1
|
Lejri I, Grimm A, Trempat P, Boujedaini N, Eckert A. Gelsemium low doses protect against serum deprivation-induced stress on mitochondria in neuronal cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118714. [PMID: 39181289 DOI: 10.1016/j.jep.2024.118714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gelsemium dynamized dilutions (GDD) are known as a remedy for a wide range of behavioral and psychological symptoms of depression and anxiety at ultra-low doses, yet the underlying mechanisms of the mode of action of G. sempervirens itself are not well understood. AIM OF THE STUDY The present study was designed to examine the neuroprotective effects of Gelsemium preparations in counteracting stress-related mitochondrial dysfunctions in neuronal cells. MATERIALS AND METHODS We started by studying how serum deprivation affects the mitochondrial functions of human neuroblastoma (SH-SY5Y) cells. Next, we looked into the potential of various Gelsemium dilutions to improve cell survival and ATP levels. After identifying the most effective dilutions, 3C and 5C, we tested their ability to protect SH-SY5Y cells from stress-induced mitochondrial deficits. We measured total and mitochondrial superoxide anion radicals using fluorescent dyes dihydroethidium (DHE) and the red mitochondrial superoxide indicator (MitoSOX). Additionally, we assessed total nitric oxide levels with 4,5-diaminofluorescein diacetate (DAF-2DA), examined the redox state using pRA305 cells stably transfected with a plasmid encoding a redox-sensitive green fluorescent protein, and analyzed mitochondrial network morphology using an automated high-content analysis device, Cytation3. Furthermore, we investigated bioenergetics by measuring ATP production with a bioluminescence assay (ViaLighTM HT) and evaluated mitochondrial respiration (OCR) and glycolysis (ECAR) using the Seahorse Bioscience XF24 Analyzer. Finally, we determined cell survival using an MTT reduction assay. RESULTS Our research indicates that Gelsemium dilutions (3C and 5C) exhibited neuroprotective effects by: - Normalizing total and mitochondrial superoxide anion radicals and total nitric oxide levels. - Regulating the mitochondrial redox environment and mitochondrial networks morphology. - Increasing ATP generation as well as OCR and ECAR levels, thereby reducing the viability loss induced by serum withdrawal stress. CONCLUSIONS These findings highlight that dynamized Gelsemium preparations may have neuroprotective effects against stress-induced cellular changes in the brain by regulating mitochondrial functions, essential for the survival, plasticity, and function of neurons in depression.
Collapse
Affiliation(s)
- Imane Lejri
- Research Cluster Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland.
| | - Amandine Grimm
- Research Cluster Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland.
| | | | | | - Anne Eckert
- Research Cluster Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland.
| |
Collapse
|
2
|
Lejri I, Cader Z, Grimm A, Eckert A. Human iPSCs from Aged Donors Retain Their Mitochondrial Aging Signature. Int J Mol Sci 2024; 25:11199. [PMID: 39456998 PMCID: PMC11508692 DOI: 10.3390/ijms252011199] [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: 09/25/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Aging represents the leading risk factor for developing neurodegenerative disorders. One of the nine hallmarks of aging is mitochondrial dysfunction. Age-related mitochondrial alterations have been shown to affect mitochondrial energy metabolism, reduction-oxidation homeostasis, and mitochondrial dynamics. Previous reports have shown that induced pluripotent stem cells (iPSCs) from aged donors do not keep the aging signature at the transcriptomic level. However, not all aspects of aging have been investigated, and especially not the mitochondria-related aging signature. Therefore, the present study compared the mitochondrial function in iPSCs from healthy aged donors compared to those of young donors. We addressed whether aged iPSCs may be used as drug-screening models of "aging in a dish" to identify therapies alleviating mitochondria aging. Compared to iPSCs from young donors, we demonstrate that iPSCs from aged donors show impaired mitochondrial bioenergetics and exhibit a rise in reactive oxygen species generation. Furthermore, aged iPSCs present a lower mitochondrial mass and alterations in the morphology of the mitochondrial network when compared to iPSCs from young donors. This study provides the first evidence that the aging phenotype is present at the mitochondrial level in iPSCs from aged donors, ranging from bioenergetics to mitochondrial network morphology. This model might be used to screen mitochondria-targeting drugs to promote healthy aging at the mitochondrial level.
Collapse
Affiliation(s)
- Imane Lejri
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| | - Zameel Cader
- Translational Molecular Neuroscience Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK;
| | - Amandine Grimm
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Department of Biomedicine, University of Basel, 4055 Basel, Switzerland
| | - Anne Eckert
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland; (I.L.); (A.G.)
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| |
Collapse
|
3
|
Tseng WW, Chu CH, Lee YJ, Zhao S, Chang C, Ho YP, Wei AC. Metabolic regulation of mitochondrial morphologies in pancreatic beta cells: coupling of bioenergetics and mitochondrial dynamics. Commun Biol 2024; 7:1267. [PMID: 39369076 PMCID: PMC11455970 DOI: 10.1038/s42003-024-06955-3] [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: 12/02/2021] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
Cellular bioenergetics and mitochondrial dynamics are crucial for the secretion of insulin by pancreatic beta cells in response to elevated levels of blood glucose. To elucidate the interactions between energy production and mitochondrial fission/fusion dynamics, we combine live-cell mitochondria imaging with biophysical-based modeling and graph-based network analysis. The aim is to determine the mechanism that regulates mitochondrial morphology and balances metabolic demands in pancreatic beta cells. A minimalistic differential equation-based model for beta cells is constructed that includes glycolysis, oxidative phosphorylation, calcium dynamics, and fission/fusion dynamics, with ATP synthase flux and proton leak flux as main regulators of mitochondrial dynamics. The model shows that mitochondrial fission occurs in response to hyperglycemia, starvation, ATP synthase inhibition, uncoupling, and diabetic conditions, in which the rate of proton leakage exceeds the rate of mitochondrial ATP synthesis. Under these metabolic challenges, the propensities of tip-to-tip fusion events simulated from the microscopy images of the mitochondrial networks are lower than those in the control group and prevent the formation of mitochondrial networks. The study provides a quantitative framework that couples bioenergetic regulation with mitochondrial dynamics, offering insights into how mitochondria adapt to metabolic challenges.
Collapse
Affiliation(s)
- Wen-Wei Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsiang Chu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yi-Ju Lee
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Shirui Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Centre for Novel Biomaterials, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of the CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- The Ministry of Education Key Laboratory of Regeneration Medicine, Shatin, New Territories, Hong Kong SAR, China
| | - Chen Chang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Centre for Novel Biomaterials, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of the CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- The Ministry of Education Key Laboratory of Regeneration Medicine, Shatin, New Territories, Hong Kong SAR, China
| | - An-Chi Wei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
4
|
Delgado-Waldo I, Dokudovskaya S, Loissell-Baltazar YA, Pérez-Arteaga E, Coronel-Hernández J, Martínez-Vázquez M, Pérez-Yépez EA, Lopez-Saavedra A, Jacobo-Herrera N, Pérez Plasencia C. Laherradurin Inhibits Colorectal Cancer Cell Growth by Induction of Mitochondrial Dysfunction and Autophagy Induction. Cells 2024; 13:1649. [PMID: 39404412 PMCID: PMC11475353 DOI: 10.3390/cells13191649] [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: 09/09/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
LAH, an acetogenin from the Annonaceae family, has demonstrated antitumor activity in several cancer cell lines and in vivo models, where it reduced the tumor size and induced programmed cell death. We focused on the effects of LAH on mitochondrial dynamics, mTOR signaling, autophagy, and apoptosis in colorectal cancer (CRC) cells to explore its anticancer potential. METHODS CRC cells were treated with LAH, and its effects on mitochondrial respiration and glycolysis were measured using Seahorse XF technology. The changes in mitochondrial dynamics were observed through fluorescent imaging, while Western blot analysis was used to examine key autophagy and apoptosis markers. RESULTS LAH significantly inhibited mitochondrial complex I activity, inducing ATP depletion and a compensatory increase in glycolysis. This disruption caused mitochondrial fragmentation, a trigger for autophagy, as shown by increased LC3-II expression and mTOR suppression. Apoptosis was also confirmed through the cleavage of caspase-3, contributing to reduced cancer cell viability. CONCLUSIONS LAH's anticancer effects in CRC cells are driven by its disruption of mitochondrial function, triggering both autophagy and apoptosis. These findings highlight its potential as a therapeutic compound for further exploration in cancer treatment.
Collapse
Affiliation(s)
- Izamary Delgado-Waldo
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico
| | - Svetlana Dokudovskaya
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Yahir A. Loissell-Baltazar
- CNRS UMR9018, Institut Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; (S.D.); (Y.A.L.-B.)
| | - Eduardo Pérez-Arteaga
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Jossimar Coronel-Hernández
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Mariano Martínez-Vázquez
- Instituto de Química, Universidad Nacional Autónoma de México, C. Exterior, C. Universitaria, Coyoacán, Ciudad de México 04510, Mexico;
| | - Eloy Andrés Pérez-Yépez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
| | - Alejandro Lopez-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA), Instituto Nacional de Cancerología, San Fernando 22. Col. Sección XVI, Tlalpan, Ciudad de México 14080, Mexico;
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey Ciudad de Mexico, C. Puente #222, Coapa, Arboledas del Sur, Tlalpan, Ciudad de Mexico 14380, Mexico
| | - Nadia Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Av. Vasco de Quiroga 15, Col. Belisario Domínguez Sección XVI, Tlalpan, Ciudad de México 14080, Mexico; (I.D.-W.); (E.P.-A.)
| | - Carlos Pérez Plasencia
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Instituto Nacional Nacional de Cancerología, Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico; (J.C.-H.); (E.A.P.-Y.)
