51
|
Vue Z, Murphy A, Le H, Neikirk K, Garza-Lopez E, Marshall AG, Mungai M, Jenkins B, Vang L, Beasley HK, Ezedimma M, Manus S, Whiteside A, Forni MF, Harris C, Crabtree A, Albritton CF, Jamison S, Demirci M, Prasad P, Oliver A, Actkins KV, Shao J, Zaganjor E, Scudese E, Rodriguez B, Koh A, Rabago I, Moore JE, Nguyen D, Aftab M, Kirk B, Li Y, Wandira N, Ahmad T, Saleem M, Kadam A, Katti P, Koh HJ, Evans C, Koo YD, Wang E, Smith Q, Tomar D, Williams CR, Sweetwyne MT, Quintana AM, Phillips MA, Hubert D, Kirabo A, Dash C, Jadiya P, Kinder A, Ajijola OA, Miller-Fleming TW, McReynolds MR, Hinton A. MICOS Complex Loss Governs Age-Associated Murine Mitochondrial Architecture and Metabolism in the Liver, While Sam50 Dictates Diet Changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599846. [PMID: 38979162 PMCID: PMC11230271 DOI: 10.1101/2024.06.20.599846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The liver, the largest internal organ and a metabolic hub, undergoes significant declines due to aging, affecting mitochondrial function and increasing the risk of systemic liver diseases. How the mitochondrial three-dimensional (3D) structure changes in the liver across aging, and the biological mechanisms regulating such changes confers remain unclear. In this study, we employed Serial Block Face-Scanning Electron Microscopy (SBF-SEM) to achieve high-resolution 3D reconstructions of murine liver mitochondria to observe diverse phenotypes and structural alterations that occur with age, marked by a reduction in size and complexity. We also show concomitant metabolomic and lipidomic changes in aged samples. Aged human samples reflected altered disease risk. To find potential regulators of this change, we examined the Mitochondrial Contact Site and Cristae Organizing System (MICOS) complex, which plays a crucial role in maintaining mitochondrial architecture. We observe that the MICOS complex is lost during aging, but not Sam50. Sam50 is a component of the sorting and assembly machinery (SAM) complex that acts in tandem with the MICOS complex to modulate cristae morphology. In murine models subjected to a high-fat diet, there is a marked depletion of the mitochondrial protein SAM50. This reduction in Sam50 expression may heighten the susceptibility to liver disease, as our human biobank studies corroborate that Sam50 plays a genetically regulated role in the predisposition to multiple liver diseases. We further show that changes in mitochondrial calcium dysregulation and oxidative stress accompany the disruption of the MICOS complex. Together, we establish that a decrease in mitochondrial complexity and dysregulated metabolism occur with murine liver aging. While these changes are partially be regulated by age-related loss of the MICOS complex, the confluence of a murine high-fat diet can also cause loss of Sam50, which contributes to liver diseases. In summary, our study reveals potential regulators that affect age-related changes in mitochondrial structure and metabolism, which can be targeted in future therapeutic techniques.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mariaassumpta Ezedimma
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Maria Fernanda Forni
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Claude F. Albritton
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ky’Era V. Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Izabella Rabago
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Johnathan E. Moore
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Desiree Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Muhammad Aftab
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Yahang Li
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Nelson Wandira
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Taseer Ahmad
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Chantell Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, Iowa, USA1
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Dhanendra Tomar
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab,40100, Pakistan
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, 37232, USA
- Vanderbilt Institute for Global Health, Nashville, TN, 37232, USA
| | - Chandravanu Dash
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN, United States
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, NC
| | - André Kinder
- Artur Sá Earp Neto University Center – UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center, University of California, Los Angeles, CA, USA
| | - Tyne W. Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
52
|
Murphy MB, Yang Z, Subati T, Farber-Eger E, Kim K, Blackwell DJ, Fleming MR, Stark JM, Van Amburg JC, Woodall KK, Van Beusecum JP, Agrawal V, Smart CD, Pitzer A, Atkinson JB, Fogo AB, Bastarache JA, Kirabo A, Wells QS, Madhur MS, Barnett JV, Murray KT. LNK/SH2B3 loss of function increases susceptibility to murine and human atrial fibrillation. Cardiovasc Res 2024; 120:899-913. [PMID: 38377486 PMCID: PMC11218690 DOI: 10.1093/cvr/cvae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/31/2023] [Accepted: 10/07/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The lymphocyte adaptor protein (LNK) is a negative regulator of cytokine and growth factor signalling. The rs3184504 variant in SH2B3 reduces LNK function and is linked to cardiovascular, inflammatory, and haematologic disorders, including stroke. In mice, deletion of Lnk causes inflammation and oxidative stress. We hypothesized that Lnk-/- mice are susceptible to atrial fibrillation (AF) and that rs3184504 is associated with AF and AF-related stroke in humans. During inflammation, reactive lipid dicarbonyls are the major components of oxidative injury, and we further hypothesized that these mediators are critical drivers of the AF substrate in Lnk-/- mice. METHODS AND RESULTS Lnk-/- or wild-type (WT) mice were treated with vehicle or 2-hydroxybenzylamine (2-HOBA), a dicarbonyl scavenger, for 3 months. Compared with WT, Lnk-/- mice displayed increased AF duration that was prevented by 2-HOBA. In the Lnk-/- atria, action potentials were prolonged with reduced transient outward K+ current, increased late Na+ current, and reduced peak Na+ current, pro-arrhythmic effects that were inhibited by 2-HOBA. Mitochondrial dysfunction, especially for Complex I, was evident in Lnk-/- atria, while scavenging lipid dicarbonyls prevented this abnormality. Tumour necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β) were elevated in Lnk-/- plasma and atrial tissue, respectively, both of which caused electrical and bioenergetic remodelling in vitro. Inhibition of soluble TNF-α prevented electrical remodelling and AF susceptibility, while IL-1β inhibition improved mitochondrial respiration but had no effect on AF susceptibility. In a large database of genotyped patients, rs3184504 was associated with AF, as well as AF-related stroke. CONCLUSION These findings identify a novel role for LNK in the pathophysiology of AF in both experimental mice and humans. Moreover, reactive lipid dicarbonyls are critical to the inflammatory AF substrate in Lnk-/- mice and mediate the pro-arrhythmic effects of pro-inflammatory cytokines, primarily through electrical remodelling.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Action Potentials/drug effects
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/genetics
- Benzylamines/pharmacology
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/drug effects
- Inflammation Mediators/metabolism
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Phenotype
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Zhenjiang Yang
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Tuerdi Subati
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Eric Farber-Eger
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Kyungsoo Kim
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Daniel J Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Matthew R Fleming
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Joshua M Stark
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joseph C Van Amburg
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Kaylen K Woodall
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Justin P Van Beusecum
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Vineet Agrawal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Charles D Smart
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - James B Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Julie A Bastarache
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Quinn S Wells
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Meena S Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joey V Barnett
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Katherine T Murray
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| |
Collapse
|
53
|
Wilmsen SM, Dzialowski EM. Chronic changes in developmental oxygen have little effect on mitochondria and tracheal density in the endothermic moth Manduca sexta. J Exp Biol 2024; 227:jeb247882. [PMID: 38873706 DOI: 10.1242/jeb.247882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Oxygen availability during development is known to impact the development of insect respiratory and metabolic systems. Drosophila adult tracheal density exhibits developmental plasticity in response to hypoxic or hyperoxic oxygen levels during larval development. Respiratory systems of insects with higher aerobic demands, such as those that are facultative endotherms, may be even more responsive to oxygen levels above or below normoxia during development. The moth Manduca sexta is a large endothermic flying insect that serves as a good study system to start answering questions about developmental plasticity. In this study, we examined the effect of developmental oxygen levels (hypoxia: 10% oxygen, and hyperoxia: 30% oxygen) on the respiratory and metabolic phenotype of adult moths, focusing on morphological and physiological cellular and intercellular changes in phenotype. Mitochondrial respiration rate in permeabilized and isolated flight muscle was measured in adults. We found that permeabilized flight muscle fibers from the hypoxic group had increased mitochondrial oxygen consumption, but this was not replicated in isolated flight muscle mitochondria. Morphological changes in the trachea were examined using confocal imaging. We used transmission electron microscopy to quantify muscle and mitochondrial density in the flight muscle. The respiratory morphology was not significantly different between developmental oxygen groups. These results suggest that the developing M. sexta trachea and mitochondrial respiration have limited developmental plasticity when faced with rearing at 10% or 30% oxygen.
Collapse
Affiliation(s)
- Sara M Wilmsen
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, Denton, TX 76201, USA
| | - Edward M Dzialowski
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, Denton, TX 76201, USA
| |
Collapse
|
54
|
Wang L, Peng T, Deng J, Gao W, Wang H, Junhong Luo O, Huang L, Chen G. Nicotinamide riboside alleviates brain dysfunction induced by chronic cerebral hypoperfusion via protecting mitochondria. Biochem Pharmacol 2024; 225:116272. [PMID: 38723719 DOI: 10.1016/j.bcp.2024.116272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) is an enduring inadequate blood flow to the brain, resulting in vascular dementia (VaD). However, the effective treatment strategies are lacking. Supplementing with nicotinamide adenine dinucleotide (NAD+) has shown neuroprotective benefits in other neurodegenerative disorders. Nicotinamide riboside (NR), as a precursor of NAD+, is believed to hold promise in improving mitochondrial health, autophagy, and cognitive function. Meanwhile, NR has unique oral bioavailability, good tolerability, and minimal side effects, and it is the most promising for clinical translation. However, the effectiveness of NR in treating CCH-related VaD is still uncertain. The present study examined the neuroprotective effects of NR supplementation and its underlying mechanisms in a CCH rat model. The rats with CCH were given NR at a daily dosage of 400 mg/kg for 3 months. NR supplementation increased blood and brain NAD+ levels and improved brain function in CCH rats, including cognitive function and oxygenation capacity. It also reduced hippocampal neuronal loss and abnormalities and mitigated the decrease in dendritic spine density. The analysis of RNA sequencing in hippocampal tissue supports these findings. Electron microscopy and protein detection results suggest that NR may maintain mitochondrial structural integrity and exert a protective role by attenuating mitochondrial fission and impaired autophagy flux caused by CCH. In conclusion, these findings offer evidence for the neuroprotective potential of NR supplementation in ameliorating cognitive impairment induced by CCH.
Collapse
Affiliation(s)
- Lina Wang
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China
| | - Tianchan Peng
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China
| | - Jieping Deng
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Wen Gao
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Haoyun Wang
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China
| | - Li'an Huang
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guobing Chen
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine, Institute of Geriatric Immunology, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou 510632, China.
| |
Collapse
|
55
|
Beasley HK, Vue Z, McReynolds MR, Garza-Lopez E, Neikirk K, Mungai M, Marshall AG, Shao B, Benjamin JI, Wanjalla CN, Williams CR, Murray SA, Jordan VK, Shuler HD, Kirabo A, Hinton A. Running a successful STEMM summer program: A week-by-week guide. J Cell Physiol 2024; 239:e31227. [PMID: 38462753 DOI: 10.1002/jcp.31227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024]
Abstract
While some established undergraduate summer programs are effective across many institutions, these programs may only be available to some principal investigators or may not fully address the diverse needs of incoming undergraduates. This article outlines a 10-week science, technology, engineering, mathematics, and medicine (STEMM) education program designed to prepare undergraduate students for graduate school through a unique model incorporating mentoring dyads and triads, cultural exchanges, and diverse activities while emphasizing critical thinking, research skills, and cultural sensitivity. Specifically, we offer a straightforward and adaptable guide that we have used for mentoring undergraduate students in a laboratory focused on mitochondria and microscopy, but which may be customized for other disciplines. Key components include self-guided projects, journal clubs, various weekly activities such as mindfulness training and laboratory techniques, and a focus on individual and cultural expression. Beyond this unique format, this 10-week program also seeks to offer an intensive research program that emulates graduate-level experiences, offering an immersive environment for personal and professional development, which has led to numerous achievements for past students, including publications and award-winning posters.
Collapse
Affiliation(s)
- Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- The Huck Institute of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Edgar Garza-Lopez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jazmine I Benjamin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Celestine N Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clintoria R Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio, USA
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Haysetta D Shuler
- Department of Biological Sciences, Winston-Salem State University, Winston-Salem, North Carolina, USA
- Shuler Consulting, Winston-Salem, North Carolina, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
56
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Su YR, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3 + T-cell: CD14 +monocyte complexes are dynamic and increased with HIV and glucose intolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.24.538020. [PMID: 37162990 PMCID: PMC10168203 DOI: 10.1101/2023.04.24.538020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
An increased risk of cardiometabolic disease accompanies persistent systemic inflammation. Yet, the innate and adaptive immune system features in persons who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled HIV as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+T-cell: CD14+monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+T-cell:CD14+monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis and may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cellmonocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Leslie M. Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L. Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin M. Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Gianella
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J. Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Spyros A. Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
57
|
Vue Z, Prasad P, Le H, Neikirk K, Harris C, Garza-Lopez E, Wang E, Murphy A, Jenkins B, Vang L, Scudese E, Shao B, Kadam A, Shao J, Marshall AG, Crabtree A, Kirk B, Koh A, Wilson G, Oliver A, Rodman T, Kabugi K, Koh HJ, Smith Q, Zaganjor E, Wanjalla CN, Dash C, Evans C, Phillips MA, Hubert D, Ajijola O, Whiteside A, Do Koo Y, Kinder A, Demirci M, Albritton CF, Wandira N, Jamison S, Ahmed T, Saleem M, Tomar D, Williams CR, Sweetwyne MT, Murray SA, Cooper A, Kirabo A, Jadiya P, Quintana A, Katti P, Fu Dai D, McReynolds MR, Hinton A. The MICOS Complex Regulates Mitochondrial Structure and Oxidative Stress During Age-Dependent Structural Deficits in the Kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598108. [PMID: 38915644 PMCID: PMC11195114 DOI: 10.1101/2024.06.09.598108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The kidney filters nutrient waste and bodily fluids from the bloodstream, in addition to secondary functions of metabolism and hormone secretion, requiring an astonishing amount of energy to maintain its functions. In kidney cells, mitochondria produce adenosine triphosphate (ATP) and help maintain kidney function. Due to aging, the efficiency of kidney functions begins to decrease. Dysfunction in mitochondria and cristae, the inner folds of mitochondria, is a hallmark of aging. Therefore, age-related kidney function decline could be due to changes in mitochondrial ultrastructure, increased reactive oxygen species (ROS), and subsequent alterations in metabolism and lipid composition. We sought to understand if there is altered mitochondrial ultrastructure, as marked by 3D morphological changes, across time in tubular kidney cells. Serial block facing-scanning electron microscope (SBF-SEM) and manual segmentation using the Amira software were used to visualize murine kidney samples during the aging process at 3 months (young) and 2 years (old). We found that 2-year mitochondria are more fragmented, compared to the 3-month, with many uniquely shaped mitochondria observed across aging, concomitant with shifts in ROS, metabolomics, and lipid homeostasis. Furthermore, we show that the mitochondrial contact site and cristae organizing system (MICOS) complex is impaired in the kidney due to aging. Disruption of the MICOS complex shows altered mitochondrial calcium uptake and calcium retention capacity, as well as generation of oxidative stress. We found significant, detrimental structural changes to aged kidney tubule mitochondria suggesting a potential mechanism underlying why kidney diseases occur more readily with age. We hypothesize that disruption in the MICOS complex further exacerbates mitochondrial dysfunction, creating a vicious cycle of mitochondrial degradation and oxidative stress, thus impacting kidney health.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Genesis Wilson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | | | - Chandravanu Dash
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, TN, United States
| | - Chantell Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Olujimi Ajijola
- UCLA Cardiac Arrhythmia Center, University of California, Los Angeles, CA, USA
| | - Aaron Whiteside
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, Iowa, USA
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Claude F. Albritton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Nelson Wandira
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Taseer Ahmed
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute for Global Health, Vanderbilt University, Nashville, TN, 37232, USA
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Anita Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Dao Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
58
|
Scudese E, Vue Z, Katti P, Marshall AG, Demirci M, Vang L, López EG, Neikirk K, Shao B, Le H, Stephens D, Hall DD, Rostami R, Rodman T, Kabugi K, Harris C, Shao J, Mungai M, AshShareef ST, Hicsasmaz I, Manus S, Wanjalla C, Whiteside A, Dasari R, Williams C, Damo SM, Gaddy JA, Glancy B, Dantas EHM, Kinder A, Kadam A, Tomar D, Scartoni F, Baffi M, McReynolds MR, Phillips MA, Cooper A, Murray SA, Quintana AM, Exil V, Kirabo A, Mobley BC, Hinton A. 3D Mitochondrial Structure in Aging Human Skeletal Muscle: Insights into MFN-2 Mediated Changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.13.566502. [PMID: 38168206 PMCID: PMC10760012 DOI: 10.1101/2023.11.13.566502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Age-related atrophy of skeletal muscle, is characterized by loss of mass, strength, endurance, and oxidative capacity during aging. Notably, bioenergetics and protein turnover studies have shown that mitochondria mediate this decline in function. Although exercise has been the only therapy to mitigate sarcopenia, the mechanisms that govern how exercise serves to promote healthy muscle aging are unclear. Mitochondrial aging is associated with decreased mitochondrial capacity, so we sought to investigate how aging affects mitochondrial structure and potential age-related regulators. Specifically, the three-dimensional (3D) mitochondrial structure associated with morphological changes in skeletal muscle during aging requires further elucidation. We hypothesized that aging causes structural remodeling of mitochondrial 3D architecture representative of dysfunction, and this effect is mitigated by exercise. We used serial block-face scanning electron microscopy to image human skeletal tissue samples, followed by manual contour tracing using Amira software for 3D reconstruction and subsequent analysis of mitochondria. We then applied a rigorous in vitro and in vivo exercise regimen during aging. Across 5 human cohorts, we correlate differences in magnetic resonance imaging, mitochondria 3D structure, exercise parameters, and plasma immune markers between young (under 50 years) and old (over 50 years) individuals. We found that mitochondria we less spherical and more complex, indicating age-related declines in contact site capacity. Additionally, aged samples showed a larger volume phenotype in both female and male humans, indicating potential mitochondrial swelling. Concomitantly, muscle area, exercise capacity, and mitochondrial dynamic proteins showed age-related losses. Exercise stimulation restored mitofusin 2 (MFN2), one such of these mitochondrial dynamic proteins, which we show is required for the integrity of mitochondrial structure. Furthermore, we show that this pathway is evolutionarily conserved as Marf, the MFN2 ortholog in Drosophila, knockdown alters mitochondrial morphology and leads to the downregulation of genes regulating mitochondrial processes. Our results define age-related structural changes in mitochondria and further suggest that exercise may mitigate age-related structural decline through modulation of mitofusin 2.
Collapse
Affiliation(s)
- Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Prassana Katti
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mert Demirci
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza López
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Duane D. Hall
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, Iowa City, IA 52242, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Salma T. AshShareef
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Innes Hicsasmaz
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sasha Manus
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celestine Wanjalla
- Division of Infection Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, 45435, USA
| | - Revathi Dasari
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Clintoria Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH, 45435, USA
| | - Steven M. Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
| | - Jennifer A. Gaddy
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- NIAMS, NIH, Bethesda, MD, 20892, USA
| | - Estélio Henrique Martin Dantas
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Doctor’s Degree Program in Nursing and Biosciences - PpgEnfBio, Federal University of the State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
- Laboratory of Human Motricity Biosciences - LABIMH, Federal University of the State of Rio de Janeiro - UNIRIO, RJ, Brazil
- Brazilian Paralympic Academy – APB
- Doctor’s Degree Program in Health and Environment - PSA, Tiradentes University - UNIT, Aracaju, SE, Brazil
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Fabiana Scartoni
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Matheus Baffi
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA, 16801, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Vernat Exil
- Department of Pediatrics, Div. of Cardiology, St. Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Annet Kirabo
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bret C. Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
59
|
Rockfield SM, Turnis ME, Rodriguez-Enriquez R, Bathina M, Ng SK, Kurtz N, Becerra Mora N, Pelletier S, Robinson CG, Vogel P, Opferman JT. Genetic ablation of Immt induces a lethal disruption of the MICOS complex. Life Sci Alliance 2024; 7:e202302329. [PMID: 38467404 PMCID: PMC10927357 DOI: 10.26508/lsa.202302329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
The mitochondrial contact site and cristae organizing system (MICOS) is important for crista junction formation and for maintaining inner mitochondrial membrane architecture. A key component of the MICOS complex is MIC60, which has been well studied in yeast and cell culture models. However, only one recent study has demonstrated the embryonic lethality of losing Immt (the gene encoding MIC60) expression. Tamoxifen-inducible ROSA-CreERT2-mediated deletion of Immt in adult mice disrupted the MICOS complex, increased mitochondria size, altered cristae morphology, and was lethal within 12 d. Pathologically, these mice displayed defective intestinal muscle function (paralytic ileus) culminating in dehydration. We also identified bone marrow (BM) hypocellularity in Immt-deleted mice, although BM transplants from wild-type mice did not improve survival. Altogether, this inducible mouse model demonstrates the importance of MIC60 in vivo, in both hematopoietic and non-hematopoietic tissues, and provides a valuable resource for future mechanistic investigations into the MICOS complex.
Collapse
Affiliation(s)
- Stephanie M Rockfield
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Meghan E Turnis
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Madhavi Bathina
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Seng Kah Ng
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nathan Kurtz
- Electron Microscopy, Department of Cellular Imaging Shared Resources, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nathalie Becerra Mora
- Electron Microscopy, Department of Cellular Imaging Shared Resources, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephane Pelletier
- Transgenic Core Facility, Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Camenzind G Robinson
- Electron Microscopy, Department of Cellular Imaging Shared Resources, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Vogel
- Comparative Pathology Core, Pathology Department, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
60
|
Balhara M, Neikirk K, Marshall A, Hinton A, Kirabo A. Endoplasmic Reticulum Stress in Hypertension and Salt Sensitivity of Blood Pressure. Curr Hypertens Rep 2024; 26:273-290. [PMID: 38602583 PMCID: PMC11166838 DOI: 10.1007/s11906-024-01300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE OF REVIEW Hypertension is a principal risk factor for cardiovascular morbidity and mortality, with its severity exacerbated by high sodium intake, particularly in individuals with salt-sensitive blood pressure. However, the mechanisms underlying hypertension and salt sensitivity are only partly understood. Herein, we review potential interactions in hypertension pathophysiology involving the immune system, endoplasmic reticulum (ER) stress, the unfolded protein response (UPR), and proteostasis pathways; identify knowledge gaps; and discuss future directions. RECENT FINDINGS Recent advancements by our research group and others reveal interactions within and between adaptive and innate immune responses in hypertension pathophysiology. The salt-immune-hypertension axis is further supported by the discovery of the role of dendritic cells in hypertension, marked by isolevuglandin (IsoLG) formation. Alongside these broadened understandings of immune-mediated salt sensitivity, the contributions of T cells to hypertension have been recently challenged by groups whose findings did not support increased resistance of Rag-1-deficient mice to Ang II infusion. Hypertension has also been linked to ER stress and the UPR. Notably, a holistic approach is needed because the UPR engages in crosstalk with autophagy, the ubiquitin proteasome, and other proteostasis pathways, that may all involve hypertension. There is a critical need for studies to establish cause and effect relationships between ER stress and the UPR in hypertension pathophysiology in humans and to determine whether the immune system and ER stress function mainly to exacerbate or initiate hypertension and target organ injury. This review of recent studies proposes new avenues for future research for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maria Balhara
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37212-8802, USA.
- Vanderbilt Center for Immunobiology, Nashville, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, USA.
- Vanderbilt Institute for Global Health, Nashville, USA.
| |
Collapse
|
61
|
Yuan C, Duan Y, Li X, Zhang Y, Cao L, Feng T, Ge J, Wang Q, Zheng H. Transcriptional and ultrastructural changes of macrophages after african swine fever virus infection. Vet Microbiol 2024; 293:110074. [PMID: 38603982 DOI: 10.1016/j.vetmic.2024.110074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
African swine fever (ASF) is a highly impactful infectious disease in the swine industry, leading to substantial economic losses globally. The causative agent, African swine fever virus (ASFV), possesses intricate pathogenesis, warranting further exploration. In this study, we investigated the impact of ASFV infection on host gene transcription and organelle changes through macrophage transcriptome sequencing and ultrastructural transmission electron microscopy observation. According to the results of the transcriptome sequencing, ASFV infection led to significant alterations in the gene expression pattern of porcine bone marrow derived macrophages (BMDMs), with 2404 genes showing upregulation and 1579 genes downregulation. Cytokines, and chemokines were significant changes in the expression of BMDMs; there was significant activation of pattern recognition receptors such as Toll-like receptors and Nod-like receptors. According to the observation of the ultrastructure, mitochondrial damage and mitochondrial autophagy were widely present in ASFV-infected cells. The reduced number of macrophage pseudopodia suggested that virus-induced structural changes may compromise pathogen recognition, phagocytosis, and signal communication in macrophages. Additionally, the decreased size and inhibited acidification of secondary lysosomes in macrophages implied suppressed phagocytosis. Overall, ASFV infection resulted in significant changes in the expression of cytokines and chemokines, accompanied by the activation of NLR and TLR signaling pathways. We reported for the first time that ASFV infection led to a reduction in pseudopodia numbers and a decrease in the size and acidification of secondary lysosomes.
Collapse
Affiliation(s)
- Cong Yuan
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yueyue Duan
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Xiangtong Li
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Yu Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Liyan Cao
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Tao Feng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Junwei Ge
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Qi Wang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China; Chengdu National Agricultural Science and Technology Center, Chengdu, China.
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| |
Collapse
|
62
|
Cheng CJ, Nizar JM, Dai DF, Huang CL. Transport activity regulates mitochondrial bioenergetics and biogenesis in renal tubules. FASEB J 2024; 38:e23703. [PMID: 38805156 PMCID: PMC11147170 DOI: 10.1096/fj.202400358rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Renal tubules are featured with copious mitochondria and robust transport activity. Mutations in mitochondrial genes cause congenital renal tubulopathies, and changes in transport activity affect mitochondrial morphology, suggesting mitochondrial function and transport activity are tightly coupled. Current methods of using bulk kidney tissues or cultured cells to study mitochondrial bioenergetics are limited. Here, we optimized an extracellular flux analysis (EFA) to study mitochondrial respiration and energy metabolism using microdissected mouse renal tubule segments. EFA detects mitochondrial respiration and glycolysis by measuring oxygen consumption and extracellular acidification rates, respectively. We show that both measurements positively correlate with sample sizes of a few centimeter-length renal tubules. The thick ascending limbs (TALs) and distal convoluted tubules (DCTs) critically utilize glucose/pyruvate as energy substrates, whereas proximal tubules (PTs) are significantly much less so. Acute inhibition of TALs' transport activity by ouabain treatment reduces basal and ATP-linked mitochondrial respiration. Chronic inhibition of transport activity by 2-week furosemide treatment or deletion of with-no-lysine kinase 4 (Wnk4) decreases maximal mitochondrial capacity. In addition, chronic inhibition downregulates mitochondrial DNA mass and mitochondrial length/density in TALs and DCTs. Conversely, gain-of-function Wnk4 mutation increases maximal mitochondrial capacity and mitochondrial length/density without increasing mitochondrial DNA mass. In conclusion, EFA is a sensitive and reliable method to investigate mitochondrial functions in isolated renal tubules. Transport activity tightly regulates mitochondrial bioenergetics and biogenesis to meet the energy demand in renal tubules. The system allows future investigation into whether and how mitochondria contribute to tubular remodeling adapted to changes in transport activity.
Collapse
Affiliation(s)
- Chih-Jen Cheng
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jonathan M Nizar
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
| | - Dao-Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, U.S.A
| | - Chou-Long Huang
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
| |
Collapse
|
63
|
Hinton AO, N'jai AU, Vue Z, Wanjalla C. Connection Between HIV and Mitochondria in Cardiovascular Disease and Implications for Treatments. Circ Res 2024; 134:1581-1606. [PMID: 38781302 PMCID: PMC11122810 DOI: 10.1161/circresaha.124.324296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
HIV infection and antiretroviral therapy alter mitochondrial function, which can progressively lead to mitochondrial damage and accelerated aging. The interaction between persistent HIV reservoirs and mitochondria may provide insight into the relatively high rates of cardiovascular disease and mortality in persons living with HIV. In this review, we explore the intricate relationship between HIV and mitochondrial function, highlighting the potential for novel therapeutic strategies in the context of cardiovascular diseases. We reflect on mitochondrial dynamics, mitochondrial DNA, and mitochondrial antiviral signaling protein in the context of HIV. Furthermore, we summarize how toxicities related to early antiretroviral therapy and current highly active antiretroviral therapy can contribute to mitochondrial dysregulation, chronic inflammation, and poor clinical outcomes. There is a need to understand the mechanisms and develop new targeted therapies. We further consider current and potential future therapies for HIV and their interplay with mitochondria. We reflect on the next-generation antiretroviral therapies and HIV cure due to the direct and indirect effects of HIV persistence, associated comorbidities, coinfections, and the advancement of interdisciplinary research fields. This includes exploring novel and creative approaches to target mitochondria for therapeutic intervention.