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México 54090, Mexico
| |
Collapse
|
5
|
Javed Z, Shin DH, Pan W, White SR, Elhaw AT, Kim YS, Kamlapurkar S, Cheng YY, Benson JC, Abdelnaby AE, Phaëton R, Wang HG, Yang S, Sullivan MLG, St Croix CM, Watkins SC, Mullett SJ, Gelhaus SL, Lee N, Coffman LG, Aird KM, Trebak M, Mythreye K, Walter V, Hempel N. Drp1 splice variants regulate ovarian cancer mitochondrial dynamics and tumor progression. EMBO Rep 2024; 25:4281-4310. [PMID: 39191946 PMCID: PMC11467262 DOI: 10.1038/s44319-024-00232-4] [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: 02/26/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Aberrant mitochondrial fission/fusion dynamics are frequently associated with pathologies, including cancer. We show that alternative splice variants of the fission protein Drp1 (DNM1L) contribute to the complexity of mitochondrial fission/fusion regulation in tumor cells. High tumor expression of the Drp1 alternative splice variant lacking exon 16 relative to other transcripts is associated with poor outcome in ovarian cancer patients. Lack of exon 16 results in Drp1 localization to microtubules and decreased association with mitochondrial fission sites, culminating in fused mitochondrial networks, enhanced respiration, changes in metabolism, and enhanced pro-tumorigenic phenotypes in vitro and in vivo. These effects are inhibited by siRNAs designed to specifically target 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 pathophysiological importance of Drp1 alternative splicing, highlight the divergent functions and consequences of changing the relative expression of Drp1 splice variants in tumor cells, and strongly warrant consideration of alternative splicing in future studies focused on Drp1.
Collapse
Affiliation(s)
- Zaineb Javed
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, 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
- School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Weihua Pan
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sierra R White
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amal Taher Elhaw
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Yeon Soo Kim
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shriya Kamlapurkar
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ya-Yun Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rébécca Phaëton
- Department of Obstetrics & Gynecology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- GlaxoSmithKline, Collegeville, 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
| | - Mara L G Sullivan
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Claudette M St Croix
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Simon C Watkins
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Health Sciences Mass Spectrometry Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stacy L Gelhaus
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Health Sciences Mass Spectrometry Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nam Lee
- 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, Pittsburgh, PA, USA
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katherine M Aird
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, 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, Pittsburgh, PA, USA
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, 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, Pittsburgh, PA, USA.
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Qu J, Tian L, Zhang M, Sun B, Chen L. SGLT2 inhibitor canagliflozin reduces visceral adipose tissue in db/db mice by modulating AMPK/KLF4 signaling and regulating mitochondrial dynamics to induce browning. Mol Cell Endocrinol 2024; 592:112320. [PMID: 38964727 DOI: 10.1016/j.mce.2024.112320] [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: 03/25/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Obesity is characterized by excessive accumulation of adipose tissue (mainly visceral). The morphology and function of mitochondria are crucial for regulating adipose browning and weight loss. Research suggests that the SGLT2 inhibitor canagliflozin may induce weight loss through an unknown mechanism, particularly targeting visceral adipose tissue. While Krueppel-Like Factor 4 (KLF4) is known to be essential for energy metabolism and mitochondrial function, its specific impact on visceral adipose tissue remains unclear. We administered canagliflozin to db/db mice for 8 weeks, or exposed adipocytes to canagliflozin for 24 h. The expression levels of browning markers, mitochondrial dynamics, and KLF4 were assessed. Then we validated the function of KLF4 through overexpression in vivo and in vitro. Adenosine monophosphate-activated protein kinase (AMPK) agonists, inhibitors, and KLF4 si-RNA were employed to elucidate the relationship between AMPK and KLF4. The findings demonstrated that canagliflozin significantly decreased body weight in db/db mice and augmented cold-induced thermogenesis. Additionally, canagliflozin increased the expression of mitochondrial fusion-related factors while reducing the levels of fission markers in epididymal white adipose tissue. These consistent findings were mirrored in canagliflozin-treated adipocytes. Similarly, overexpression of KLF4 in both adipocytes and db/db mice yielded comparable results. In all, canagliflozin mitigates obesity in db/db mice by promoting the brown visceral adipocyte phenotype through enhanced mitochondrial fusion via AMPK/KLF4 signaling.
Collapse
Affiliation(s)
- Jingru Qu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Lei Tian
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Man Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| |
Collapse
|
7
|
Suh J, Lee YS. The multifaceted roles of mitochondria in osteoblasts: from energy production to mitochondrial-derived vesicle secretion. J Bone Miner Res 2024; 39:1205-1214. [PMID: 38907370 PMCID: PMC11371665 DOI: 10.1093/jbmr/zjae088] [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: 01/12/2024] [Revised: 05/03/2024] [Indexed: 06/24/2024]
Abstract
Mitochondria in osteoblasts have been demonstrated to play multiple crucial functions in bone formation from intracellular adenosine triphosphate production to extracellular secretion of mitochondrial components. The present review explores the current knowledge about mitochondrial biology in osteoblasts, including mitochondrial biogenesis, bioenergetics, oxidative stress generation, and dynamic changes in morphology. Special attention is given to recent findings, including mitochondrial donut formation in osteoblasts, which actively generates mitochondrial-derived vesicles (MDVs), followed by extracellular secretion of small mitochondria and MDVs. We also discuss the therapeutic effects of targeting osteoblast mitochondria, highlighting their potential applications in improving bone health.
Collapse
Affiliation(s)
- Joonho Suh
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun-Sil Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
8
|
Sword J, Fomitcheva IV, Kirov SA. Spreading depolarization causes reversible neuronal mitochondria fragmentation and swelling in healthy, normally perfused neocortex. J Cereb Blood Flow Metab 2024:271678X241257887. [PMID: 39053498 PMCID: PMC11574936 DOI: 10.1177/0271678x241257887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/28/2024] [Accepted: 05/12/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial function is tightly linked to morphology, and fragmentation of dendritic mitochondria during noxious conditions suggests loss of function. In the normoxic cortex, spreading depolarization (SD) is a phenomenon underlying migraine aura. It is unknown whether mitochondria structure is affected by normoxic SD. In vivo two-photon imaging followed by quantitative serial section electron microscopy (ssEM) was used to monitor dendritic mitochondria in the normoxic cortex of urethane-anesthetized mature male and female mice during and after SD initiated by focal KCl microinjection. Structural dynamics of dendrites and their mitochondria were visualized by transfecting excitatory, glutamatergic neurons of the somatosensory cortex with bicistronic AAV, which induced tdTomoto labeling in neuronal cytoplasm and mitochondria labeling with roGFP. Normoxic SD triggered rapidly reversible fragmentation of dendritic mitochondria alongside dendritic beading; however, mitochondria took significantly longer to recover. Several rounds of SD resulted in transient mitochondrial fragmentation and dendritic beading without accumulating injury, as both recovered. SsEM corroborated normoxic SD-elicited dendritic and mitochondrial swelling and transformation of the filamentous mitochondrial network into shorter, swollen tubular, and globular structures. Our results revealed normoxic SD-induced disruption of the dendritic mitochondrial structure that might impact mitochondrial bioenergetics during migraine with aura.
Collapse
Affiliation(s)
- Jeremy Sword
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ioulia V Fomitcheva
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Sergei A Kirov
- Dept. of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Dept. of Neurosurgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
9
|
Hosen MHA, Whitworth DJ, Leusch FDL, Yuen N, Bengtson Nash SM. Bioenergetic Shifts in Humpback Whale Fibroblasts Upon Chemical Exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:12313-12319. [PMID: 38958666 DOI: 10.1021/acs.est.3c10595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Southern Hemisphere humpback whales accumulate persistent and toxic chemicals, which are transported to Antarctica through distant sources and in situ usage. The extreme seasonal migration-associated fast of humpback whales results in the remobilization of persistent and lipophilic environmental contaminants from liberated fat stores. Mitochondria play a key role in lipid metabolism, and any disruption to mitochondrial function is expected to influence whole-organism bioenergetics. It is therefore of interest to advance understanding of the impact of known contaminants of the Antarctic sea-ice ecosystem upon humpback whale cellular bioenergetics. Using cell line-based in vitro testing, this study employed the Seahorse Extracellular Flux Analyzer to study cellular metabolic activity in live humpback whale fibroblast cells. The assay, based on oxygen consumption rate, provides insights into the cause of cellular bioenergetic disruption. Immortalized skin fibroblasts were exposed to four priority environmental chemicals found in the Antarctic sea-ice ecosystem. Our findings reveal chemical-dependent functional alterations and varying bioenergetic profile responses. Chlorpyrifos was observed to decrease mitochondrial basal oxygen consumption; dieldrin increased basal oxygen consumption; trifluralin's impact was dose-specific, and endosulfan displayed no effect. Our results provide unique insights into environmental chemical mechanisms of action on cellular bioenergetics, generating much-needed taxa-specific chemical effect data in support of evidence-based conservation policy and management.