Collapse
Affiliation(s)
- Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Alhaji U N'jai
- Biological Sciences, Fourah Bay College and College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone and Koinadugu College, Kabala (A.U.N.)
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Celestine Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.W.)
| |
Collapse
|
64
|
Xu M, Li LP, He X, Lu XZ, Bi XY, Li Q, Xue XR. Metformin induction of heat shock factor 1 activation and the mitochondrial unfolded protein response alleviate cardiac remodeling in spontaneously hypertensive rats. FASEB J 2024; 38:e23654. [PMID: 38717442 DOI: 10.1096/fj.202400070r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/30/2024] [Accepted: 04/23/2024] [Indexed: 06/07/2024]
Abstract
Heart failure and cardiac remodeling are both characterized by mitochondrial dysfunction. Healthy mitochondria are required for adequate contractile activity and appropriate regulation of cell survival. In the mammalian heart, enhancement of the mitochondrial unfolded protein response (UPRmt) is cardioprotective under pressure overload conditions. We explored the UPRmt and the underlying regulatory mechanism in terms of hypertension-induced cardiac remodeling and the cardioprotective effect of metformin. Male spontaneously hypertensive rats and angiotensin II-treated neonatal rat cardiomyocytes were used to induce cardiac hypertrophy. The results showed that hypertension induced the formation of aberrant mitochondria, characterized by a reduced mtDNA/nDNA ratio and swelling, as well as lower levels of mitochondrial complexes I to V and inhibition of the expression of one protein subunit of each of complexes I to IV. Such changes eventually enlarged cardiomyocytes and increased cardiac fibrosis. Metformin treatment increased the mtDNA/nDNA ratio and regulated the UPRmt, as indicated by increased expression of activating transcription factor 5, Lon protease 1, and heat shock protein 60, and decreased expression of C/EBP homologous protein. Thus, metformin improved mitochondrial ultrastructure and function in spontaneously hypertensive rats. In vitro analyses revealed that metformin reduced the high levels of angiotensin II-induced mitochondrial reactive oxygen species in such animals and stimulated nuclear translocation of heat shock factor 1 (HSF1). Moreover, HSF1 small-interfering RNA reduced the metformin-mediated improvements in mitochondrial morphology and the UPRmt by suppressing hypertrophic signals and cardiomyocyte apoptosis. These results suggest that HSF1/UPRmt signaling contributes to the beneficial effects of metformin. Metformin-mediated targeting of mitochondrial protein homeostasis and modulation of HSF1 levels have potential therapeutic implications in terms of cardiac remodeling.
Collapse
Affiliation(s)
- Man Xu
- Department of Pharmacy, Xi'an People's Hospital (Xi'an Fourth Hospital), Northwest University Affiliated People's Hospital, Xi'an, Shaanxi, China
| | - Li-Peng Li
- Department of Pharmacy, Xi'an People's Hospital (Xi'an Fourth Hospital), Northwest University Affiliated People's Hospital, Xi'an, Shaanxi, China
| | - Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xing-Zhu Lu
- Department of Pharmacy, Second Affiliated Hospital of Xi'an Jiaotong University Medical School, Xi'an, Shaanxi, China
| | - Xue-Yuan Bi
- Department of Pharmacy, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qi Li
- Department of Science and Education, Xi'an People's Hospital (Xi'an Fourth Hospital), Northwest University Affiliated People's Hospital, Xi'an, China
| | - Xiao-Rong Xue
- Department of Pharmacy, Xi'an People's Hospital (Xi'an Fourth Hospital), Northwest University Affiliated People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
65
|
Cheng D, Lei ZG, Chu K, Lam OJH, Chiang CY, Zhang ZJ. N, N-Dimethyltryptamine, a natural hallucinogen, ameliorates Alzheimer's disease by restoring neuronal Sigma-1 receptor-mediated endoplasmic reticulum-mitochondria crosstalk. Alzheimers Res Ther 2024; 16:95. [PMID: 38693554 PMCID: PMC11061967 DOI: 10.1186/s13195-024-01462-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Aberrant neuronal Sigma-1 receptor (Sig-1r)-mediated endoplasmic reticulum (ER)- mitochondria signaling plays a key role in the neuronal cytopathology of Alzheimer's disease (AD). The natural psychedelic N, N-dimethyltryptamine (DMT) is a Sig-1r agonist that may have the anti-AD potential through protecting neuronal ER-mitochondrial interplay. METHODS 3×TG-AD transgenic mice were administered with chronic DMT (2 mg/kg) for 3 weeks and then performed water maze test. The Aβ accumulation in the mice brain were determined. The Sig-1r level upon DMT treatment was tested. The effect of DMT on the ER-mitochondrial contacts site and multiple mitochondria-associated membrane (MAM)-associated proteins were examined. The effect of DMT on calcium transport between ER and mitochondria and the mitochondrial function were also evaluated. RESULTS chronic DMT (2 mg/kg) markedly alleviated cognitive impairment of 3×TG-AD mice. In parallel, it largely diminished Aβ accumulation in the hippocampus and prefrontal cortex. DMT restored the decreased Sig-1r levels of 3×TG-AD transgenic mice. The hallucinogen reinstated the expression of multiple MAM-associated proteins in the brain of 3×TG-AD mice. DMT also prevented physical contact and calcium dynamic between the two organelles in in vitro and in vivo pathological circumstances. DMT modulated oxidative phosphorylation (OXPHOS) and ATP synthase in the in vitro model of AD. CONCLUSION The anti-AD effects of DMT are associated with its protection of neuronal ER-mitochondria crosstalk via the activation of Sig-1r. DMT has the potential to serve as a novel preventive and therapeutic agent against AD.
Collapse
Affiliation(s)
- Dan Cheng
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhuo-Gui Lei
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Kin Chu
- Department of Psychology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Oi Jin Honey Lam
- School of Biomedical Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Chun Yuan Chiang
- Digital Centre of State Key Laboratory of Quality Research in Chinese Medicine, Macau, China
| | - Zhang-Jin Zhang
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China.
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
66
|
You W, Knoops K, Berendschot TTJM, Benedikter BJ, Webers CAB, Reutelingsperger CPM, Gorgels TGMF. PGC-1a mediated mitochondrial biogenesis promotes recovery and survival of neuronal cells from cellular degeneration. Cell Death Discov 2024; 10:180. [PMID: 38632223 PMCID: PMC11024166 DOI: 10.1038/s41420-024-01953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Neurodegenerative disorders are characterized by the progressive loss of structure and function of neurons, often including the death of the neuron. Previously, we reported that, by removing the cell death stimulus, dying/injured neurons could survive and recover from the process of regulated cell death, even if the cells already displayed various signs of cellular damage. Now we investigated the role of mitochondrial dynamics (fission/fusion, biogenesis, mitophagy) in both degeneration and in recovery of neuronal cells. In neuronal PC12 cells, exposure to ethanol (EtOH) induced massive neurite loss along with widespread mitochondrial fragmentation, mitochondrial membrane potential loss, reduced ATP production, and decreased total mitochondrial volume. By removing EtOH timely all these mitochondrial parameters recovered to normal levels. Meanwhile, cells regrew neurites and survived. Study of the mitochondrial dynamics showed that autophagy was activated only during the cellular degeneration phase (EtOH treatment) but not in the recovery phase (EtOH removed), and it was not dependent on the Parkin/PINK1 mediated mitophagy pathway. Protein expression of key regulators of mitochondrial fission, phospho-Drp1Ser616 and S-OPA1, increased during EtOH treatment and recovered to normal levels after removing EtOH. In addition, the critical role of PGC-1α mediated mitochondrial biogenesis in cellular recovery was revealed: inhibition of PGC-1α using SR-18292 after EtOH removal significantly impeded recovery of mitochondrial damage, regeneration of neurites, and cell survival in a concentration-dependent manner. Taken together, our study showed reversibility of mitochondrial morphological and functional damage in stressed neuronal cells and revealed that PGC-1α mediated mitochondrial biogenesis played a critical role in the cellular recovery. This molecular mechanism could be a target for neuroprotection and neurorescue in neurodegenerative diseases.
Collapse
Affiliation(s)
- Wenting You
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands
- Department of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Kèvin Knoops
- The Microscopy CORE lab, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Tos T J M Berendschot
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Birke J Benedikter
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands.
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
67
|
Li M, Zhang Y, Yu G, Gu L, Zhu H, Feng S, Xiong X, Jian Z. Mitochondria-associated endoplasmic reticulum membranes tethering protein VAPB-PTPIP51 protects against ischemic stroke through inhibiting the activation of autophagy. CNS Neurosci Ther 2024; 30:e14707. [PMID: 38584329 PMCID: PMC10999572 DOI: 10.1111/cns.14707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/09/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024] Open
Abstract
AIMS Mitochondria-associated endoplasmic reticulum membranes (MAMs) serve as a crucial bridge connecting the endoplasmic reticulum (ER) and mitochondria within cells. Vesicle-associated membrane protein-associated protein B (VAPB) and protein tyrosine phosphatase interacting protein 51 (PTPIP51) are responsible for the formation and stability of MAMs, which have been implicated in the pathogenesis of various diseases. However, the role of MAMs in ischemic stroke (IS) remains unclear. We aimed to investigate the role of MAMs tethering protein VAPB-PTPIP51 in experimental cerebral ischemia. METHODS We simulated cerebral ischemia-reperfusion injury (CIRI) by using a mouse middle cerebral artery occlusion (MCAO) model. RESULTS We observed a decrease in VAPB-PTPIP51 expression in the brain tissue. Our findings suggested compromised MAMs after MCAO, as a decreased mitochondria-ER contact (MERC) coverage and an increased distance were observed through the transmission electron microscope (TEM). Upon VAPB or PTPIP51 knockdown, the damage to MAMs was exacerbated, accompanied by excessive autophagy activation and increased reactive oxygen species (ROS) production, resulting in an enlarged infarct area and exacerbated neurological deficits. Notably, we observed that this damage was concomitant with the inhibition of the PI3K/AKT/mTOR pathway and was successfully mitigated by the treatment with the PI3K activator. CONCLUSIONS Our findings suggest that the downregulation of VAPB-PTPIP51 expression after IS mediates structural damage to MAMs. This may exacerbate CIRI by inhibiting the PI3K pathway and activating autophagy, thus providing new therapeutic targets for IS.
Collapse
Affiliation(s)
- Mingyang Li
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yonggang Zhang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Guixiang Yu
- Department of OphthalmologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Lijuan Gu
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hua Zhu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Shi Feng
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhihong Jian
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
68
|
Neikirk K, Stephens DC, Beasley HK, Marshall AG, Gaddy JA, Damo SM, Hinton AO. Considerations for developing mitochondrial transplantation techniques for individualized medicine. Biotechniques 2024; 76:125-134. [PMID: 38420889 DOI: 10.2144/btn-2023-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Tweetable abstract Mitochondrial transplantation has been used to treat various diseases associated with mitochondrial dysfunction. Here, we highlight the considerations in quality control mechanisms that should be considered in the context of mitochondrial transplantation.
Collapse
Affiliation(s)
- Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Dominique C Stephens
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Heather K Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven M Damo
- Department of Life & Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Antentor O Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
69
|
Jenkins BC, Neikirk K, Katti P, Claypool SM, Kirabo A, McReynolds MR, Hinton A. Mitochondria in disease: changes in shapes and dynamics. Trends Biochem Sci 2024; 49:346-360. [PMID: 38402097 PMCID: PMC10997448 DOI: 10.1016/j.tibs.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/14/2024] [Accepted: 01/26/2024] [Indexed: 02/26/2024]
Abstract
Mitochondrial structure often determines the function of these highly dynamic, multifunctional, eukaryotic organelles, which are essential for maintaining cellular health. The dynamic nature of mitochondria is apparent in descriptions of different mitochondrial shapes [e.g., donuts, megamitochondria (MGs), and nanotunnels] and crista dynamics. This review explores the significance of dynamic alterations in mitochondrial morphology and regulators of mitochondrial and cristae shape. We focus on studies across tissue types and also describe new microscopy techniques for detecting mitochondrial morphologies both in vivo and in vitro that can improve understanding of mitochondrial structure. We highlight the potential therapeutic benefits of regulating mitochondrial morphology and discuss prospective avenues to restore mitochondrial bioenergetics to manage diseases related to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Brenita C Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Steven M Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801, USA.
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
70
|
Hinton A, Katti P, Mungai M, Hall DD, Koval O, Shao J, Vue Z, Lopez EG, Rostami R, Neikirk K, Ponce J, Streeter J, Schickling B, Bacevac S, Grueter C, Marshall A, Beasley HK, Do Koo Y, Bodine SC, Nava NGR, Quintana AM, Song LS, Grumbach I, Pereira RO, Glancy B, Abel ED. ATF4-dependent increase in mitochondrial-endoplasmic reticulum tethering following OPA1 deletion in skeletal muscle. J Cell Physiol 2024; 239:e31204. [PMID: 38419397 PMCID: PMC11144302 DOI: 10.1002/jcp.31204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
Mitochondria and endoplasmic reticulum (ER) contact sites (MERCs) are protein- and lipid-enriched hubs that mediate interorganellar communication by contributing to the dynamic transfer of Ca2+, lipid, and other metabolites between these organelles. Defective MERCs are associated with cellular oxidative stress, neurodegenerative disease, and cardiac and skeletal muscle pathology via mechanisms that are poorly understood. We previously demonstrated that skeletal muscle-specific knockdown (KD) of the mitochondrial fusion mediator optic atrophy 1 (OPA1) induced ER stress and correlated with an induction of Mitofusin-2, a known MERC protein. In the present study, we tested the hypothesis that Opa1 downregulation in skeletal muscle cells alters MERC formation by evaluating multiple myocyte systems, including from mice and Drosophila, and in primary myotubes. Our results revealed that OPA1 deficiency induced tighter and more frequent MERCs in concert with a greater abundance of MERC proteins involved in calcium exchange. Additionally, loss of OPA1 increased the expression of activating transcription factor 4 (ATF4), an integrated stress response (ISR) pathway effector. Reducing Atf4 expression prevented the OPA1-loss-induced tightening of MERC structures. OPA1 reduction was associated with decreased mitochondrial and sarcoplasmic reticulum, a specialized form of ER, calcium, which was reversed following ATF4 repression. These data suggest that mitochondrial stress, induced by OPA1 deficiency, regulates skeletal muscle MERC formation in an ATF4-dependent manner.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Prasanna Katti
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA, 20892
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Duane D. Hall
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
| | - Olha Koval
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Jianqiang Shao
- Central Microscopy Research Facility, Iowa City, IA USA 52242
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Edgar Garza Lopez
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
| | - Rahmati Rostami
- Department of Genetic Medicine, Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA, 10065
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Jessica Ponce
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Jennifer Streeter
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Brandon Schickling
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Department of Medicine, Duke University, Durham, NC, USA 27708
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Chad Grueter
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Andrea Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Sue C. Bodine
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA 73104
| | - Nayeli G. Reyes Nava
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA 79968
| | - Anita M. Quintana
- Department of Biological Sciences and Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA 79968
| | - Long-Sheng Song
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Isabella Grumbach
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Renata O. Pereira
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA, 20892
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA 20892
| | - E. Dale Abel
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, Iowa City, IA, USA 52242
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, USA 52242
- Department of Medicine, UCLA School of Medicine, Los Angeles, CA, USA 90095
| |
Collapse
|
71
|
Liang W, Najor RH, Gustafsson ÅB. Protocol to separate small and large extracellular vesicles from mouse and human cardiac tissues. STAR Protoc 2024; 5:102914. [PMID: 38386549 PMCID: PMC10897917 DOI: 10.1016/j.xpro.2024.102914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/05/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Extracellular vesicles (EVs) are secreted by cells under various conditions and can contribute to the disease progression in tissues. Here, we present a protocol to separate small and large EVs from mouse hearts and cardiac tissues collected from patients. We describe steps for utilizing enzymatic digestion for release of EVs from interstitial space followed by differential centrifugation and immunoaffinity purification. The isolated EVs can be used for various experiments to gain insight into their in vivo functions. For complete details on the use and execution of this protocol, please refer to Liang et al. (2023).1.
Collapse
Affiliation(s)
- Wenjing Liang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
72
|
Shati AA, Eid RA, El-kott AF, Alqahtani YA, Shatoor AS, Ahmed Zaki MS. Curcumin attenuates doxorubicin-induced cardiotoxicity via suppressing oxidative Stress, preventing inflammation and apoptosis: Ultrastructural and computational approaches. Heliyon 2024; 10:e27164. [PMID: 38468941 PMCID: PMC10926088 DOI: 10.1016/j.heliyon.2024.e27164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Currently, doxorubicin (DOX) is one of the medications commonly used in chemotherapy to treat different types of tumors.Nonetheless, despite being effective in multiple tumors, yet its use is limited owing to its cytotoxic effects, the therapeutic use of DOX has been limited. This work aimed to explore whether curcumin (CMN) can prevents DOX-induced cardiotoxicity in rats. Four groups of rats were created, with the first functioning as a control, while the second group received CMN. DOX alone was administered to the third group, whereas CMN and DOX were administered to the fourth group. Lipid peroxidation assessed as Malondialdehyde (MDA), aspartate aminotransferase (AST), alanine aminotransferase (ALT), oxidative stress markers as catalase (CAT), superoxide dismutase (SOD), and inflammatory markers as tumor necrosis factor-alpha (TNF-α) in heart homogenates, each one was assessed. Heart specimens was investigated histologically and ultrastructurally. Increased, AST, and ALT serum levels, increased MDA levels, decreased SOD and CAT levels, and increased TNF-α concentrations in heart homogenates were all signs of DOX-induced myocardial injury. Histological and ultrastructural examinations revealed vacuoles and larger, swollen mitochondria in the cytoplasm. Furthermore, DOX caused significant changes in the myocardium, most notably nuclei disintegration, myofibrillar loss, and myocyte vacuolization. Using CMN with DOX reduced the harmful consequences of DOX on the myocardium by returning the increased AST and ALT levels to their original levels as compared to the control and reducing them. In cardiac tissue, CMN significantly increased the concentrations of SOD and CAT and significantly decreased the concentrations of MDA and TNF-α. Biochemical and histological studies have demonstrated that CMN has a heart-protective effect that might be related to its antioxidant and anti-inflammatory capabilities.
Collapse
Affiliation(s)
- Ayed A. Shati
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Refaat A. Eid
- Department of Pathology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Attalla F. El-kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
- Department of Zoology, College of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Youssef A. Alqahtani
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdullah S. Shatoor
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
73
|
Kawatani K, Holm ML, Starling SC, Martens YA, Zhao J, Lu W, Ren Y, Li Z, Jiang P, Jiang Y, Baker SK, Wang N, Roy B, Parsons TM, Perkerson RB, Bao H, Han X, Bu G, Kanekiyo T. ABCA7 deficiency causes neuronal dysregulation by altering mitochondrial lipid metabolism. Mol Psychiatry 2024; 29:809-819. [PMID: 38135757 PMCID: PMC11153016 DOI: 10.1038/s41380-023-02372-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
ABCA7 loss-of-function variants are associated with increased risk of Alzheimer's disease (AD). Using ABCA7 knockout human iPSC models generated with CRISPR/Cas9, we investigated the impacts of ABCA7 deficiency on neuronal metabolism and function. Lipidomics revealed that mitochondria-related phospholipids, such as phosphatidylglycerol and cardiolipin were reduced in the ABCA7-deficient iPSC-derived cortical organoids. Consistently, ABCA7 deficiency-induced alterations of mitochondrial morphology accompanied by reduced ATP synthase activity and exacerbated oxidative damage in the organoids. Furthermore, ABCA7-deficient iPSC-derived neurons showed compromised mitochondrial respiration and excess ROS generation, as well as enlarged mitochondrial morphology compared to the isogenic controls. ABCA7 deficiency also decreased spontaneous synaptic firing and network formation in iPSC-derived neurons, in which the effects were rescued by supplementation with phosphatidylglycerol or NAD+ precursor, nicotinamide mononucleotide. Importantly, effects of ABCA7 deficiency on mitochondria morphology and synapses were recapitulated in synaptosomes isolated from the brain of neuron-specific Abca7 knockout mice. Together, our results provide evidence that ABCA7 loss-of-function contributes to AD risk by modulating mitochondria lipid metabolism.
Collapse
Affiliation(s)
- Keiji Kawatani
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Marie-Louise Holm
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Skylar C Starling
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- SciNeuro Pharmaceuticals, Rockville, MD, 20850, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Peizhou Jiang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yangying Jiang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Samantha K Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ni Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Bhaskar Roy
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ralph B Perkerson
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Hanmei Bao
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
74
|
Dash S, Ueda T, Komuro A, Honda M, Sugisawa R, Okada H. Deoxycytidine kinase inactivation enhances gemcitabine resistance and sensitizes mitochondrial metabolism interference in pancreatic cancer. Cell Death Dis 2024; 15:131. [PMID: 38346958 PMCID: PMC10861559 DOI: 10.1038/s41419-024-06531-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most lethal forms of cancer. Although in the last decade, an increase in 5-year patient survival has been observed, the mortality rate remains high. As a first-line treatment for PDAC, gemcitabine alone or in combination (gemcitabine plus paclitaxel) has been used; however, drug resistance to this regimen is a growing issue. In our previous study, we reported MYC/glutamine dependency as a therapeutic target in gemcitabine-resistant PDAC secondary to deoxycytidine kinase (DCK) inactivation. Moreover, enrichment of oxidative phosphorylation (OXPHOS)-associated genes was a common property shared by PDAC cell lines, and patient clinical samples coupled with low DCK expression was also demonstrated, which implicates DCK in cancer metabolism. In this article, we reveal that the expression of most genes encoding mitochondrial complexes is remarkably upregulated in PDAC patients with low DCK expression. The DCK-knockout (DCK KO) CFPAC-1 PDAC cell line model reiterated this observation. Particularly, OXPHOS was functionally enhanced in DCK KO cells as shown by a higher oxygen consumption rate and mitochondrial ATP production. Electron microscopic observations revealed abnormal mitochondrial morphology in DCK KO cells. Furthermore, DCK inactivation exhibited reactive oxygen species (ROS) reduction accompanied with ROS-scavenging gene activation, such as SOD1 and SOD2. SOD2 inhibition in DCK KO cells clearly induced cell growth suppression. In combination with increased anti-apoptotic gene BCL2 expression in DCK KO cells, we finally reveal that venetoclax and a mitochondrial complex I inhibitor are therapeutically efficacious for DCK-inactivated CFPAC-1 cells in in vitro and xenograft models. Hence, our work provides insight into inhibition of mitochondrial metabolism as a novel therapeutic approach to overcome DCK inactivation-mediated gemcitabine resistance in PDAC patient treatment.
Collapse
Affiliation(s)
- Suman Dash
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Ryoichi Sugisawa
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, 589-8511, Japan.
- Anti-aging Center, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan.
| |
Collapse
|
75
|
Cheng CJ, Nizar JM, Dai DF, Huang CL. Transport activity regulates mitochondrial bioenergetics and biogenesis in renal tubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.04.578838. [PMID: 38370657 PMCID: PMC10871199 DOI: 10.1101/2024.02.04.578838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Renal tubules are featured with copious mitochondria and robust transport activity. Mutations in mitochondrial genes cause congenital renal tubulopathies, and changes in transport activity affect mitochondrial morphology, suggesting mitochondrial function and transport activity are tightly coupled. Current methods of using bulk kidney tissues or cultured cells to study mitochondrial bioenergetics are limited. Here, we optimized an extracellular flux analysis (EFA) to study mitochondrial respiration and energy metabolism using microdissected mouse renal tubule segments. EFA detects mitochondrial respiration and glycolysis by measuring oxygen consumption and extracellular acidification rates, respectively. We show that both measurements positively correlate with sample sizes of a few centimeter-length renal tubules. The thick ascending limbs (TALs) and distal convoluted tubules (DCTs) predominantly utilize glucose/pyruvate as energy substrates, whereas proximal tubules (PTs) are significantly much less so. Acute inhibition of TALs' transport activity by ouabain treatment reduces basal and ATP-linked mitochondrial respiration. Chronic inhibition of transport activity by 2-week furosemide treatment or deletion of with-no-lysine kinase 4 (Wnk4) decreases maximal mitochondrial capacity. In addition, chronic inhibition downregulates mitochondrial DNA mass and mitochondrial length/density in TALs and DCTs. Conversely, gain-of-function Wnk4 mutation increases maximal mitochondrial capacity and mitochondrial length/density without increasing mitochondrial DNA mass. In conclusion, EFA is a sensitive and reliable method to investigate mitochondrial functions in isolated renal tubules. Transport activity tightly regulates mitochondrial bioenergetics and biogenesis to meet the energy demand in renal tubules. The system allows future investigation into whether and how mitochondria contribute to tubular remodeling adapted to changes in transport activity. Key points A positive correlation between salt reabsorption and oxygen consumption in mammalian kidneys hints at a potential interaction between transport activity and mitochondrial respiration in renal tubules.Renal tubules are heterogeneous in transport activity and mitochondrial metabolism, and traditional assays using bulk kidney tissues cannot provide segment-specific information.Here, we applied an extracellular flux analysis to investigate mitochondrial respiration and energy metabolism in isolated renal tubules. This assay is sensitive in detecting oxygen consumption and acid production in centimeter-length renal tubules and reliably recapitulates segment-specific metabolic features.Acute inhibition of transport activity reduces basal and ATP-linked mitochondrial respirations without changing maximal mitochondrial respiratory capacity. Chronic alterations of transport activity further adjust maximal mitochondrial respiratory capacity via regulating mitochondrial biogenesis or non-transcriptional mechanisms.Our findings support the concept that renal tubular cells finely adjust mitochondrial bioenergetics and biogenesis to match the new steady state of transport activity.
Collapse
|
76
|
Hurtado KA, Schnellmann RG. Mitophagy regulates mitochondrial number following pharmacological induction of mitochondrial biogenesis in renal proximal tubule cells. Front Pharmacol 2024; 15:1344075. [PMID: 38375036 PMCID: PMC10875001 DOI: 10.3389/fphar.2024.1344075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/21/2024] Open
Abstract
Background: Mitochondrial biogenesis (MB) induction through the activation of the 5-Hydroxytriptamine (5-HT) 1F receptor (HTR1F) is a promising mechanism for the treatment of diseases characterized by mitochondrial dysfunction, such as acute kidney injury (AKI). While several studies report pharmacological activation of MB in the proximal tubule, it is unclear how the proximal tubule regulates itself once the pharmacological activation is removed. Mitophagy is the process of selective mitochondria degradation. We hypothesize that mitophagy decreases mitochondrial number after pharmacological stimulation and restore mitochondrial homeostasis. Methods: Renal proximal tubules were treated at time 0hr with LY344864 or vehicle for 24 h and then removed. LY344864, a selective HTR1F agonist, induces MB in renal proximal tubules as previously reported (Gibbs et al., Am J Physiol Renal Physiol, 2018, 314(2), F260-F268). Vehicle and pharmacological reagents were added at the 24 h time point. Electron microscopy was used to assess mitochondrial morphology, number, and autolysosomes. Seahorse Bioscience XF-96 extracellular flux analyzer was used to measure maximal mitochondrial oxygen consumption rates (FCCP-OCR), a functional marker of MB. Results: LY344864 treatment increased FCCP-OCR, phosphorylation of protein kinase B (AKT), peroxisome proliferator-activated receptor γ coactivator-1alpha (PGC-1α), and mitochondrial number after 24 h. These endpoints decreased to baseline 24 h after LY344864 removal. Treatment with ROC-325, an autophagy inhibitor, increased Sequestosome-1 (SQSTM1/P62) and microtubule-associated protein-1 light chain 3 (LC3B) after 24 h of treatment. Also, ROC-325 treatment sustained the elevated mitochondrial number after LY344864 pre-treatment and removal. Conclusion: These data revealed that inhibition of autophagy extends elevated mitochondrial number and function by preventing the lysosomal degradation of mitochondria after the removal of LY344864.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
- Southern Arizona VA Health Care System, Tucson, AZ, United States
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
77
|
Baruah N, Haajanen R, Rahman MT, Pirttilä AM, Koskimäki JJ. Biosynthesis of polyhydroxybutyrate by Methylorubrum extorquens DSM13060 is essential for intracellular colonization in plant endosymbiosis. FRONTIERS IN PLANT SCIENCE 2024; 15:1302705. [PMID: 38390299 PMCID: PMC10883064 DOI: 10.3389/fpls.2024.1302705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/15/2024] [Indexed: 02/24/2024]
Abstract
Methylorubrum extorquens DSM13060 is an endosymbiont that lives in the cells of shoot tip meristems. The bacterium is methylotrophic and consumes plant-derived methanol for the production of polyhydroxybutyrate (PHB). The PHB provides protection against oxidative stress for both host and endosymbiont cells through its fragments, methyl-esterified 3-hydroxybutyrate (ME-3HB) oligomers. We evaluated the role of the genes involved in the production of ME-3HB oligomers in the host colonization by the endosymbiont M. extorquens DSM13060 through targeted genetic mutations. The strains with deletions in PHB synthase (phaC), PHB depolymerase (phaZ1), and a transcription factor (phaR) showed altered PHB granule characteristics, as ΔphaC had a significantly low number of granules, ΔphaR had a significantly increased number of granules, and ΔphaZ1 had significantly large PHB granules in the bacterial cells. When the deletion strains were exposed to oxidative stress, the ΔphaC strain was sensitive to 10 mM HO· and 20 mM H2O2. The colonization of the host, Scots pine (Pinus sylvestris L.), by the deletion strains varied greatly. The deletion strain ΔphaR colonized the host mainly intercellularly, whereas the ΔphaZ1 strain was a slightly poorer colonizer than the control. The deletion strain ΔphaC lacked the colonization potential, living mainly on the surfaces of the epidermis of pine roots and shoots in contrast to the control, which intracellularly colonized all pine tissues within the study period. In earlier studies, deletions within the PHB metabolic pathway have had a minor effect on plant colonization by rhizobia. We have previously shown the association between ME-3HB oligomers, produced by PhaC and PhaZ1, and the ability to alleviate host-generated oxidative stress during plant infection by the endosymbiont M. extorquens DSM13060. Our current results show that the low capacity for PHB synthesis leads to poor tolerance of oxidative stress and loss of colonization potential by the endosymbiont. Altogether, our findings demonstrate that the metabolism of PHB in M. extorquens DSM13060 is an important trait in the non-rhizobial endosymbiosis.