Collapse
Affiliation(s)
- Md Hafiz All Hosen
- Centre for Planetary Health and Food Security, School of Environment and Science, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Deanne J Whitworth
- The School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Frederic D L Leusch
- Australian Rivers Institute, School of Environment and Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Nicholas Yuen
- The School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Susan M Bengtson Nash
- Centre for Planetary Health and Food Security, School of Environment and Science, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
10
|
Ghani M, Szabó B, Alkhatibe M, Amsalu H, Zohar P, Janka EA, Mótyán JA, Tar K. Serine 39 in the GTP-binding domain of Drp1 is involved in shaping mitochondrial morphology. FEBS Open Bio 2024; 14:1147-1165. [PMID: 38760979 PMCID: PMC11216946 DOI: 10.1002/2211-5463.13820] [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: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
Continuous fusion and fission are critical for mitochondrial health. In this study, we further characterize the role played by dynamin-related protein 1 (Drp1) in mitochondrial fission. We show that a single amino acid change in Drp1 at position 39 from serine to alanine (S39A) within the GTP-binding (GTPase) domain results in a fused mitochondrial network in human SH-SY5Y neuroblastoma cells. Interestingly, the phosphorylation of Ser-616 and Ser-637 of Drp1 remains unaffected by the S39A mutation, and mitochondrial bioenergetic profile and cell viability in the S39A mutant were comparable to those observed in the control. This leads us to propose that the serine 39 residue of Drp1 plays a crucial role in mitochondrial distribution through its involvement in the GTPase activity. Furthermore, this amino acid mutation leads to structural anomalies in the mitochondrial network. Taken together, our results contribute to a better understanding of the function of the Drp1 protein.
Collapse
Affiliation(s)
- Marvi Ghani
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenHungary
| | - Bernadett Szabó
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
| | - Mahmoud Alkhatibe
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
| | - Hailemariam Amsalu
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
- Doctoral School of Molecular MedicineUniversity of DebrecenHungary
| | - Peleg Zohar
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
| | - Eszter Anna Janka
- Department of Dermatology, MTA Centre of Excellence, Faculty of MedicineUniversity of DebrecenHungary
- HUN‐REN‐UD Allergology Research GroupUniversity of DebrecenHungary
| | - János András Mótyán
- Department of Biochemistry and Molecular Biology, Faculty of MedicineUniversity of DebrecenHungary
| | - Krisztina Tar
- Department of Medical Chemistry, Faculty of MedicineUniversity of DebrecenHungary
| |
Collapse
|
11
|
Dong Z, Han W, Jiang P, Hao L, Fu X. Regulation of mitochondrial network architecture and function in mesenchymal stem cells by micropatterned surfaces. Regen Biomater 2024; 11:rbae052. [PMID: 38854681 PMCID: PMC11162196 DOI: 10.1093/rb/rbae052] [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: 01/08/2024] [Revised: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 06/11/2024] Open
Abstract
Mitochondrial network architecture, which is closely related to mitochondrial function, is mechanically sensitive and regulated by multiple stimuli. However, the effects of microtopographic cues on mitochondria remain poorly defined. Herein, polycaprolactone (PCL) surfaces were used as models to investigate how micropatterns regulate mitochondrial network architecture and function in rat adipose-derived stem cells (rASCs). It was found that large pit (LP)-induced rASCs to form larger and more complex mitochondrial networks. Consistently, the expression of key genes related to mitochondrial dynamics revealed that mitochondrial fusion (MFN1 and MFN2) and midzone fission (DRP1 and MFF) were increased in rASCs on LP. In contrast, the middle pit (MP)-enhanced mitochondrial biogenesis, as evidenced by the larger mitochondrial area and higher expression of PGC-1. Both LP and MP promoted ATP production in rASCs. It is likely that LP increased ATP levels through modulating mitochondrial network architecture while MP stimulated mitochondria biogenesis to do so. Our study clarified the regulation of micropatterned surfaces on mitochondria, highlighting the potential of LP and MP as a simple platform to stimulate mitochondria and the subsequent cellular function of MSCs.
Collapse
Affiliation(s)
- Zixuan Dong
- The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Weiju Han
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Panyu Jiang
- The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Lijing Hao
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xiaoling Fu
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
12
|
Hao Q, He X, Wang KN, Niu J, Meng F, Fu J, Zong C, Liu Z, Yu X. Long-Chain Fluorescent Probe for Straightforward and Nondestructive Staining Mitochondria in Fixed Cells and Tissues. Anal Chem 2024. [PMID: 38330436 DOI: 10.1021/acs.analchem.3c05660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Normally, small-molecule fluorescent probes dependent on the mitochondrial membrane potential (MMP) are invalid for fixed cells and tissues, which limits their clinical applications when the fixation of pathological specimens is imperative. Given that mitochondrial morphology is closely associated with disease, we developed a long-chain mitochondrial probe for fixed cells and tissues, DMPQ-12, by installing a C12-alkyl chain into the quinoline moiety. In fixed cells stained with DMPQ-12, filament mitochondria and folded cristae were observed with confocal and structural illumination microscopy, respectively. In titration test with three major phospholipids, DMPQ-12 exhibited a stronger binding force to mitochondria-exclusive cardiolipin, revealing its targeting mechanism. Moreover, mitochondrial morphological changes in the three lesion models were clearly visualized in fixed cells. Finally, by DMPQ-12, three kinds of mitochondria with different morphologies were observed in situ in fixed muscle tissues. This work breaks the conventional concept that organic fluorescent probes only stain mitochondria with normal membrane potentials and opens new avenues for comprehensive mitochondrial investigations in research and clinical settings.
Collapse
Affiliation(s)
- Qiuhua Hao
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Xiuquan He
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, P. R. China
| | - Kang-Nan Wang
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Jie Niu
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Fangfang Meng
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Jinyu Fu
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Chong Zong
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Zhiqiang Liu
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| | - Xiaoqiang Yu
- State Key Laboratory of Crystal Materials, Advanced Medical Research Institute, Shandong University, Jinan 250100, P. R. China
| |
Collapse
|
13
|
Vega-Vásquez T, Langgartner D, Wang JY, Reber SO, Picard M, Basualto-Alarcón C. Mitochondrial morphology in the mouse adrenal cortex: Influence of chronic psychosocial stress. Psychoneuroendocrinology 2024; 160:106683. [PMID: 38086320 PMCID: PMC10872515 DOI: 10.1016/j.psyneuen.2023.106683] [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: 02/10/2023] [Revised: 10/16/2023] [Accepted: 11/19/2023] [Indexed: 01/02/2024]
Abstract
Mitochondria within the adrenal cortex play a key role in synthesizing steroid hormones. The adrenal cortex is organized in three functionally specialized zones (glomerulosa, fasciculata, and reticularis) that produce different classes of steroid hormones in response to various stimuli, including psychosocial stress. Given that the functions and morphology of mitochondria are dynamically related and respond to stress, we applied transmission electron microscopy (TEM) to examine potential differences in mitochondrial morphology under basal and chronic psychosocial stress conditions. We used the chronic subordinate colony housing (CSC) paradigm, a murine model of chronic psychosocial stress. Our findings quantitatively define how mitochondrial morphology differs among each of the three adrenal cortex zones under basal conditions, and show that chronic psychosocial stress mainly affected mitochondria in the zona glomerulosa, shifting their morphology towards the more typical glucocorticoid-producing zona fasciculata mitochondrial phenotype. Analysis of adrenocortical lipid droplets that provide cholesterol for steroidogenesis showed that chronic psychosocial stress altered lipid droplet diameter, without affecting droplet number or inter-organellar mitochondria-lipid droplet interactions. Together, our findings support the hypothesis that each adrenal cortex layer is characterized by morphologically distinct mitochondria and that this adrenal zone-specific mitochondrial morphology is sensitive to environmental stimuli, including chronic psychosocial stressors. Further research is needed to define the role of these stress-induced changes in mitochondrial morphology, particularly in the zona glomerulosa, on stress resilience and related behaviors.
Collapse
Affiliation(s)
- Tamara Vega-Vásquez
- Laboratory of Cellular Physiology and Metabolism, Health Sciences Department, University of Aysén, Coyhaique, Chile
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Jennifer Y Wang
- School of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, Ulm, Germany
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA; Department of Neurology, H. Houston Merritt Center, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, USA; New York State Psychiatric Institute, New York, USA
| | - Carla Basualto-Alarcón
- Laboratory of Cellular Physiology and Metabolism, Health Sciences Department, University of Aysén, Coyhaique, Chile; Anatomy and Legal Medicine Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
14
|
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
|
15
|
Sword J, Fomitcheva IV, Kirov SA. Spreading depolarization causes reversible neuronal mitochondria fragmentation and swelling in healthy, normally perfused neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576364. [PMID: 38328069 PMCID: PMC10849532 DOI: 10.1101/2024.01.22.576364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Mitochondrial function is tightly linked to their morphology, and fragmentation of dendritic mitochondria during noxious conditions suggests loss of function. In the normoxic cortex, spreading depolarization (SD) is a phenomenon underlying migraine aura. It is unknown whether mitochondria structure is affected by normoxic SD. In vivo two-photon imaging followed by quantitative serial section electron microscopy (ssEM) was used to monitor dendritic mitochondria in the normoxic cortex of urethane-anesthetized mature male and female mice during and after SD initiated by focal KCl microinjection. Structural dynamics of dendrites and their mitochondria were visualized by transfecting excitatory, glutamatergic neurons of the somatosensory cortex with bicistronic AAV, which induced tdTomoto labeling in neuronal cytoplasm and mitochondria labeling with roGFP. Normoxic SD triggered a rapid fragmentation of dendritic mitochondria alongside dendritic beading, both reversible; however, mitochondria took significantly longer to recover. Several rounds of SD resulted in transient mitochondrial fragmentation and dendritic beading without accumulating injury, as both recovered. SsEM corroborated normoxic SD-elicited dendritic and mitochondrial swelling and transformation of the filamentous mitochondrial network into shorter, swollen tubular and globular structures. Our results revealed normoxic SD-induced disruption of the dendritic mitochondrial structure that might impact mitochondrial bioenergetics during migraine with aura.