Collapse
Affiliation(s)
- Namrata Baruah
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
| | - Roosa Haajanen
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
| | - Mohammad Tanvir Rahman
- Disease Networks, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | | | - Janne J Koskimäki
- Ecology and Genetics Research Unit, University of Oulu, Oulu, Finland
| |
Collapse
|
78
|
You W, Knoops K, Boesten I, Berendschot TTJM, van Zandvoort MAMJ, Benedikter BJ, Webers CAB, Reutelingsperger CPM, Gorgels TGMF. A time window for rescuing dying retinal ganglion cells. Cell Commun Signal 2024; 22:88. [PMID: 38297331 PMCID: PMC10832163 DOI: 10.1186/s12964-023-01427-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) degeneration and death cause vision loss in patients with glaucoma. Regulated cell death, once initiated, is generally considered to be an irreversible process. Recently, we showed that, by timely removing the cell death stimulus, stressed neuronal PC12 cells can recover from phosphatidylserine (PS) exposure, nuclear shrinkage, DNA damage, mitochondrial fragmentation, mitochondrial membrane potential loss, and retraction of neurites, all hallmarks of an activated cell death program. Whether the cell death process can be reversed in neurons of the central nervous system, like RGCs, is still unknown. Here, we studied reversibility of the activated cell death program in primary rat RGCs (prRGCs). METHODS prRGCs were exposed to ethanol (5%, vol/vol) to induce cell death. At different stages of the cell death process, ethanol was removed by washing and injured prRGCs were further cultured in fresh medium to see whether they recovered. The dynamics of single cells were monitored by high-resolution live-cell spinning disk microscopy. PS exposure, mitochondrial structure, membrane potential, and intracellular Ca2+ were revealed by annexin A5-FITC, Mito-tracker, TMRM, and Fluo 8-AM staining, respectively. The distribution of cytochrome c was investigated by immunofluorescence. The ultrastructure of mitochondria was studied by electron microscopy. RESULTS Analysis of temporal relationships between mitochondrial changes and PS exposure showed that fragmentation of the mitochondrial network and loss of mitochondrial membrane potential occurred before PS exposure. Mitochondrial changes proceeded caspase-independently, while PS exposure was caspase dependent. Interestingly, prRGCs recovered quickly from these mitochondrial changes but not from PS exposure at the plasma membrane. Correlative light and electron microscopy showed that stress-induced decrease in mitochondrial area, length and cristae number was reversible. Intracellular Ca2+ was elevated during this stage of reversible mitochondrial injury, but there was no sign of mitochondrial cytochrome c release. CONCLUSIONS Our study demonstrates that RGCs with impaired mitochondrial structure and function can fully recover if there is no mitochondrial cytochrome c release yet, and no PS is exposed at the plasma membrane. This finding indicates that there is a time window for rescuing dying or injured RGCs, by simply removing the cell death stimulus. Video Abstract.
Collapse
Affiliation(s)
- Wenting You
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Department of Mental Health and Neuroscience, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Kèvin Knoops
- The Microscopy CORE lab, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Iris Boesten
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Tos T J M Berendschot
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Marc A M J van Zandvoort
- Department of Molecular Cell Biology, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Institute of Molecular Cardiovascular Research (IMCAR), Universitätsklinikum Aachen, 52074, Aachen, Germany
| | - Birke J Benedikter
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands.
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands.
| |
Collapse
|
79
|
Javed Z, Shin DH, Pan W, White SR, Kim YS, Elhaw AT, Kamlapurkar S, Cheng YY, Benson JC, Abdelnaby AE, Phaëton R, Wang HG, Yang S, Sullivan ML, St.Croix CM, Watkins SC, Mullett SJ, Gelhaus SL, Lee N, Coffman LG, Aird KM, Trebak M, Mythreye K, Walter V, Hempel N. Alternative splice variants of the mitochondrial fission protein DNM1L/Drp1 regulate mitochondrial dynamics and tumor progression in ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.20.558501. [PMID: 37790404 PMCID: PMC10542115 DOI: 10.1101/2023.09.20.558501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Aberrant mitochondrial fission/fusion dynamics have been reported in cancer cells. While post translational modifications are known regulators of the mitochondrial fission/fusion machinery, we show that alternative splice variants of the fission protein Drp1 (DNM1L) have specific and unique roles in cancer, adding to the complexity of mitochondrial fission/fusion regulation in tumor cells. Ovarian cancer specimens express an alternative splice transcript variant of Drp1 lacking exon 16 of the variable domain, and high expression of this splice variant relative to other transcripts is associated with poor patient outcome. Unlike the full-length variant, expression of Drp1 lacking exon 16 leads to decreased association of Drp1 to mitochondrial fission sites, more fused mitochondrial networks, enhanced respiration, and TCA cycle metabolites, and is associated with a more metastatic phenotype in vitro and in vivo. These pro-tumorigenic effects can also be inhibited by specific siRNA-mediated inhibition of the endogenously expressed transcript lacking exon 16. Moreover, lack of exon 16 abrogates mitochondrial fission in response to pro-apoptotic stimuli and leads to decreased sensitivity to chemotherapeutics. These data emphasize the significance of the pathophysiological consequences of Drp1 alternative splicing and divergent functions of Drp1 splice variants, and strongly warrant consideration of Drp1 splicing in future studies.
Collapse
Affiliation(s)
- Zaineb Javed
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Dong Hui Shin
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Weihua Pan
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
| | - Sierra R. White
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, PA, USA
| | - Yeon Soo Kim
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Amal Taher Elhaw
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Shriya Kamlapurkar
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
| | - Ya-Yun Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
| | - J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA
| | - Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA
| | - Rébécca Phaëton
- Department of Obstetrics & Gynecology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Hong-Gang Wang
- Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Health Sciences Mass Spectrometry Core, University of Pittsburgh, PA, USA
| | - Mara L.G. Sullivan
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, PA, USA; Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Claudette M. St.Croix
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, PA, USA; Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Simon C. Watkins
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, PA, USA; Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Steven J. Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Health Sciences Mass Spectrometry Core, University of Pittsburgh, PA, USA
| | - Stacy L. Gelhaus
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA; Health Sciences Mass Spectrometry Core, University of Pittsburgh, PA, USA
| | - Nam Lee
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, PA, USA; Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Lan G. Coffman
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
| | - Katherine M. Aird
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Mohamed Trebak
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, PA, USA
| | - Karthikeyan Mythreye
- Department of Pathology and O’Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vonn Walter
- Department of Public Health Sciences, Division of Biostatistics and Bioinformatics and Department of Biochemistry and Molecular Biology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Nadine Hempel
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, PA, USA
| |
Collapse
|
80
|
Natrus L, Lisakovska O, Smirnov A, Osadchuk Y, Savosko S, Klys Y. Could the Propionic Acid Treatment in Combination with Metformin be Safe for the Small Intestine of Diabetic Rats? Endocr Metab Immune Disord Drug Targets 2024; 24:1335-1345. [PMID: 38265384 DOI: 10.2174/0118715303273125231121062111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/10/2023] [Accepted: 09/22/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Effects of propionic acid (PA) on the cellular and molecular processes in the small intestine under type 2 diabetes mellitus (T2DM)-induced endoplasmic reticulum (ER) stress remain incompletely studied. OBJECTIVES The aim of the study was to assess the state of unfolded protein response (UPR) system in the small intestine of diabetic rats and to explore PA's influence on metformin treatment. METHODS Male Wistar rats were divided into 1) control and 2) T2DM groups, and groups receiving (14 days, orally) 3) metformin (60 mg/kg), 4) PA (60 mg/kg), and 5) PA+metformin. Western blotting, RT-PCR, and transmission electron microscopy were performed. RESULTS We found that T2DM induced elevation of ER intermembrane space and UPR overactivation based on increased GRP78, ATF6 and PERK levels in small intestine. Metformin treatment led to a further UPR activation. PA supplementation partially restored enterocytes functioning via normalization of ATF6 and PERK content, while IRE1 level reached the maximum value, compared to all groups. The most pronounced effect of adaptation to the T2DMinduced ER stress was observed after combined metformin and PA action. In particular, decreased ER intermembrane space in enterocytes was detected compared to separate metformin and PA administration, which was accompanied by restored GRP78, PERK and IRE1 levels. CONCLUSION Our study proves the safety of additional therapy with propionic acid in combination with metformin for the functional state of small intestine. Due to its ability to modulate UPR signaling, PA may be considered a safe and perspective candidate for supportive therapy in T2DM, especially for neuroprotection.
Collapse
Affiliation(s)
- Larysa Natrus
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomoletz National Medical University, Kyiv 03115, Ukraine
| | - Olha Lisakovska
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv 01054, Ukraine
| | - Anton Smirnov
- Department of Socio-Humanitarian and Biomedical Sciences, Kharkiv Institute of Medicine and Biomedical Sciences, Kharkiv 61002, Ukraine
| | - Yuliia Osadchuk
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomoletz National Medical University, Kyiv 03115, Ukraine
| | - Serhyi Savosko
- Department of Histology and Embryology, Bogomoletz National Medical University, Kyiv 03115, Ukraine
| | - Yuliia Klys
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomoletz National Medical University, Kyiv 03115, Ukraine
| |
Collapse
|
81
|
Crabtree A, Neikirk K, Marshall AG, Vang L, Whiteside AJ, Williams Q, Altamura CT, Owens TC, Stephens D, Shao B, Koh A, Killion M, Lopez EG, Lam J, Rodriguez B, Mungai M, Stanley J, Dean ED, Koh HJ, Gaddy JA, Scudese E, Sweetwyne MT, Davis J, Zaganjor E, Murray SA, Katti P, Damo SM, Vue Z, Hinton A. Defining Mitochondrial Cristae Morphology Changes Induced by Aging in Brown Adipose Tissue. Adv Biol (Weinh) 2024; 8:e2300186. [PMID: 37607124 PMCID: PMC10869235 DOI: 10.1002/adbi.202300186] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/20/2023] [Indexed: 08/24/2023]
Abstract
Mitochondria are required for energy production and even give brown adipose tissue (BAT) its characteristic color due to their high iron content and abundance. The physiological function and bioenergetic capacity of mitochondria are connected to the structure, folding, and organization of its inner-membrane cristae. During the aging process, mitochondrial dysfunction is observed, and the regulatory balance of mitochondrial dynamics is often disrupted, leading to increased mitochondrial fragmentation in aging cells. Therefore, it is hypothesized that significant morphological changes in BAT mitochondria and cristae will be present with aging. A quantitative 3D electron microscopy approach is developed to map cristae network organization in mouse BAT to test this hypothesis. Using this methodology, the 3D morphology of mitochondrial cristae is investigated in adult (3-month) and aged (2-year) murine BAT tissue via serial block face-scanning electron microscopy (SBF-SEM) and 3D reconstruction software for manual segmentation, analysis, and quantification. Upon investigation, an increase is found in mitochondrial volume, surface area, and complexity and decreased sphericity in aged BAT, alongside significant decreases in cristae volume, area, perimeter, and score. Overall, these data define the nature of the mitochondrial structure in murine BAT across aging.
Collapse
Affiliation(s)
- Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron J Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Qiana Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Christopher T Altamura
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Trinity Celeste Owens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mason Killion
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Ben Rodriguez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jade Stanley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - E Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ho-Jin Koh
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jennifer A Gaddy
- Department of Biological Sciences, Tennessee State University, Nashville, TN, 37209, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH), Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, 22290-240, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Rio de Janeiro, 22290-240, Brazil
| | - Mariya T Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience, Pharmacology, Meharry Medical College, Nashville, TN, 37208, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sandra A Murray
- Department of Cell Biology, University of Pittsburgh, Pittsburg, PA, 15261, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
82
|
Stephens DC, Mungai M, Crabtree A, Beasley HK, Garza-Lopez E, Vang L, Neikirk K, Vue Z, Vue N, Marshall AG, Turner K, Shao JQ, Sarker B, Murray S, Gaddy JA, Davis J, Damo SM, Hinton AO. Protocol for isolating mice skeletal muscle myoblasts and myotubes via differential antibody validation. STAR Protoc 2023; 4:102591. [PMID: 37938976 PMCID: PMC10663959 DOI: 10.1016/j.xpro.2023.102591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/06/2023] [Accepted: 09/01/2023] [Indexed: 11/10/2023] Open
Abstract
Isolation of skeletal muscles allows for the exploration of many complex diseases. Here, we present a protocol for isolating mice skeletal muscle myoblasts and myotubes that have been differentiated through antibody validation. We describe steps for collecting and preparing murine skeletal tissue, myoblast cell maintenance, plating, and cell differentiation. We then detail procedures for cell incubation, immunostaining, slide preparation and storage, and imaging for immunofluorescence validation.
Collapse
Affiliation(s)
- Dominique C Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Neng Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Kyrin Turner
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA
| | - Jian-Qiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA 52242, USA
| | - Bishnu Sarker
- School of Applied Computational Sciences, Meharry Medical College, Nashville, TN 37232, USA
| | - Sandra Murray
- Department of Cell Biology, College of Medicine, University of Pittsburgh, Pittsburgh, TN 15260, USA
| | - Jennifer A Gaddy
- Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, TN, USA; Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Jamaine Davis
- Department of Biochemistry and Cancer Biology. Meharry Medical College, Nashville, TN, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37232, USA.
| | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
83
|
Elagamy SH, Adly L, Abdel Hamid MA. Smartphone based colorimetric approach for quantitative determination of uric acid using Image J. Sci Rep 2023; 13:21888. [PMID: 38081872 PMCID: PMC10713523 DOI: 10.1038/s41598-023-48962-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Recently, significant attention has been directed towards digital image colorimetry DIC using mobile applications or available software programs, which offer the advantage of analyzing samples without the need for sophisticated instruments. One such image processing program is Image J, widely used for obtaining quantitative information from scientific images. Image J could measure the color intensities by quantifying of the RGB (red-green-blue) gray levels across the images of colored substances. These values are correlated to the color intensities through conversion to CMY (cyan-magenta-yellow) values which are proportional to the color intensities. The objective of this study is to develop an innovative analytical method for the quantitative determination of uric acid using Image J for color quantification. Image J was utilized to analyze images captured by smart phone for successive concentrations of uric acid that were previously treated with phosphotungstate to develop a blue color. The proposed method has been applied for determination of uric acid in real urine using standard addition method and the results were compared with UV/VIS spectrophotometry as a reference method. In this research, we will also assess the effectiveness of quantitative analysis using Image J in comparison to a mobile application, namely RGB Color Detector.
Collapse
Affiliation(s)
- Samar H Elagamy
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Latifa Adly
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mohamed Ahmed Abdel Hamid
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
- Department of pharmaceutical chemistry, Al Salam university, Tanta, Egypt
| |
Collapse
|
84
|
Neikirk K, Lopez EG, Marshall AG, Alghanem A, Krystofiak E, Kula B, Smith N, Shao J, Katti P, Hinton A. Call to action to properly utilize electron microscopy to measure organelles to monitor disease. Eur J Cell Biol 2023; 102:151365. [PMID: 37864884 DOI: 10.1016/j.ejcb.2023.151365] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023] Open
Abstract
This review provides an overview of the current methods for quantifying mitochondrial ultrastructure, including cristae morphology, mitochondrial contact sites, and recycling machinery and a guide to utilizing electron microscopy to effectively measure these organelles. Quantitative analysis of mitochondrial ultrastructure is essential for understanding mitochondrial biology and developing therapeutic strategies for mitochondrial-related diseases. Techniques such as transmission electron microscopy (TEM) and serial block face-scanning electron microscopy, as well as how they can be combined with other techniques including confocal microscopy, super-resolution microscopy, and correlative light and electron microscopy are discussed. Beyond their limitations and challenges, we also offer specific magnifications that may be best suited for TEM analysis of mitochondrial, endoplasmic reticulum, and recycling machinery. Finally, perspectives on future quantification methods are offered.
Collapse
Affiliation(s)
- Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Edgar-Garza Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Ahmad Alghanem
- King Abdullah International Medical Research Center (KAIMRC), Ali Al Arini, Ar Rimayah, Riyadh 11481, Saudi Arabia
| | - Evan Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Bartosz Kula
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester 14642, USA
| | - Nathan Smith
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester 14642, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
85
|
Marshall AG, Neikirk K, Stephens D, Garza-Lopez E, Vue Z, Beasley HK, Janumyan Doe Y, Campbell D, Fears L, Alghanem A, Spencer EC, Scudese E, Owens B, Vang C, Morton DJ, Conley Z, Hinton A. A workshop on mitochondria for students to improve understanding of science and hypothesis forming. ADVANCES IN PHYSIOLOGY EDUCATION 2023; 47:823-830. [PMID: 37650143 PMCID: PMC10854815 DOI: 10.1152/advan.00116.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
There remains a clear deficiency in recruiting middle school students in science, technology, engineering, mathematics, and medicine fields, especially for those students entering physiology from underrepresented backgrounds. A large part of this may be arising from a disconnect between how science is typically practiced at a collegiate and K-12 level. Here, we have envisioned mitochondria and their diverse subcellular structures as an involver for middle school students. We present the framework for a workshop that familiarizes students with mitochondria, employing three-dimensional visual-spatial learning and real-time critical thinking and hypothesis forming. This workshop had the goal of familiarizing middle school students with the unique challenges the field currently faces and better understanding the actuality of being a scientist through critical analysis including hypothesis forming. Findings show that middle school students responded positively to the program and felt as though they had a better understanding of mitochondria. Future implications for hands-on programs to involve underrepresented students in science are discussed, as well as potential considerations to adapt it for high school and undergraduate students.NEW & NOTEWORTHY Here we employ a workshop that utilizes blended and tactile learning to teach middle schoolers about mitochondrial structure. By creating an approachable and fun workshop that can be utilized for middle school students, we seek to encourage them to join a career in physiology.
Collapse
Affiliation(s)
- Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Yelena Janumyan Doe
- Collaborative for STEM Education and Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, Tennessee, United States
| | - Desmond Campbell
- Collaborative for STEM Education and Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, Tennessee, United States
| | - Letimicia Fears
- Collaborative for STEM Education and Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, Tennessee, United States
| | - Ahmad Alghanem
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Elsie C Spencer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Teachers College, Columbia University, New York, New York, United States
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
- Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Rio de Janeiro, Brazil
| | - Beverly Owens
- Department of Chemistry, Cleveland Early College High School, Shelby, North Carolina, United States
| | - Chia Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Counseling Department, New Highlands University, Las Vegas, New Mexico, United States
| | - Derrick J Morton
- Kaiser Permanente Bernard J. Tyson School of Medicine, Department of Biomedical Science, Pasadena, California, United States
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States
| | - Zachary Conley
- Collaborative for STEM Education and Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, Tennessee, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
86
|
Hurtado KA, Janda J, Schnellmann RG. Lasmiditan restores mitochondrial quality control mechanisms and accelerates renal recovery after ischemia-reperfusion injury. Biochem Pharmacol 2023; 218:115855. [PMID: 37866804 PMCID: PMC10872401 DOI: 10.1016/j.bcp.2023.115855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND Mitochondrial dysfunction is a well-established result of acute kidney injury (AKI). Previously, we identified that 5-hydroxytryptamine 1F (5-HT1F) receptor agonism with lasmiditan induces mitochondrial biogenesis (MB) and improves renal vasculature and function in an AKI mouse model. We hypothesize that lasmiditan also modulates mitochondrial dynamics and mitophagy in a mouse model of AKI. METHODS Male mice were subjected to renal ischemia/reperfusion (I/R) and treated daily with lasmiditan (0.3 mg/kg) or vehicle beginning 24 h after injury for 3 or 6d. Serum creatinine was measured to estimate glomerular filtration. Electron microscopy was used to assess mitochondrial morphology and mitophagy. Mitochondrial-related protein were confirmed with immunoblotting. Mitochondrial function was assessed with ATP measurements. RESULTS Lasmiditan treatment improved mitochondrial and kidney recovery as early as 3d post-AKI, as evidenced by increased ATP, and decreased serum creatinine, respectively. Electron micrographs of renal cortices revealed that lasmiditan also decreased mitochondrial damage and increased mitochondrial area and size by 6d after I/R injury. Additionally, lasmiditan treatment increased mitolysosomes by 3d, indicating induction of mitophagy. Phosphorylation of mitophagy-related proteins were also increased in the renal cortices of lasmiditan-treated AKI mice 3d after I/R injury, whereas fusion-related proteins were increased at 6d after I/R injury. CONCLUSION These data reveal that lasmiditan accelerates renal recovery, restores normal mitochondrial membrane and cristae morphology, decreases excessive mitochondrial fission, and accelerates mitophagy post-AKI in a time-dependent manner, establishing mitochondrial function and recovery from AKI.
Collapse
Affiliation(s)
- Kevin A Hurtado
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Jaroslav Janda
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA; Southern Arizona VA Health Care System, Tucson, AZ, USA; Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
87
|
Vue Z, Garza‐Lopez E, Neikirk K, Katti P, Vang L, Beasley H, Shao J, Marshall AG, Crabtree A, Murphy AC, Jenkins BC, Prasad P, Evans C, Taylor B, Mungai M, Killion M, Stephens D, Christensen TA, Lam J, Rodriguez B, Phillips MA, Daneshgar N, Koh H, Koh A, Davis J, Devine N, Saleem M, Scudese E, Arnold KR, Vanessa Chavarin V, Daniel Robinson R, Chakraborty M, Gaddy JA, Sweetwyne MT, Wilson G, Zaganjor E, Kezos J, Dondi C, Reddy AK, Glancy B, Kirabo A, Quintana AM, Dai D, Ocorr K, Murray SA, Damo SM, Exil V, Riggs B, Mobley BC, Gomez JA, McReynolds MR, Hinton A. 3D reconstruction of murine mitochondria reveals changes in structure during aging linked to the MICOS complex. Aging Cell 2023; 22:e14009. [PMID: 37960952 PMCID: PMC10726809 DOI: 10.1111/acel.14009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/01/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023] Open
Abstract
During aging, muscle gradually undergoes sarcopenia, the loss of function associated with loss of mass, strength, endurance, and oxidative capacity. However, the 3D structural alterations of mitochondria associated with aging in skeletal muscle and cardiac tissues are not well described. Although mitochondrial aging is associated with decreased mitochondrial capacity, the genes responsible for the morphological changes in mitochondria during aging are poorly characterized. We measured changes in mitochondrial morphology in aged murine gastrocnemius, soleus, and cardiac tissues using serial block-face scanning electron microscopy and 3D reconstructions. We also used reverse transcriptase-quantitative PCR, transmission electron microscopy quantification, Seahorse analysis, and metabolomics and lipidomics to measure changes in mitochondrial morphology and function after loss of mitochondria contact site and cristae organizing system (MICOS) complex genes, Chchd3, Chchd6, and Mitofilin. We identified significant changes in mitochondrial size in aged murine gastrocnemius, soleus, and cardiac tissues. We found that both age-related loss of the MICOS complex and knockouts of MICOS genes in mice altered mitochondrial morphology. Given the critical role of mitochondria in maintaining cellular metabolism, we characterized the metabolomes and lipidomes of young and aged mouse tissues, which showed profound alterations consistent with changes in membrane integrity, supporting our observations of age-related changes in muscle tissues. We found a relationship between changes in the MICOS complex and aging. Thus, it is important to understand the mechanisms that underlie the tissue-dependent 3D mitochondrial phenotypic changes that occur in aging and the evolutionary conservation of these mechanisms between Drosophila and mammals.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Kit Neikirk
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
| | - Larry Vang
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Heather Beasley
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Jianqiang Shao
- Central Microscopy Research FacilityUniversity of IowaIowaIowa CityUSA
| | - Andrea G. Marshall
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Amber Crabtree
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Alexandria C. Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Brenita C. Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Chantell Evans
- Department of Cell BiologyDuke University School of MedicineNorth CarolinaDurhamUSA
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of FloridaFloridaGainesvilleUSA
| | - Margaret Mungai
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Mason Killion
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Dominique Stephens
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Jacob Lam
- Department of Internal MedicineUniversity of IowaIowaIowa CityUSA
| | | | - Mark A. Phillips
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Nastaran Daneshgar
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Ho‐Jin Koh
- Department of Biological SciencesTennessee State UniversityTennesseeNashvilleUSA
| | - Alice Koh
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience, and PharmacologyMeharry Medical CollegeTennesseeNashvilleUSA
| | - Nina Devine
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Mohammad Saleem
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO)Rio de JaneiroBrazil
- Sport Sciences and Exercise Laboratory (LaCEE)Catholic University of Petrópolis (UCP)PetrópolisState of Rio de JaneiroBrazil
| | - Kenneth Ryan Arnold
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Valeria Vanessa Chavarin
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Ryan Daniel Robinson
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | | | - Jennifer A. Gaddy
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Medicine Health and SocietyVanderbilt UniversityTennesseeNashvilleUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Veterans AffairsTennessee Valley Healthcare SystemsTennesseeNashvilleUSA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and PathologyUniversity of WashingtonWashingtonSeattleUSA
| | - Genesis Wilson
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Elma Zaganjor
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Cristiana Dondi
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | | | - Brian Glancy
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of HealthMarylandBethesdaUSA
| | - Annet Kirabo
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research CenterUniversity of Texas at El PasoTexasEl PasoUSA
| | - Dao‐Fu Dai
- Department of PathologyUniversity of Johns Hopkins School of MedicineMarylandBaltimoreUSA
| | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Sandra A. Murray
- Department of Cell Biology, School of MedicineUniversity of PittsburghPennsylvaniaPittsburghUSA
| | - Steven M. Damo
- Department of Life and Physical SciencesFisk UniversityTennesseeNashvilleUSA
- Center for Structural BiologyVanderbilt UniversityTennesseeNashvilleUSA
| | - Vernat Exil
- Department of Pediatrics, Carver College of MedicineUniversity of IowaIowaIowa CityUSA
- Department of Pediatrics, Division of CardiologySt. Louis University School of MedicineMissouriSt. LouisUSA
| | - Blake Riggs
- Department of BiologySan Francisco State UniversityCaliforniaSan FranciscoUSA
| | - Bret C. Mobley
- Department of PathologyVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jose A. Gomez
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Antentor Hinton
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| |
Collapse
|
88
|
Li J, Xue C, Yang H, Zhang J, Li G, Li J, Kuang F, Chen J, Zhang S, Gao F, Kou Z, Zhang X, Dong L. Simulated weightlessness induces hippocampal insulin resistance and cognitive impairment. Life Sci 2023; 333:122112. [PMID: 37758017 DOI: 10.1016/j.lfs.2023.122112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Growing evidence highlights the potential consequences of long-term spaceflight, including gray matter volume reduction and cognitive dysfunction with subclinical manifestations of diabetes mellitus among astronauts, but the underlying mechanisms remain unknown. In this study, we found that long-term simulated weightlessness induced hippocampal insulin resistance and subsequent neuronal damage and cognitive impairment in rats. Rats subjected to 4-week tail suspension exhibited peripheral insulin resistance, evidenced by increased fasting blood glucose and abnormal glucose tolerance and insulin tolerance, alongside reduced spontaneous activity and impaired recognition memory. In addition, 4 weeks of simulated weightlessness induced neuronal apoptosis and degeneration in the hippocampus, as evidenced by increased TUNEL and Fluoro-Jade B staining-positive neurons. Mechanistically, insulin-stimulated hippocampal Akt phosphorylation was decreased, while PTEN, the negative regulator of insulin signaling, was increased in the hippocampus in tail-suspended rats. Interestingly, treatment with berberine, an insulin sensitizer, partly reversed the above-mentioned effects induced by simulated weightlessness. These data suggest that long-term simulated weightlessness induces cognitive impairment as well as neuronal apoptosis and neural degeneration, partially through hippocampal insulin resistance via PTEN up-regulation. Berberine treatment attenuates hippocampal insulin resistance and improves cognitive function.
Collapse
Affiliation(s)
- Jiahui Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China; Department of Psychology, Air Force Hospital, Western Theater Command, Chengdu, China
| | - Caiyan Xue
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Hongyan Yang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Jiaxin Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Guohua Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Jijun Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Fang Kuang
- Department of Neurobiology, Air Force Medical University, Xi'an, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China
| | - Shu Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China
| | - Zhenzhen Kou
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Air Force Medical University, Xi'an, China.