Collapse
|
16
|
Hu L, Wang N, Bryant JD, Liu L, Xie L, West AP, Walsh AJ. Label-free spatially maintained measurements of metabolic phenotypes in cells. Front Bioeng Biotechnol 2023; 11:1293268. [PMID: 38090715 PMCID: PMC10715269 DOI: 10.3389/fbioe.2023.1293268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/14/2023] [Indexed: 02/01/2024] Open
Abstract
Metabolic reprogramming at a cellular level contributes to many diseases including cancer, yet few assays are capable of measuring metabolic pathway usage by individual cells within living samples. Here, autofluorescence lifetime imaging is combined with single-cell segmentation and machine-learning models to predict the metabolic pathway usage of cancer cells. The metabolic activities of MCF7 breast cancer cells and HepG2 liver cancer cells were controlled by growing the cells in culture media with specific substrates and metabolic inhibitors. Fluorescence lifetime images of two endogenous metabolic coenzymes, reduced nicotinamide adenine dinucleotide (NADH) and oxidized flavin adenine dinucleotide (FAD), were acquired by a multi-photon fluorescence lifetime microscope and analyzed at the cellular level. Quantitative changes of NADH and FAD lifetime components were observed for cells using glycolysis, oxidative phosphorylation, and glutaminolysis. Conventional machine learning models trained with the autofluorescence features classified cells as dependent on glycolytic or oxidative metabolism with 90%-92% accuracy. Furthermore, adapting convolutional neural networks to predict cancer cell metabolic perturbations from the autofluorescence lifetime images provided improved performance, 95% accuracy, over traditional models trained via extracted features. Additionally, the model trained with the lifetime features of cancer cells could be transferred to autofluorescence lifetime images of T cells, with a prediction that 80% of activated T cells were glycolytic, and 97% of quiescent T cells were oxidative. In summary, autofluorescence lifetime imaging combined with machine learning models can detect metabolic perturbations between glycolysis and oxidative metabolism of living samples at a cellular level, providing a label-free technology to study cellular metabolism and metabolic heterogeneity.
Collapse
Affiliation(s)
- Linghao Hu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Nianchao Wang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Joshua D. Bryant
- Microbial Pathogenesis and Immunology, Health Science Center, Texas A&M University, College Station, TX, United States
| | - Lin Liu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - A. Phillip West
- Microbial Pathogenesis and Immunology, Health Science Center, Texas A&M University, College Station, TX, United States
| | - Alex J. Walsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
17
|
Su É, Villard C, Manneville JB. Mitochondria: At the crossroads between mechanobiology and cell metabolism. Biol Cell 2023; 115:e2300010. [PMID: 37326132 DOI: 10.1111/boc.202300010] [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: 01/17/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Metabolism and mechanics are two key facets of structural and functional processes in cells, such as growth, proliferation, homeostasis and regeneration. Their reciprocal regulation has been increasingly acknowledged in recent years: external physical and mechanical cues entail metabolic changes, which in return regulate cell mechanosensing and mechanotransduction. Since mitochondria are pivotal regulators of metabolism, we review here the reciprocal links between mitochondrial morphodynamics, mechanics and metabolism. Mitochondria are highly dynamic organelles which sense and integrate mechanical, physical and metabolic cues to adapt their morphology, the organization of their network and their metabolic functions. While some of the links between mitochondrial morphodynamics, mechanics and metabolism are already well established, others are still poorly documented and open new fields of research. First, cell metabolism is known to correlate with mitochondrial morphodynamics. For instance, mitochondrial fission, fusion and cristae remodeling allow the cell to fine-tune its energy production through the contribution of mitochondrial oxidative phosphorylation and cytosolic glycolysis. Second, mechanical cues and alterations in mitochondrial mechanical properties reshape and reorganize the mitochondrial network. Mitochondrial membrane tension emerges as a decisive physical property which regulates mitochondrial morphodynamics. However, the converse link hypothesizing a contribution of morphodynamics to mitochondria mechanics and/or mechanosensitivity has not yet been demonstrated. Third, we highlight that mitochondrial mechanics and metabolism are reciprocally regulated, although little is known about the mechanical adaptation of mitochondria in response to metabolic cues. Deciphering the links between mitochondrial morphodynamics, mechanics and metabolism still presents significant technical and conceptual challenges but is crucial both for a better understanding of mechanobiology and for potential novel therapeutic approaches in diseases such as cancer.
Collapse
Affiliation(s)
- Émilie Su
- Laboratoire Matière et Systèmes Complexes (MSC), Université Paris Cité - CNRS, UMR 7057, Paris, France
- Laboratoire Interdisciplinaire des Énergies de Demain (LIED), Université Paris Cité - CNRS, UMR 8236, Paris, France
| | - Catherine Villard
- Laboratoire Interdisciplinaire des Énergies de Demain (LIED), Université Paris Cité - CNRS, UMR 8236, Paris, France
| | - Jean-Baptiste Manneville
- Laboratoire Matière et Systèmes Complexes (MSC), Université Paris Cité - CNRS, UMR 7057, Paris, France
| |
Collapse
|
18
|
Siemers KM, Klein AK, Baack ML. Mitochondrial Dysfunction in PCOS: Insights into Reproductive Organ Pathophysiology. Int J Mol Sci 2023; 24:13123. [PMID: 37685928 PMCID: PMC10488260 DOI: 10.3390/ijms241713123] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex, but relatively common endocrine disorder associated with chronic anovulation, hyperandrogenism, and micro-polycystic ovaries. In addition to reduced fertility, people with PCOS have a higher risk of obesity, insulin resistance, and metabolic disease, all comorbidities that are associated with mitochondrial dysfunction. This review summarizes human and animal data that report mitochondrial dysfunction and metabolic dysregulation in PCOS to better understand how mitochondria impact reproductive organ pathophysiology. This in-depth review considers all the elements regulating mitochondrial quantity and quality, from mitochondrial biogenesis under the transcriptional regulation of both the nuclear and mitochondrial genome to the ultrastructural and functional complexes that regulate cellular metabolism and reactive oxygen species production, as well as the dynamics that regulate subcellular interactions that are key to mitochondrial quality control. When any of these mitochondrial functions are disrupted, the energetic equilibrium within the cell changes, cell processes can fail, and cell death can occur. If this process is ongoing, it affects tissue and organ function, causing disease. The objective of this review is to consolidate and classify a broad number of PCOS studies to understand how various mitochondrial processes impact reproductive organs, including the ovary (oocytes and granulosa cells), uterus, placenta, and circulation, causing reproductive pathophysiology. A secondary objective is to uncover the potential role of mitochondria in the transgenerational transmission of PCOS and metabolic disorders.
Collapse
Affiliation(s)
- Kyle M. Siemers
- Physician Scientist (MD/Ph.D.) Program, Sanford School of Medicine, University of South Dakota, 414 E. Clark Street, Vermillion, SD 57069, USA;
| | - Abigail K. Klein
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Lee Medical Building, 414 E. Clark St., Sioux Falls, SD 57069, USA;
| | - Michelle L. Baack
- Department of Pediatrics, Division of Neonatology, Sanford School of Medicine, University of South Dakota, 1400 W. 22nd St., Sioux Falls, SD 57105, USA
- Environmental Influences on Health and Disease Group, Sanford Research, 2301 E. 60th St., Sioux Falls, SD 57104, USA
| |
Collapse
|
19
|
Chapa-Dubocq XR, Rodríguez-Graciani KM, García-Báez J, Vadovsky A, Bazil JN, Javadov S. The Role of Swelling in the Regulation of OPA1-Mediated Mitochondrial Function in the Heart In Vitro. Cells 2023; 12:2017. [PMID: 37626827 PMCID: PMC10453793 DOI: 10.3390/cells12162017] [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: 06/18/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Optic atrophy-1 (OPA1) plays a crucial role in the regulation of mitochondria fusion and participates in maintaining the structural integrity of mitochondrial cristae. Here we elucidate the role of OPA1 cleavage induced by calcium swelling in the presence of Myls22 (an OPA1 GTPase activity inhibitor) and TPEN (an OMA1 inhibitor). The rate of ADP-stimulated respiration was found diminished by both inhibitors, and they did not prevent Ca2+-induced mitochondrial respiratory dysfunction, membrane depolarization, or swelling. L-OPA1 cleavage was stimulated at state 3 respiration; therefore, our data suggest that L-OPA1 cleavage produces S-OPA1 to maintain mitochondrial bioenergetics in response to stress.
Collapse
Affiliation(s)
- Xavier R. Chapa-Dubocq
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Keishla M. Rodríguez-Graciani
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Jorge García-Báez
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| | - Alyssa Vadovsky
- Department of Physiology, Michigan State University, East Lansing, MI 48824-1046, USA; (A.V.); (J.N.B.)
| | - Jason N. Bazil
- Department of Physiology, Michigan State University, East Lansing, MI 48824-1046, USA; (A.V.); (J.N.B.)
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (J.G.-B.)
| |
Collapse
|
20
|
Bhatti JS, Kaur S, Mishra J, Dibbanti H, Singh A, Reddy AP, Bhatti GK, Reddy PH. Targeting dynamin-related protein-1 as a potential therapeutic approach for mitochondrial dysfunction in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166798. [PMID: 37392948 DOI: 10.1016/j.bbadis.2023.166798] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that manifests its pathology through synaptic damage, mitochondrial abnormalities, microRNA deregulation, hormonal imbalance, increased astrocytes & microglia, accumulation of amyloid β (Aβ) and phosphorylated Tau in the brains of AD patients. Despite extensive research, the effective treatment of AD is still unknown. Tau hyperphosphorylation and mitochondrial abnormalities are involved in the loss of synapses, defective axonal transport and cognitive decline in patients with AD. Mitochondrial dysfunction is evidenced by enhanced mitochondrial fragmentation, impaired mitochondrial dynamics, mitochondrial biogenesis and defective mitophagy in AD. Hence, targeting mitochondrial proteins might be a promising therapeutic strategy in treating AD. Recently, dynamin-related protein 1 (Drp1), a mitochondrial fission protein, has gained attention due to its interactions with Aβ and hyperphosphorylated Tau, altering mitochondrial morphology, dynamics, and bioenergetics. These interactions affect ATP production in mitochondria. A reduction in Drp1 GTPase activity protects against neurodegeneration in AD models. This article provides a comprehensive overview of Drp1's involvement in oxidative damage, apoptosis, mitophagy, and axonal transport of mitochondria. We also highlighted the interaction of Drp1 with Aβ and Tau, which may contribute to AD progression. In conclusion, targeting Drp1 could be a potential therapeutic approach for preventing AD pathology.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India.
| | - Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Jayapriya Mishra
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | | | - Arti Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - P Hemachandra Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA; Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
21
|
San J, Hu J, Pang H, Zuo W, Su N, Guo Z, Wu G, Yang J. Taurine Protects against the Fatty Liver Hemorrhagic Syndrome in Laying Hens through the Regulation of Mitochondrial Homeostasis. Int J Mol Sci 2023; 24:10360. [PMID: 37373507 DOI: 10.3390/ijms241210360] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease caused by fat deposition in the liver of humans and mammals, while fatty liver hemorrhagic syndrome (FLHS) is a fatty liver disease in laying hens which can increase the mortality and cause severe economic losses to the laying industry. Increasing evidence has shown a close relationship between the occurrence of fatty liver disease and the disruption of mitochondrial homeostasis. Studies have proven that taurine can regulate hepatic fat metabolism, reduce hepatic fatty deposition, inhibit oxidative stress, and alleviate mitochondrial dysfunction. However, the mechanisms by which taurine regulates mitochondrial homeostasis in hepatocytes need to be further studied. In this study, we determined the effects and mechanisms of taurine on high-energy low-protein diet-induced FLHS in laying hens and in cultured hepatocytes in free fatty acid (FFA)-induced steatosis. The liver function, lipid metabolism, antioxidant capacity, mitochondrial function, mitochondrial dynamics, autophagy, and biosynthesis were detected. The results showed impaired liver structure and function, mitochondrial damage and dysfunction, lipid accumulation, and imbalance between mitochondrial fusion and fission, mitochondrial autophagy, and biosynthesis in both FLHS hens and steatosis hepatocytes. Taurine administration can significantly inhibit the occurrence of FLHS, protect mitochondria in hepatocytes from disease induced by lipid accumulation and FFA, up-regulate the expression levels of Mfn1, Mfn2, Opa1, LC3I, LC3II, PINK1, PGC-1α, Nrf1, Nrf2, and Tfam, and down-regulate the expression levels of Fis1, Drp1, and p62. In conclusion, taurine can protect laying hens from FLHS through the regulation of mitochondrial homeostasis, including the regulation of mitochondrial dynamics, autophagy, and biosynthesis.
Collapse
Affiliation(s)
- Jishuang San
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Huiping Pang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Wenjun Zuo
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Na Su
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Zimeng Guo
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
22
|
Abstract
Over the last half century, the autofluorescence of the metabolic cofactors NADH (reduced nicotinamide adenine dinucleotide) and FAD (flavin adenine dinucleotide) has been quantified in a variety of cell types and disease states. With the spread of nonlinear optical microscopy techniques in biomedical research, NADH and FAD imaging has offered an attractive solution to noninvasively monitor cell and tissue status and elucidate dynamic changes in cell or tissue metabolism. Various tools and methods to measure the temporal, spectral, and spatial properties of NADH and FAD autofluorescence have been developed. Specifically, an optical redox ratio of cofactor fluorescence intensities and NADH fluorescence lifetime parameters have been used in numerous applications, but significant work remains to mature this technology for understanding dynamic changes in metabolism. This article describes the current understanding of our optical sensitivity to different metabolic pathways and highlights current challenges in the field. Recent progress in addressing these challenges and acquiring more quantitative information in faster and more metabolically relevant formats is also discussed.
Collapse
Affiliation(s)
- Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering and the Arkansas Integrative Metabolic Research Center, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
23
|
Szabo L, Grimm A, García-León JA, Verfaillie CM, Eckert A. Genetically Engineered Triple MAPT-Mutant Human-Induced Pluripotent Stem Cells (N279K, P301L, and E10+16 Mutations) Exhibit Impairments in Mitochondrial Bioenergetics and Dynamics. Cells 2023; 12:1385. [PMID: 37408218 DOI: 10.3390/cells12101385] [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: 03/27/2023] [Revised: 04/28/2023] [Accepted: 05/11/2023] [Indexed: 07/07/2023] Open
Abstract
Pathological abnormalities in the tau protein give rise to a variety of neurodegenerative diseases, conjointly termed tauopathies. Several tau mutations have been identified in the tau-encoding gene MAPT, affecting either the physical properties of tau or resulting in altered tau splicing. At early disease stages, mitochondrial dysfunction was highlighted with mutant tau compromising almost every aspect of mitochondrial function. Additionally, mitochondria have emerged as fundamental regulators of stem cell function. Here, we show that compared to the isogenic wild-type triple MAPT-mutant human-induced pluripotent stem cells, bearing the pathogenic N279K, P301L, and E10+16 mutations, exhibit deficits in mitochondrial bioenergetics and present altered parameters linked to the metabolic regulation of mitochondria. Moreover, we demonstrate that the triple tau mutations disturb the cellular redox homeostasis and modify the mitochondrial network morphology and distribution. This study provides the first characterization of disease-associated tau-mediated mitochondrial impairments in an advanced human cellular tau pathology model at early disease stages, ranging from mitochondrial bioenergetics to dynamics. Consequently, comprehending better the influence of dysfunctional mitochondria on the development and differentiation of stem cells and their contribution to disease progression may thus assist in the potential prevention and treatment of tau-related neurodegeneration.
Collapse
Affiliation(s)
- Leonora Szabo
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| | - Amandine Grimm
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
- Department of Biomedicine, University of Basel, 4055 Basel, Switzerland
| | - Juan Antonio García-León
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, 3000 Leuven, Belgium
| | - Anne Eckert
- Research Cluster Molecular and Cognitive Neurosciences, University of Basel, 4002 Basel, Switzerland
- Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, 4002 Basel, Switzerland
| |
Collapse
|
24
|
Rickard BP, Overchuk M, Chappell VA, Kemal Ruhi M, Sinawang PD, Nguyen Hoang TT, Akin D, Demirci U, Franco W, Fenton SE, Santos JH, Rizvi I. Methods to Evaluate Changes in Mitochondrial Structure and Function in Cancer. Cancers (Basel) 2023; 15:2564. [PMID: 37174030 PMCID: PMC10177605 DOI: 10.3390/cancers15092564] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Mitochondria are regulators of key cellular processes, including energy production and redox homeostasis. Mitochondrial dysfunction is associated with various human diseases, including cancer. Importantly, both structural and functional changes can alter mitochondrial function. Morphologic and quantifiable changes in mitochondria can affect their function and contribute to disease. Structural mitochondrial changes include alterations in cristae morphology, mitochondrial DNA integrity and quantity, and dynamics, such as fission and fusion. Functional parameters related to mitochondrial biology include the production of reactive oxygen species, bioenergetic capacity, calcium retention, and membrane potential. Although these parameters can occur independently of one another, changes in mitochondrial structure and function are often interrelated. Thus, evaluating changes in both mitochondrial structure and function is crucial to understanding the molecular events involved in disease onset and progression. This review focuses on the relationship between alterations in mitochondrial structure and function and cancer, with a particular emphasis on gynecologic malignancies. Selecting methods with tractable parameters may be critical to identifying and targeting mitochondria-related therapeutic options. Methods to measure changes in mitochondrial structure and function, with the associated benefits and limitations, are summarized.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
| | - Vesna A. Chappell
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul 34684, Turkey
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Tina Thuy Nguyen Hoang
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Suzanne E. Fenton
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
25
|
Xiao Z, Zhang X, Li G, Sun L, Li J, Jing Z, Qiu Q, He G, Gao C, Sun X. Tibial fracture surgery in elderly mice caused postoperative neurocognitive disorder via SOX2OT lncRNA in the hippocampus. Mol Brain 2023; 16:36. [PMID: 37098623 PMCID: PMC10131420 DOI: 10.1186/s13041-023-01024-y] [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: 12/08/2022] [Accepted: 03/30/2023] [Indexed: 04/27/2023] Open
Abstract
Increasing evidence indicates the major role of mitochondrial function in neurodegenerative disease. However, it is unclear whether mitochondrial dynamics directly affect postoperative neurocognitive disorder (PND). This study aimed to analyze the underlying mechanisms of mitochondrial dynamics in the pathogenesis of PND. Tibial fracture surgery was performed in elderly mice to generate a PND model in vivo. Cognitive behavior was evaluated 3 days post-surgery using novel object recognition and fear conditioning. A gradual increase in the SOX2OT mRNA level and decrease in the SOX2 mRNA level were noted, with impaired cognitive function, in the mice 3 days after tibial surgery compared with mice in the sham group. To evaluate the role of SOX2OT in PND, SOX2OT knockdown was performed in vitro and in vivo using lentivirus transfection in HT22 cells and via brain stereotactic injection of lentivirus, respectively. SOX2OT knockdown reduced apoptosis, inhibited oxidative stress, suppressed mitochondrial hyperdivision, attenuated surgery-induced cognitive dysfunction, and promoted downstream SOX2 expression in elderly mice. Furthermore, Sox2 alleviated mitochondrial functional damage by inhibiting the transcription of mitochondrial division protein Drp1. Our study findings indicate that SOX2OT knockout alleviates surgery-induced mitochondrial fission and cognitive function defects by upregulating the expression of Sox2 in mice, resulting in the inhibition of drp1 transcription. Therefore, regulation of the SOX2/Drp1 pathway may be a potential mechanism for the treatment of patients with PND.