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China.
| | - Ling Dong
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Air Force Medical University, Xi'an, China.
| |
Collapse
|
89
|
Child JR, Hofler AC, Chen Q, Yang BH, Kristofich J, Zheng T, Hannigan MM, Elles AL, Reid DW, Nicchitta CV. Examining SRP pathway function in mRNA localization to the endoplasmic reticulum. RNA (NEW YORK, N.Y.) 2023; 29:1703-1724. [PMID: 37643813 PMCID: PMC10578483 DOI: 10.1261/rna.079643.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/17/2023] [Indexed: 08/31/2023]
Abstract
Signal recognition particle (SRP) pathway function in protein translocation across the endoplasmic reticulum (ER) is well established; its role in RNA localization to the ER remains, however, unclear. In current models, mRNAs undergo translation- and SRP-dependent trafficking to the ER, with ER localization mediated via interactions between SRP-bound translating ribosomes and the ER-resident SRP receptor (SR), a heterodimeric complex comprising SRA, the SRP-binding subunit, and SRB, an integral membrane ER protein. To study SRP pathway function in RNA localization, SR knockout (KO) mammalian cell lines were generated and the consequences of SR KO on steady-state and dynamic mRNA localization examined. CRISPR/Cas9-mediated SRPRB KO resulted in profound destabilization of SRA. Pairing siRNA silencing of SRPRA in SRPRB KO cells yielded viable SR KO cells. Steady-state mRNA compositions and ER-localization patterns in parental and SR KO cells were determined by cell fractionation and deep sequencing. Notably, steady-state cytosol and ER mRNA compositions and partitioning patterns were largely unaltered by loss of SR expression. To examine SRP pathway function in RNA localization dynamics, the subcellular trafficking itineraries of newly exported mRNAs were determined by 4-thiouridine (4SU) pulse-labeling/4SU-seq/cell fractionation. Newly exported mRNAs were distinguished by high ER enrichment, with ER localization being SR-independent. Intriguingly, under conditions of translation initiation inhibition, the ER was the default localization site for all newly exported mRNAs. These data demonstrate that mRNA localization to the ER can be uncoupled from the SRP pathway function and reopen questions regarding the mechanism of RNA localization to the ER.
Collapse
Affiliation(s)
- Jessica R Child
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Alex C Hofler
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Qiang Chen
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Brenda H Yang
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - JohnCarlo Kristofich
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Tianli Zheng
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Molly M Hannigan
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Andrew L Elles
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - David W Reid
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Christopher V Nicchitta
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
90
|
Vue Z, Neikirk K, Vang L, Garza-Lopez E, Christensen TA, Shao J, Lam J, Beasley HK, Marshall AG, Crabtree A, Anudokem J, Rodriguez B, Kirk B, Bacevac S, Barongan T, Shao B, Stephens DC, Kabugi K, Koh HJ, Koh A, Evans CS, Taylor B, Reddy AK, Miller-Fleming T, Actkins KV, Zaganjor E, Daneshgar N, Murray SA, Mobley BC, Damo SM, Gaddy JA, Riggs B, Wanjalla C, Kirabo A, McReynolds M, Gomez JA, Phillips MA, Exil V, Dai DF, Hinton A. Three-dimensional mitochondria reconstructions of murine cardiac muscle changes in size across aging. Am J Physiol Heart Circ Physiol 2023; 325:H965-H982. [PMID: 37624101 PMCID: PMC10977873 DOI: 10.1152/ajpheart.00202.2023] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/26/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
With sparse treatment options, cardiac disease remains a significant cause of death among humans. As a person ages, mitochondria breakdown and the heart becomes less efficient. Heart failure is linked to many mitochondria-associated processes, including endoplasmic reticulum stress, mitochondrial bioenergetics, insulin signaling, autophagy, and oxidative stress. The roles of key mitochondrial complexes that dictate the ultrastructure, such as the mitochondrial contact site and cristae organizing system (MICOS), in aging cardiac muscle are poorly understood. To better understand the cause of age-related alteration in mitochondrial structure in cardiac muscle, we used transmission electron microscopy (TEM) and serial block facing-scanning electron microscopy (SBF-SEM) to quantitatively analyze the three-dimensional (3-D) networks in cardiac muscle samples of male mice at aging intervals of 3 mo, 1 yr, and 2 yr. Here, we present the loss of cristae morphology, the inner folds of the mitochondria, across age. In conjunction with this, the three-dimensional (3-D) volume of mitochondria decreased. These findings mimicked observed phenotypes in murine cardiac fibroblasts with CRISPR/Cas9 knockout of Mitofilin, Chchd3, Chchd6 (some members of the MICOS complex), and Opa1, which showed poorer oxidative consumption rate and mitochondria with decreased mitochondrial length and volume. In combination, these data show the need to explore if loss of the MICOS complex in the heart may be involved in age-associated mitochondrial and cristae structural changes.NEW & NOTEWORTHY This article shows how mitochondria in murine cardiac changes, importantly elucidating age-related changes. It also is the first to show that the MICOS complex may play a role in outer membrane mitochondrial structure.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Trace A Christensen
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, Rochester, Minnesota, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, Iowa, United States
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Josephs Anudokem
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Taylor Barongan
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Dominique C Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee, United States
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Chantell S Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Anilkumar K Reddy
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tyne Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ky'Era V Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Jennifer A Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Tennessee Valley Healthcare Systems, United States Department of Veterans Affairs, Nashville, Tennessee, United States
| | - Blake Riggs
- Department of Biology at San Francisco State University, San Francisco, California, United States
| | - Celestine Wanjalla
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States
| | - Jose A Gomez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Mark A Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, Oregon, United States
| | - Vernat Exil
- Division of Cardiology, Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Dao-Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
91
|
Zhu Q, Jia Z, Song Y, Dou W, Scharf DH, Wu X, Xu Z, Guan W. Impact of PpSpi1, a glycosylphosphatidylinositol-anchored cell wall glycoprotein, on cell wall defects of N-glycosylation-engineered Pichia pastoris. mBio 2023; 14:e0061723. [PMID: 37606451 PMCID: PMC10653784 DOI: 10.1128/mbio.00617-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/14/2023] [Indexed: 08/23/2023] Open
Abstract
IMPORTANCE Engineering of biological pathways in various microorganisms is a promising direction for biotechnology. Since the existing microbial cells have evolved over a long period of time, any artificial engineering may cause some unexpected and harmful effects on them. Systematically studying and evaluating these engineered strains are very important and necessary. In order to produce therapeutic proteins with human-like N-glycan structures, much progress has been achieved toward the humanization of N-glycosylation pathways in yeasts. The properties of a P. pastoris strain with humanized N-glycosylation machinery were carefully evaluated in this study. Our work has identified a key glycoprotein (PpSpi1) responsible for the poor growth and morphological defects of this glycoengineered strain. Overexpression of PpSpi1 could significantly rescue the growth defect of the glycoengineered P. pastoris and facilitate its future industrial applications.
Collapse
Affiliation(s)
- Quanchao Zhu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zuyuan Jia
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuchao Song
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwang Dou
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Daniel Henry Scharf
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- China Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, China
| | - Xiaodan Wu
- Analysis Center of Agrobiology and Environmental Science of Zhejiang University, Hangzhou, China
| | - Zhihao Xu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjun Guan
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- China Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, China
| |
Collapse
|
92
|
Tao E, Wu Y, Hu C, Zhu Z, Ye D, Long G, Chen B, Guo R, Shu X, Zheng W, Zhang T, Jia X, Du X, Fang M, Jiang M. Early life stress induces irritable bowel syndrome from childhood to adulthood in mice. Front Microbiol 2023; 14:1255525. [PMID: 37849921 PMCID: PMC10577190 DOI: 10.3389/fmicb.2023.1255525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
Background Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal disorder. Traditionally, early life stress (ELS) is predisposed to IBS in adult. However, whether ELS induces IBS in early life remains unclear. Methods Separated cohort studies were conducted in neonatal male pups of C57BL/6 mice by maternal separation (MS) model. MS and non-separation mice were scheduled to be evaluated for prime IBS-phenotypes, including visceral hypersensitivity, intestinal motility, intestinal permeability, and anxiety-like behavior. Ileal contents and fecal samples were collected and analyzed by 16S rRNA gene sequencing and bacterial community analyses. Subcellular structures of intestinal epithelial, such as epithelial tight junctions and mitochondria, were observed under transmission electron microscopy. Results MS induced visceral hypersensitivity and decreased total intestinal transit time from childhood to adulthood. In addition, MS induced intestinal hyperpermeability and anxiety-like behavior from adolescence to adulthood. Besides, MS affected intestinal microbial composition from childhood to adulthood. Moreover, MS disrupted intestinal mitochondrial structure from childhood to adulthood. Conclusion The study showed for the first time that MS induced IBS from early life to adulthood in mice. The disrupted intestinal mitochondrial structure and the significant dysbiosis of intestinal microbiota in early life may contribute to the initiation and progress of IBS from early life to adulthood.
Collapse
Affiliation(s)
- Enfu Tao
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Department of Neonatology and NICU, Wenling Maternal and Child Health Care Hospital, Wenling, China
| | - Yuhao Wu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Chenmin Hu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Zhenya Zhu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Diya Ye
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Gao Long
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Bo Chen
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Rui Guo
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Xiaoli Shu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Wei Zheng
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Ting Zhang
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Xinyi Jia
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Xiao Du
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mizu Jiang
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| |
Collapse
|
93
|
Ruegsegger GN, Pataky MW, Simha S, Robinson MM, Klaus KA, Nair KS. High-intensity aerobic, but not resistance or combined, exercise training improves both cardiometabolic health and skeletal muscle mitochondrial dynamics. J Appl Physiol (1985) 2023; 135:763-774. [PMID: 37616334 PMCID: PMC10642518 DOI: 10.1152/japplphysiol.00405.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023] Open
Abstract
This study investigated how different exercise training modalities influence skeletal muscle mitochondrial dynamics. Healthy [average body mass index (BMI): 25.8 kg/m2], sedentary younger and older participants underwent 12 wk of supervised high-intensity aerobic interval training (HIIT; n = 13), resistance training (RT; n = 14), or combined training (CT; n = 11). Mitochondrial structure was assessed using transmission electron microscopy (TEM). Regulators of mitochondrial fission and fusion, cardiorespiratory fitness (V̇o2peak), insulin sensitivity via a hyperinsulinemic-euglycemic clamp, and muscle mitochondrial respiration were assessed. TEM showed increased mitochondrial volume, number, and perimeter following HIIT (P < 0.01), increased mitochondrial number following CT (P < 0.05), and no change in mitochondrial abundance after RT. Increased mitochondrial volume associated with increased mitochondrial respiration and insulin sensitivity following HIIT (P < 0.05). Increased mitochondrial perimeter associated with increased mitochondrial respiration, insulin sensitivity, and V̇o2peak following HIIT (P < 0.05). No such relationships were observed following CT or RT. OPA1, a regulator of fusion, was increased following HIIT (P < 0.05), whereas FIS1, a regulator of fission, was decreased following HIIT and CT (P < 0.05). HIIT also increased the ratio of OPA1/FIS1 (P < 0.01), indicative of the balance between fission and fusion, which positively correlated with improvements in respiration, insulin sensitivity, and V̇o2peak (P < 0.05). In conclusion, HIIT induces a larger, more fused mitochondrial tubular network. Changes indicative of increased fusion following HIIT associate with improvements in mitochondrial respiration, insulin sensitivity, and V̇o2peak supporting the idea that enhanced mitochondrial fusion accompanies notable health benefits of HIIT.NEW & NOTEWORTHY We assessed the effects of 12 wk of supervised high-intensity interval training (HIIT), resistance training, and combined training (CT) on skeletal muscle mitochondrial abundance and markers of fission and fusion. HIIT increased mitochondrial area and size and promoted protein changes indicative of increased mitochondrial fusion, whereas lessor effects were observed after CT and no changes were observed after RT. Furthermore, increased mitochondrial area and size after HIIT associated with improved mitochondrial respiration, cardiorespiratory fitness, and insulin sensitivity.