Collapse
Affiliation(s)
- Zhibin Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
- Department of Anesthesiology, The 986th Air Force Hospital, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiajing Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710032, Shaanxi, China
| | - Guangyao Li
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Li Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiangjing Li
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Ziwei Jing
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Qingya Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Guangxiang He
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Changjun Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xude Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
26
|
Barichello T, Giridharan VV, Catalão CHR, Ritter C, Dal-Pizzol F. Neurochemical effects of sepsis on the brain. Clin Sci (Lond) 2023; 137:401-414. [PMID: 36942500 PMCID: PMC11315270 DOI: 10.1042/cs20220549] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Sepsis is a life-threatening organ dysfunction triggered by a dysregulated host immune response to eliminate an infection. After the host immune response is activated, a complex, dynamic, and time-dependent process is triggered. This process promotes the production of inflammatory mediators, including acute-phase proteins, complement system proteins, cytokines, chemokines, and antimicrobial peptides, which are required to initiate an inflammatory environment for eliminating the invading pathogen. The physiological response of this sepsis-induced systemic inflammation can affect blood-brain barrier (BBB) function; subsequently, endothelial cells produce inflammatory mediators, including cytokines, chemokines, and matrix metalloproteinases (MMPs) that degrade tight junction (TJ) proteins and decrease BBB function. The resulting BBB permeability allows peripheral immune cells from the bloodstream to enter the brain, which then release a range of inflammatory mediators and activate glial cells. The activated microglia and astrocytes release reactive oxygen species (ROS), cytokines, chemokines, and neurochemicals, initiate mitochondrial dysfunction and neuronal damage, and exacerbate the inflammatory milieu in the brain. These changes trigger sepsis-associated encephalopathy (SAE), which has the potential to increase cognitive deterioration and susceptibility to cognitive decline later in life.
Collapse
Affiliation(s)
- Tatiana Barichello
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, U.S.A
- Graduate Program in Health Sciences, Department of Medicine, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, U.S.A
| | - Carlos Henrique R Catalão
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, U.S.A
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of São Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Cristiane Ritter
- Graduate Program in Health Sciences, Department of Medicine, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Graduate Program in Health Sciences, Department of Medicine, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| |
Collapse
|
27
|
Reduced OPA1, Mitochondrial Fragmentation and Increased Susceptibility to Apoptosis in Granular Corneal Dystrophy Type 2 Corneal Fibroblasts. Genes (Basel) 2023; 14:genes14030566. [PMID: 36980838 PMCID: PMC10048436 DOI: 10.3390/genes14030566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The progressive degeneration of granular corneal dystrophy type 2 (GCD2) corneal fibroblasts is associated with altered mitochondrial function, but the underlying mechanisms are incompletely understood. We investigated whether an imbalance of mitochondrial dynamics contributes to mitochondrial dysfunction of GCD2 corneal fibroblasts. Transmission electron microscopy revealed several small, structurally abnormal mitochondria with altered cristae morphology in GCD2 corneal fibroblasts. Confocal microscopy showed enhanced mitochondrial fission and fragmented mitochondrial tubular networks. Western blotting revealed higher levels of MFN1, MFN2, and pDRP1 and decreased levels of OPA1 and FIS1 in GCD2. OPA1 reduction by short hairpin RNA (shRNA) resulted in fragmented mitochondrial tubular networks and increased susceptibility to mitochondrial stress-induced apoptosis. A decrease in the mitochondrial biogenesis-related transcription factors NRF1 and PGC1α was observed, while there was an increase in the mitochondrial membrane proteins TOM20 and TIM23. Additionally, reduced levels of mitochondrial DNA (mtDNA) were exhibited in GCD2 corneal fibroblasts. These observations suggest that altered mitochondrial fission/fusion and biogenesis are the critical molecular mechanisms that cause mitochondrial dysfunction contributing to the degeneration of GCD2 corneal fibroblasts.
Collapse
|
28
|
Wang B, Shi H, Yang B, Miao Z, Sun M, Yang H, Xu X. The mitochondrial Ahi1/GR participates the regulation on mtDNA copy numbers and brain ATP levels and modulates depressive behaviors in mice. Cell Commun Signal 2023; 21:21. [PMID: 36691038 PMCID: PMC9869592 DOI: 10.1186/s12964-022-01034-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Previous studies have shown that depression is often accompanied by an increase in mtDNA copy number and a decrease in ATP levels; however, the exact regulatory mechanisms remain unclear. METHODS In the present study, Western blot, cell knockdown, immunofluorescence, immunoprecipitation and ChIP-qPCR assays were used to detect changes in the Ahi1/GR-TFAM-mtDNA pathway in the brains of neuronal Abelson helper integration site-1 (Ahi1) KO mice and dexamethasone (Dex)-induced mice to elucidate the pathogenesis of depression. In addition, a rescue experiment was performed to determine the effects of regular exercise on the Ahi1/GR-TFAM-mtDNA-ATP pathway and depression-like behavior in Dex-induced mice and Ahi1 KO mice under stress. RESULTS In this study, we found that ATP levels decreased and mitochondrial DNA (mtDNA) copy numbers increased in depression-related brain regions in Dex-induced depressive mice and Ahi1 knockout (KO) mice. In addition, Ahi1 and glucocorticoid receptor (GR), two important proteins related to stress and depressive behaviors, were significantly decreased in the mitochondria under stress. Intriguingly, GR can bind to the D-loop control region of mitochondria and regulate mitochondrial replication and transcription. Importantly, regular exercise significantly increased mitochondrial Ahi1/GR levels and ATP levels and thus improved depression-like behaviors in Dex-induced depressive mice but not in Ahi1 KO mice under stress. CONCLUSIONS In summary, our findings demonstrated that the mitochondrial Ahi1/GR complex and TFAM coordinately regulate mtDNA copy numbers and brain ATP levels by binding to the D-loop region of mtDNA Regular exercise increases the levels of the mitochondrial Ahi1/GR complex and improves depressive behaviors. Video Abstract.
Collapse
Affiliation(s)
- Bin Wang
- Department of Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Haixia Shi
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Bo Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Miao Sun
- Department of Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hao Yang
- Department of Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Xingshun Xu
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
29
|
Lin DS, Huang YW, Ho CS, Huang TS, Lee TH, Wu TY, Huang ZD, Wang TJ. Impact of Mitochondrial A3243G Heteroplasmy on Mitochondrial Bioenergetics and Dynamics of Directly Reprogrammed MELAS Neurons. Cells 2022; 12:cells12010015. [PMID: 36611807 PMCID: PMC9818214 DOI: 10.3390/cells12010015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The MELAS syndrome primarily affecting the CNS is mainly caused by the m.A3243G mutation. The heteroplasmy in different tissues affects the phenotypic spectrum, yet the impact of various levels of m.A3243G heteroplasmy on CNS remains elusive due to the lack of a proper neuronal model harboring m.A3243G mutation. We generated induced neurons (iNs) through the direct reprogramming of MELAS patients, with derived fibroblasts harboring high (>95%), intermediate (68%), and low (20%) m.A3243G mutation. iNs demonstrated neuronal morphology with neurite outgrowth, branching, and dendritic spines. The heteroplasmy and deficiency of respiratory chain complexes were retained in MELAS iNs. High heteroplasmy elicited the elevation in ROS levels and the disruption of mitochondrial membrane potential. Furthermore, high and intermediate heteroplasmy led to the impairment of mitochondrial bioenergetics and a change in mitochondrial dynamics toward the fission and fragmentation of mitochondria, with a reduction in mitochondrial networks. Moreover, iNs derived from aged individuals manifested with mitochondrial fission. These results help us in understanding the impact of various heteroplasmic levels on mitochondrial bioenergetics and mitochondrial dynamics in neurons as the underlying pathomechanism of neurological manifestations of MELAS syndrome. Furthermore, these findings provide targets for further pharmacological approaches of mitochondrial diseases and validate iNs as a reliable platform for studies in neuronal aspects of aging, neurodegenerative disorders, and mitochondrial diseases.
Collapse
Affiliation(s)
- Dar-Shong Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei 25245, Taiwan
- Correspondence: ; Tel.: +886-2-2809-4661; Fax: +886-2-2809-4679
| | - Yu-Wen Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Che-Sheng Ho
- Department of Medicine, Mackay Medical College, New Taipei 25245, Taiwan
- Department of Neurology, Mackay Children’s Hospital, Taipei 10449, Taiwan
| | - Tung-Sun Huang
- Department of Surgery, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsung-Han Lee
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsu-Yen Wu
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Zon-Darr Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
| |
Collapse
|
30
|
Xie W, Guo D, Li J, Yue L, Kang Q, Chen G, Zhou T, Wang H, Zhuang K, Leng L, Li H, Chen Z, Gao W, Zhang J. CEND1 deficiency induces mitochondrial dysfunction and cognitive impairment in Alzheimer's disease. Cell Death Differ 2022; 29:2417-2428. [PMID: 35732922 PMCID: PMC9751129 DOI: 10.1038/s41418-022-01027-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disease featured with memory loss and cognitive function impairments. Chronic mitochondrial stress is a vital pathogenic factor for AD and finally leads to massive neuronal death. However, the underlying mechanism is unclear. By proteomic analysis, we identified a new mitochondrial protein, cell-cycle exit and neuronal differentiation 1 (CEND1), which was decreased significantly in the brain of 5xFAD mice. CEND1 is a neuronal specific protein and locates in the presynaptic mitochondria. Depletion of CEND1 leads to increased mitochondrial fission mediated by upregulation of dynamin related protein 1 (Drp1), resulting in abnormal mitochondrial functions. CEND1 deficiency leads to cognitive impairments in mice. Overexpression of CEND1 in the hippocampus of 5xFAD mice rescued cognitive deficits. Moreover, we identified that CDK5/p25 interacted with and phosphorylated CEND1 which promoted its degradation. Our study provides new mechanistic insights in mitochondrial function regulations by CEND1 in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenting Xie
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Dong Guo
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jieyin Li
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Lei Yue
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Qi Kang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Guimiao Chen
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Tingwen Zhou
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Han Wang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Kai Zhuang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Lige Leng
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Huifang Li
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhenyi Chen
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, China
| | - Weiwei Gao
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, Fujian, 350004, China.
| | - Jie Zhang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, Fujian, 350004, China.
- Department of Anesthesiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
31
|
Ley-Ngardigal S, Bertolin G. Approaches to monitor ATP levels in living cells: where do we stand? FEBS J 2022; 289:7940-7969. [PMID: 34437768 DOI: 10.1111/febs.16169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
ATP is the most universal and essential energy molecule in cells. This is due to its ability to store cellular energy in form of high-energy phosphate bonds, which are extremely stable and readily usable by the cell. This energy is key for a variety of biological functions such as cell growth and division, metabolism, and signaling, and for the turnover of biomolecules. Understanding how ATP is produced and hydrolyzed with a spatiotemporal resolution is necessary to understand its functions both in physiological and in pathological contexts. In this review, first we will describe the organization of the electron transport chain and ATP synthase, the main molecular motor for ATP production in mitochondria. Second, we will review the biochemical assays currently available to estimate ATP quantities in cells, and we will compare their readouts, strengths, and weaknesses. Finally, we will explore the palette of genetically encoded biosensors designed for microscopy-based approaches, and show how their spatiotemporal resolution opened up the possibility to follow ATP levels in living cells.
Collapse
Affiliation(s)
- Seyta Ley-Ngardigal
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France.,LVMH Research Perfumes and Cosmetics, Saint-Jean-de-Braye, France
| | - Giulia Bertolin
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France
| |
Collapse
|
32
|
Shackleford G, Marziali LN, Sasaki Y, Claessens A, Ferri C, Weinstock NI, Rossor AM, Silvestri NJ, Wilson ER, Hurley E, Kidd GJ, Manohar S, Ding D, Salvi RJ, Feltri ML, D’Antonio M, Wrabetz L. A new mouse model of Charcot-Marie-Tooth 2J neuropathy replicates human axonopathy and suggest alteration in axo-glia communication. PLoS Genet 2022; 18:e1010477. [PMID: 36350884 PMCID: PMC9707796 DOI: 10.1371/journal.pgen.1010477] [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: 05/20/2022] [Revised: 11/29/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
Myelin is essential for rapid nerve impulse propagation and axon protection. Accordingly, defects in myelination or myelin maintenance lead to secondary axonal damage and subsequent degeneration. Studies utilizing genetic (CNPase-, MAG-, and PLP-null mice) and naturally occurring neuropathy models suggest that myelinating glia also support axons independently from myelin. Myelin protein zero (MPZ or P0), which is expressed only by Schwann cells, is critical for myelin formation and maintenance in the peripheral nervous system. Many mutations in MPZ are associated with demyelinating neuropathies (Charcot-Marie-Tooth disease type 1B [CMT1B]). Surprisingly, the substitution of threonine by methionine at position 124 of P0 (P0T124M) causes axonal neuropathy (CMT2J) with little to no myelin damage. This disease provides an excellent paradigm to understand how myelinating glia support axons independently from myelin. To study this, we generated targeted knock-in MpzT124M mutant mice, a genetically authentic model of T124M-CMT2J neuropathy. Similar to patients, these mice develop axonopathy between 2 and 12 months of age, characterized by impaired motor performance, normal nerve conduction velocities but reduced compound motor action potential amplitudes, and axonal damage with only minor compact myelin modifications. Mechanistically, we detected metabolic changes that could lead to axonal degeneration, and prominent alterations in non-compact myelin domains such as paranodes, Schmidt-Lanterman incisures, and gap junctions, implicated in Schwann cell-axon communication and axonal metabolic support. Finally, we document perturbed mitochondrial size and distribution along MpzT124M axons suggesting altered axonal transport. Our data suggest that Schwann cells in P0T124M mutant mice cannot provide axons with sufficient trophic support, leading to reduced ATP biosynthesis and axonopathy. In conclusion, the MpzT124M mouse model faithfully reproduces the human neuropathy and represents a unique tool for identifying the molecular basis for glial support of axons.
Collapse
Affiliation(s)
- Ghjuvan’Ghjacumu Shackleford
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Leandro N. Marziali
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Yo Sasaki
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, United States of America
| | - Anke Claessens
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Ferri
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Nadav I. Weinstock
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Alexander M. Rossor
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Nicholas J. Silvestri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Emma R. Wilson
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Edward Hurley
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Grahame J. Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Dalian Ding
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Richard J. Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - M. Laura Feltri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Maurizio D’Antonio
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Lawrence Wrabetz
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| |
Collapse
|
33
|
Jiang C, Okazaki T. Control of mitochondrial dynamics and apoptotic pathways by peroxisomes. Front Cell Dev Biol 2022; 10:938177. [PMID: 36158224 PMCID: PMC9500405 DOI: 10.3389/fcell.2022.938177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Peroxisomes are organelles containing different enzymes that catalyze various metabolic pathways such as β-oxidation of very long-chain fatty acids and synthesis of plasmalogens. Peroxisome biogenesis is controlled by a family of proteins called peroxins, which are required for peroxisomal membrane formation, matrix protein transport, and division. Mutations of peroxins cause metabolic disorders called peroxisomal biogenesis disorders, among which Zellweger syndrome (ZS) is the most severe. Although patients with ZS exhibit severe pathology in multiple organs such as the liver, kidney, brain, muscle, and bone, the pathogenesis remains largely unknown. Recent findings indicate that peroxisomes regulate intrinsic apoptotic pathways and upstream fission-fusion processes, disruption of which causes multiple organ dysfunctions reminiscent of ZS. In this review, we summarize recent findings about peroxisome-mediated regulation of mitochondrial morphology and its possible relationship with the pathogenesis of ZS.
Collapse
Affiliation(s)
- Chenxing Jiang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomohiko Okazaki
- Laboratory of Molecular Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- *Correspondence: Tomohiko Okazaki,
| |
Collapse
|
34
|
Mdivi-1 Induced Mitochondrial Fusion as a Potential Mechanism to Enhance Stress Tolerance in Wheat. Life (Basel) 2022; 12:life12091386. [PMID: 36143422 PMCID: PMC9503966 DOI: 10.3390/life12091386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Mitochondria play a key role in providing energy to cells. This paper is dedicated to elucidating mitochondria-dependent mechanisms that may enhance abiotic stress tolerance in wheat. Mitochondria are constantly undergoing dynamic processes of fusion and fission. In plants, stressful conditions tend to favor mitochondrial fusion processes. The role of mitochondrial fusion was studied by applying Mdivi-1, an inhibitor of mitochondrial fission, to wheat roots subjected to a wounding stress. Increased mitochondrial functional activity and upregulation of genes involved in energy metabolism suggest that mitochondrial fusion is associated with a general activation of energy metabolism. Controlling mitochondrial fusion rates could change the physiology of wheat plants by altering the energy status of the cell and helping to reduce the effects of stress. Abstract Mitochondria play a key role in providing energy to cells. These organelles are constantly undergoing dynamic processes of fusion and fission that change in stressful conditions. The role of mitochondrial fusion in wheat root cells was studied using Mdivi-1, an inhibitor of the mitochondrial fragmentation protein Drp1. The effect of the inhibitor was studied on mitochondrial dynamics in the roots of wheat seedlings subjected to a wounding stress, simulated by excision. Treatment of the stressed roots with the inhibitor increased the size of the mitochondria, enhanced their functional activity, and elevated their membrane potentials. Mitochondrial fusion was accompanied by a decrease in ROS formation and associated cell damage. Exposure to Mdivi-1 also upregulated genes encoding the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and an energy sensor AMP-dependent protein sucrose non-fermenting-related kinase (SnRK1), suggesting that mitochondrial fusion is associated with a general activation of energy metabolism. Controlling mitochondrial fusion rates could change the physiology of wheat plants by altering the energy status of the cell and helping to mitigate the effects of stress.