Collapse
Affiliation(s)
- Gregory N Ruegsegger
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, United States
- Department of Health and Human Performance, University of Wisconsin-River Falls, River Falls, Wisconsin, United States
| | - Mark W Pataky
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, United States
| | - Suvyaktha Simha
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, United States
| | - Matthew M Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon, United States
| | - Katherine A Klaus
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, United States
| | - K Sreekumaran Nair
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
94
|
Hinton A, Katti P, Christensen TA, Mungai M, Shao J, Zhang L, Trushin S, Alghanem A, Jaspersen A, Geroux RE, Neikirk K, Biete M, Lopez EG, Shao B, Vue Z, Vang L, Beasley HK, Marshall AG, Stephens D, Damo S, Ponce J, Bleck CKE, Hicsasmaz I, Murray SA, Edmonds RAC, Dajles A, Koo YD, Bacevac S, Salisbury JL, Pereira RO, Glancy B, Trushina E, Abel ED. A Comprehensive Approach to Sample Preparation for Electron Microscopy and the Assessment of Mitochondrial Morphology in Tissue and Cultured Cells. Adv Biol (Weinh) 2023; 7:e2200202. [PMID: 37140138 PMCID: PMC10615857 DOI: 10.1002/adbi.202200202] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 03/24/2023] [Indexed: 05/05/2023]
Abstract
Mitochondria respond to metabolic demands of the cell and to incremental damage, in part, through dynamic structural changes that include fission (fragmentation), fusion (merging of distinct mitochondria), autophagic degradation (mitophagy), and biogenic interactions with the endoplasmic reticulum (ER). High resolution study of mitochondrial structural and functional relationships requires rapid preservation of specimens to reduce technical artifacts coupled with quantitative assessment of mitochondrial architecture. A practical approach for assessing mitochondrial fine structure using two dimensional and three dimensional high-resolution electron microscopy is presented, and a systematic approach to measure mitochondrial architecture, including volume, length, hyperbranching, cristae morphology, and the number and extent of interaction with the ER is described. These methods are used to assess mitochondrial architecture in cells and tissue with high energy demand, including skeletal muscle cells, mouse brain tissue, and Drosophila muscles. The accuracy of assessment is validated in cells and tissue with deletion of genes involved in mitochondrial dynamics.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
| | - Prasanna Katti
- National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Trace A Christensen
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Liang Zhang
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Sergey Trushin
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ahmad Alghanem
- Department of Internal Medicine, Division of Cardiology, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO, 63130, USA
- Eastern Region, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh 11481, Al Hasa, Saudi Arabia
| | - Adam Jaspersen
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Rachel E Geroux
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kit Neikirk
- College of Natural and Health Sciences, University of Hawaii at Hilo, 200 West Kawili St, Hilo, HI, 96720, USA
| | - Michelle Biete
- College of Natural and Health Sciences, University of Hawaii at Hilo, 200 West Kawili St, Hilo, HI, 96720, USA
| | - Edgar Garza Lopez
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, 37208, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN, 37235, USA
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
| | - Steven Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
| | - Jessica Ponce
- School of Medicine, University of Utah, 30 N 1900 E, Salt Lake City, UT, 84132, USA
| | - Christopher K E Bleck
- National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Innes Hicsasmaz
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
| | - Sandra A Murray
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Ranthony A C Edmonds
- Department of Mathematics, Ohio State University, 281 W Lane Ave, Columbus, OH, 43210, USA
| | - Andres Dajles
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
| | - Jeffrey L Salisbury
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Renata O Pereira
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
| | - Brian Glancy
- National Heart, Lung, and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - E Dale Abel
- Department of Internal Medicine, University of Iowa - Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, 169 Newton Rd, Iowa City, IA, 52242, USA
- Department of Medicine, UCLA, 757 Westwood Plaza, Suite 7236, David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| |
Collapse
|
95
|
Elsaid HOA, Rivedal M, Skandalou E, Svarstad E, Tøndel C, Birkeland E, Eikrem Ø, Babickova J, Marti HP, Furriol J. Proteomic analysis unveils Gb3-independent alterations and mitochondrial dysfunction in a gla -/- zebrafish model of Fabry disease. J Transl Med 2023; 21:591. [PMID: 37670295 PMCID: PMC10478213 DOI: 10.1186/s12967-023-04475-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/28/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Fabry disease (FD) is a rare lysosomal storage disorder caused by mutations in the GLA gene, resulting in reduced or lack of α-galactosidase A activity. This results in the accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids in lysosomes causing cellular impairment and organ failures. While current therapies focus on reversing Gb3 accumulation, they do not address the altered cellular signaling in FD. Therefore, this study aims to explore Gb3-independent mechanisms of kidney damage in Fabry disease and identify potential biomarkers. METHODS To investigate these mechanisms, we utilized a zebrafish (ZF) gla-/- mutant (MU) model. ZF naturally lack A4GALT gene and, therefore, cannot synthesize Gb3. We obtained kidney samples from both wild-type (WT) (n = 8) and MU (n = 8) ZF and conducted proteome profiling using untargeted mass spectrometry. Additionally, we examined mitochondria morphology and cristae morphology using electron microscopy. To assess oxidative stress, we measured total antioxidant activity. Finally, immunohistochemistry was conducted on kidney samples to validate specific proteins. RESULTS Our proteomics analysis of renal tissues from zebrafish revealed downregulation of lysosome and mitochondrial-related proteins in gla-/- MU renal tissues, while energy-related pathways including carbon, glycolysis, and galactose metabolisms were disturbed. Moreover, we observed abnormal mitochondrial shape, disrupted cristae morphology, altered mitochondrial volume and lower antioxidant activity in gla-/- MU ZF. CONCLUSIONS These results suggest that the alterations observed at the proteome and mitochondrial level closely resemble well-known GLA mutation-related alterations in humans. Importantly, they also unveil novel Gb3-independent pathogenic mechanisms in Fabry disease. Understanding these mechanisms could potentially lead to the development of innovative drug screening approaches. Furthermore, the findings pave the way for identifying new clinical targets, offering new avenues for therapeutic interventions in Fabry disease. The zebrafish gla-/- mutant model proves valuable in elucidating these mechanisms and may contribute significantly to advancing our knowledge of this disorder.
Collapse
Affiliation(s)
- Hassan Osman Alhassan Elsaid
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Mariell Rivedal
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Eleni Skandalou
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Einar Svarstad
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Camilla Tøndel
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Even Birkeland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Øystein Eikrem
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Janka Babickova
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jessica Furriol
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
- Department of Medicine, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
96
|
Silva-Palacios A, Arana-Hidalgo D, Colín-Val Z, Castrejón-Téllez V, Soria-Castro E, Pedraza-Chaverrí J, López-Marure R, Zazueta C. Sulforaphane modifies mitochondrial-endoplasmic reticulum associations through reductive stress in cardiomyocytes. Chem Biol Interact 2023; 382:110616. [PMID: 37385402 DOI: 10.1016/j.cbi.2023.110616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/01/2023]
Abstract
Mitochondria-endoplasmic reticulum (ER) communication relies on platforms formed at the ER membrane with the mitochondrial outer membrane contact sites (MERCs). MERCs are involved in several processes including the unfolded protein response (UPR) and calcium (Ca2+) signaling. Therefore, as alterations in MERCs greatly impact cellular metabolism, pharmacological interventions to preserve productive mitochondrial-ER communication have been explored to maintain cellular homeostasis. In this regard, extensive information has documented the beneficial and potential effects of sulforaphane (SFN) in different pathological conditions; however, controversy has arisen regarding the effect of this compound on mitochondria-ER interaction. Therefore, in this study, we investigated whether SFN could induce changes in MERCs under normal culture conditions without damaging stimuli. Our results indicate that non-cytotoxic concentration of 2.5 μM SFN increased ER stress in cardiomyocytes in conjunction with a reductive stress environment, that diminishes ER-mitochondria association. Additionally, reductive stress promotes Ca2+ accumulation in the ER of cardiomyocytes. These data show an unexpected effect of SFN on cardiomyocytes grown under standard culture conditions, promoted by the cellular redox unbalance. Therefore, it is necessary to rationalize the use of compounds with antioxidant properties to avoid triggering cellular side effects.
Collapse
Affiliation(s)
- Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico.
| | - Dana Arana-Hidalgo
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Zaira Colín-Val
- Department of Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | | | - Elizabeth Soria-Castro
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - José Pedraza-Chaverrí
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Rebeca López-Marure
- Department of Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
97
|
Buchanan E, Mahony C, Bam S, Jaffer M, Macleod S, Mangali A, van der Watt M, de Wet S, Theart R, Jacobs C, Loos B, O'Ryan C. Propionic acid induces alterations in mitochondrial morphology and dynamics in SH-SY5Y cells. Sci Rep 2023; 13:13248. [PMID: 37582965 PMCID: PMC10427685 DOI: 10.1038/s41598-023-40130-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
Propionic acid (PPA) is used to study the role of mitochondrial dysfunction in neurodevelopmental conditions like autism spectrum disorders. PPA is known to disrupt mitochondrial biogenesis, metabolism, and turnover. However, the effect of PPA on mitochondrial dynamics, fission, and fusion remains challenging to study due to the complex temporal nature of these mechanisms. Here, we use complementary quantitative visualization techniques to examine how PPA influences mitochondrial ultrastructure, morphology, and dynamics in neuronal-like SH-SY5Y cells. PPA (5 mM) induced a significant decrease in mitochondrial area (p < 0.01), Feret's diameter and perimeter (p < 0.05), and in area2 (p < 0.01). Mitochondrial event localiser analysis demonstrated a significant increase in fission and fusion events (p < 0.05) that preserved mitochondrial network integrity under stress. Moreover, mRNA expression of cMYC (p < 0.0001), NRF1 (p < 0.01), TFAM (p < 0.05), STOML2 (p < 0.0001), and OPA1 (p < 0.01) was significantly decreased. This illustrates a remodeling of mitochondrial morphology, biogenesis, and dynamics to preserve function under stress. Our data provide new insights into the influence of PPA on mitochondrial dynamics and highlight the utility of visualization techniques to study the complex regulatory mechanisms involved in the mitochondrial stress response.
Collapse
Affiliation(s)
- Erin Buchanan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Caitlyn Mahony
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Sophia Bam
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Mohamed Jaffer
- Electron Microscope Unit, University of Cape Town, Cape Town, 7700, South Africa
| | - Sarah Macleod
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Asandile Mangali
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Mignon van der Watt
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Rensu Theart
- Department of Electrical and Electronic Engineering, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Caron Jacobs
- Department of Pathology, Wellcome Centre for Infectious Diseases Research in Africa and IDM Microscopy Platform, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Colleen O'Ryan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, 7700, South Africa.
| |
Collapse
|
98
|
Crabtree A, Neikirk K, Marshall AG, Vang L, Whiteside AJ, Williams Q, Altamura CT, Owens TC, Stephens D, Shao B, Koh A, Killion M, Lopez EG, Lam J, Rodriguez B, Mungai M, Stanley J, Dean ED, Koh HJ, Gaddy JA, Scudese E, Sweetwyne M, Davis J, Zaganjor E, Murray SA, Katti P, Damo SM, Vue Z, Hinton A. Defining Mitochondrial Cristae Morphology Changes Induced by Aging in Brown Adipose Tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.12.540609. [PMID: 37577723 PMCID: PMC10418056 DOI: 10.1101/2023.05.12.540609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Mitochondria are required for energy production and even give brown adipose tissue (BAT) its characteristic color due to their high iron content and abundance. The physiological function and bioenergetic capacity of mitochondria are connected to the structure, folding, and organization of its inner-membrane cristae. During the aging process, mitochondrial dysfunction is observed, and the regulatory balance of mitochondrial dynamics is often disrupted, leading to increased mitochondrial fragmentation in aging cells. Therefore, we hypothesized that significant morphological changes in BAT mitochondria and cristae would be present with aging. We developed a quantitative three-dimensional (3D) electron microscopy approach to map cristae network organization in mouse BAT to test this hypothesis. Using this methodology, we investigated the 3D morphology of mitochondrial cristae in adult (3-month) and aged (2-year) murine BAT tissue via serial block face-scanning electron microscopy (SBF-SEM) and 3D reconstruction software for manual segmentation, analysis, and quantification. Upon investigation, we found increases in mitochondrial volume, surface area, and complexity and decreased sphericity in aged BAT, alongside significant decreases in cristae volume, area, perimeter, and score. Overall, these data define the nature of the mitochondrial structure in murine BAT across aging. Abstract Figure
Collapse
Affiliation(s)
- Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron J Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Qiana Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Christopher T Altamura
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Trinity Celeste Owens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mason Killion
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Ben Rodriguez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jade Stanley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - E Danielle Dean
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, TN, 37232, USA
| | - Ho-Jin Koh
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jennifer A Gaddy
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil; Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Mariya Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience, Pharmacology, Meharry Medical College, Nashville, TN 37208 USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Sandra A Murray
- Department of Cell Biology, University of Pittsburgh; Pittsburg h, PA, 15261 USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
99
|
Marinho D, Ferreira IL, Lorenzoni R, Cardoso SM, Santana I, Rego AC. Reduction of class I histone deacetylases ameliorates ER-mitochondria cross-talk in Alzheimer's disease. Aging Cell 2023; 22:e13895. [PMID: 37358017 PMCID: PMC10410063 DOI: 10.1111/acel.13895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023] Open
Abstract
Several molecular mechanisms have been described in Alzheimer's disease (AD), including repressed gene transcription and mitochondrial and endoplasmic reticulum (ER) dysfunction. In this study, we evaluate the potential efficacy of transcriptional modifications exerted by inhibition or knockdown of class I histone deacetylases (HDACs) in ameliorating ER-mitochondria cross-talk in AD models. Data show increased HDAC3 protein levels and decreased acetyl-H3 in AD human cortex, and increased HDAC2-3 in MCI peripheral human cells, HT22 mouse hippocampal cells exposed to Aβ1-42 oligomers (AβO) and APP/PS1 mouse hippocampus. Tacedinaline (Tac, a selective class I HDAC inhibitor) counteracted the increase in ER-Ca2+ retention and mitochondrial Ca2+ accumulation, mitochondrial depolarization and impaired ER-mitochondria cross-talk, as observed in 3xTg-AD mouse hippocampal neurons and AβO-exposed HT22 cells. We further demonstrated diminished mRNA levels of proteins involved in mitochondrial-associated ER membranes (MAM) in cells exposed to AβO upon Tac treatment, along with reduction in ER-mitochondria contacts (MERCS) length. HDAC2 silencing reduced ER-mitochondria Ca2+ transfer and mitochondrial Ca2+ retention, while knockdown of HDAC3 decreased ER-Ca2+ accumulation in AβO-treated cells. APP/PS1 mice treated with Tac (30 mg/kg/day) also showed regulation of mRNA levels of MAM-related proteins, and reduced Aβ levels. These data demonstrate that Tac normalizes Ca2+ signaling between mitochondria and ER, involving the tethering between the two organelles in AD hippocampal neural cells. Tac-mediated AD amelioration occurs through the regulation of protein expression at MAM, as observed in AD cells and animal models. Data support transcriptional regulation of ER-mitochondria communication as a promising target for innovative therapeutics in AD.
Collapse
Affiliation(s)
- Daniela Marinho
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Ildete Luísa Ferreira
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Ricardo Lorenzoni
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Sandra M. Cardoso
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| | - Isabel Santana
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
- Neurology DepartmentCHUC‐Centro Hospitalar e Universitário de CoimbraCoimbraPortugal
| | - A. Cristina Rego
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
100
|
Jang HJ, Lee YH, Dao T, Jo Y, Khim KW, Eom HJ, Lee JE, Song YJ, Choi SS, Park K, Ji H, Chae YC, Myung K, Kim H, Ryu D, Park NH, Park SH, Choi JH. Thrap3 promotes nonalcoholic fatty liver disease by suppressing AMPK-mediated autophagy. Exp Mol Med 2023; 55:1720-1733. [PMID: 37524868 PMCID: PMC10474030 DOI: 10.1038/s12276-023-01047-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/14/2023] [Accepted: 04/30/2023] [Indexed: 08/02/2023] Open
Abstract
Autophagy functions in cellular quality control and metabolic regulation. Dysregulation of autophagy is one of the major pathogenic factors contributing to the progression of nonalcoholic fatty liver disease (NAFLD). Autophagy is involved in the breakdown of intracellular lipids and the maintenance of healthy mitochondria in NAFLD. However, the mechanisms underlying autophagy dysregulation in NAFLD remain unclear. Here, we demonstrate that the hepatic expression level of Thrap3 was significantly increased in NAFLD conditions. Liver-specific Thrap3 knockout improved lipid accumulation and metabolic properties in a high-fat diet (HFD)-induced NAFLD model. Furthermore, Thrap3 deficiency enhanced autophagy and mitochondrial function. Interestingly, Thrap3 knockout increased the cytosolic translocation of AMPK from the nucleus and enhanced its activation through physical interaction. The translocation of AMPK was regulated by direct binding with AMPK and the C-terminal domain of Thrap3. Our results indicate a role for Thrap3 in NAFLD progression and suggest that Thrap3 is a potential target for NAFLD treatment.
Collapse
Affiliation(s)
- Hyun-Jun Jang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Yo Han Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Yunju Jo
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Keon Woo Khim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hye-Jin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ju Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yi Jin Song
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Sun Sil Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kieun Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Haneul Ji
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Republic of Korea
| | - Hongtae Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Neung Hwa Park
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, 44033, Republic of Korea.
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Jang Hyun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|