Collapse
|
35
|
ANT1 overexpression models: Some similarities with facioscapulohumeral muscular dystrophy. Redox Biol 2022; 56:102450. [PMID: 36030628 PMCID: PMC9434167 DOI: 10.1016/j.redox.2022.102450] [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: 07/22/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by progressive muscle weakness. Adenine nucleotide translocator 1 (ANT1), the only 4q35 gene involved in mitochondrial function, is strongly expressed in FSHD skeletal muscle biopsies. However, its role in FSHD is unclear. In this study, we evaluated ANT1 overexpression effects in primary myoblasts from healthy controls and during Xenopus laevis organogenesis. We also compared ANT1 overexpression effects with the phenotype of FSHD muscle cells and biopsies. Here, we report that the ANT1 overexpression-induced phenotype presents some similarities with FSHD muscle cells and biopsies. ANT1-overexpressing muscle cells showed disorganized morphology, altered cytoskeletal arrangement, enhanced mitochondrial respiration/glycolysis, ROS production, oxidative stress, mitochondrial fragmentation and ultrastructure alteration, as observed in FSHD muscle cells. ANT1 overexpression in Xenopus laevis embryos affected skeletal muscle development, impaired skeletal muscle, altered mitochondrial ultrastructure and led to oxidative stress as observed in FSHD muscle biopsies. Moreover, ANT1 overexpression in X. laevis embryos affected heart structure and mitochondrial ultrastructure leading to cardiac arrhythmia, as described in some patients with FSHD. Overall our data suggest that ANT1 could contribute to mitochondria dysfunction and oxidative stress in FSHD muscle cells by modifying their bioenergetic profile associated with ROS production. Such interplay between energy metabolism and ROS production in FSHD will be of significant interest for future prospects.
Collapse
|
36
|
Fine-tuning cell organelle dynamics during mitosis by small GTPases. Front Med 2022; 16:339-357. [PMID: 35759087 DOI: 10.1007/s11684-022-0926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/24/2022] [Indexed: 11/04/2022]
Abstract
During mitosis, the allocation of genetic material concurs with organelle transformation and distribution. The coordination of genetic material inheritance with organelle dynamics directs accurate mitotic progression, cell fate determination, and organismal homeostasis. Small GTPases belonging to the Ras superfamily regulate various cell organelles during division. Being the key regulators of membrane dynamics, the dysregulation of small GTPases is widely associated with cell organelle disruption in neoplastic and non-neoplastic diseases, such as cancer and Alzheimer's disease. Recent discoveries shed light on the molecular properties of small GTPases as sophisticated modulators of a remarkably complex and perfect adaptors for rapid structure reformation. This review collects current knowledge on small GTPases in the regulation of cell organelles during mitosis and highlights the mediator role of small GTPase in transducing cell cycle signaling to organelle dynamics during mitosis.
Collapse
|
37
|
Shiu J, Zhang L, Lentsch G, Flesher JL, Jin S, Polleys CM, Jo SJ, Mizzoni C, Mobasher P, Kwan J, Rius-Diaz F, Tromberg BJ, Georgakoudi I, Nie Q, Balu M, Ganesan AK. Multimodal analyses of vitiligo skin identifies tissue characteristics of stable disease. JCI Insight 2022; 7:154585. [PMID: 35653192 PMCID: PMC9310536 DOI: 10.1172/jci.insight.154585] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Vitiligo is an autoimmune skin disease characterized by the destruction of melanocytes by autoreactive CD8+ T cells. Melanocyte destruction in active vitiligo is mediated by CD8+ T cells, but the persistence of white patches in stable disease is poorly understood. The interaction between immune cells, melanocytes, and keratinocytes in situ in human skin has been difficult to study due to the lack of proper tools. We combine noninvasive multiphoton microscopy (MPM) imaging and single-cell RNA-Seq (scRNA-Seq) to identify subpopulations of keratinocytes in stable vitiligo patients. We show that, compared with nonlesional skin, some keratinocyte subpopulations are enriched in lesional vitiligo skin and shift their energy utilization toward oxidative phosphorylation. Systematic investigation of cell-to-cell communication networks show that this small population of keratinocyte secrete CXCL9 and CXCL10 to potentially drive vitiligo persistence. Pseudotemporal dynamics analyses predict an alternative differentiation trajectory that generates this new population of keratinocytes in vitiligo skin. Further MPM imaging of patients undergoing punch grafting treatment showed that keratinocytes favoring oxidative phosphorylation persist in nonresponders but normalize in responders. In summary, we couple advanced imaging with transcriptomics and bioinformatics to discover cell-to-cell communication networks and keratinocyte cell states that can perpetuate inflammation and prevent repigmentation.
Collapse
Affiliation(s)
- Jessica Shiu
- Department of Dermatology, University of California, Irvine, Irvine, United States of America
| | - Lihua Zhang
- Department of Mathematics, University of California, Irvine, Irvine, United States of America
| | - Griffin Lentsch
- Beckman Laser Institute and Medical Clinic, University of California, Irvine, Irvine, United States of America
| | - Jessica L Flesher
- Department of Dermatology, Massachusetts General Hospital, Boston, United States of America
| | - Suoqin Jin
- Department of Mathematics, University of California, Irvine, Irvine, United States of America
| | - Christopher M Polleys
- Department of Biomedical Engineering, Tufts University, Medford, United States of America
| | - Seong Jin Jo
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Craig Mizzoni
- Department of Biomedical Engineering, Tufts University, Medford, United States of America
| | - Pezhman Mobasher
- Department of Dermatology, University of California, Irvine, Irvine, United States of America
| | - Jasmine Kwan
- Department of Biomedical Engineering, Tufts University, Medford, United States of America
| | - Francisca Rius-Diaz
- Department of Preventive Medicine and Public Health, University of Malaga, Malaga, Spain
| | - Bruce J Tromberg
- Beckman Laser Institute and Medical Clinic, University of California, Irvine, Irvine, United States of America
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, United States of America
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, United States of America
| | - Mihaela Balu
- Department of Surgery, Beckman Laser Institute and Medical Clinic, University of California, Irvine, Irvine, United States of America
| | - Anand K Ganesan
- Department of Dermatology, University of California, Irvine, Irvine, United States of America
| |
Collapse
|
38
|
Dantas WS, Zunica ERM, Heintz EC, Vandanmagsar B, Floyd ZE, Yu Y, Fujioka H, Hoppel CL, Belmont KP, Axelrod CL, Kirwan JP. Mitochondrial uncoupling attenuates sarcopenic obesity by enhancing skeletal muscle mitophagy and quality control. J Cachexia Sarcopenia Muscle 2022; 13:1821-1836. [PMID: 35304976 PMCID: PMC9178352 DOI: 10.1002/jcsm.12982] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/28/2022] [Accepted: 02/21/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Sarcopenic obesity is a highly prevalent disease with poor survival and ineffective medical interventions. Mitochondrial dysfunction is purported to be central in the pathogenesis of sarcopenic obesity by impairing both organelle biogenesis and quality control. We have previously identified that a mitochondrial-targeted furazano[3,4-b]pyrazine named BAM15 is orally available and selectively lowers respiratory coupling efficiency and protects against diet-induced obesity in mice. Here, we tested the hypothesis that mitochondrial uncoupling simultaneously attenuates loss of muscle function and weight gain in a mouse model of sarcopenic obesity. METHODS Eighty-week-old male C57BL/6J mice with obesity were randomized to 10 weeks of high fat diet (CTRL) or BAM15 (BAM15; 0.1% w/w in high fat diet) treatment. Body weight and food intake were measured weekly. Body composition, muscle function, energy expenditure, locomotor activity, and glucose tolerance were determined after treatment. Skeletal muscle was harvested and evaluated for histology, gene expression, protein signalling, and mitochondrial structure and function. RESULTS BAM15 decreased body weight (54.0 ± 2.0 vs. 42.3 ± 1.3 g, P < 0.001) which was attributable to increased energy expenditure (10.1 ± 0.1 vs. 11.3 ± 0.4 kcal/day, P < 0.001). BAM15 increased muscle mass (52.7 ± 0.4 vs. 59.4 ± 1.0%, P < 0.001), strength (91.1 ± 1.3 vs. 124.9 ± 1.2 g, P < 0.0001), and locomotor activity (347.0 ± 14.4 vs. 432.7 ± 32.0 m, P < 0.001). Improvements in physical function were mediated in part by reductions in skeletal muscle inflammation (interleukin 6 and gp130, both P < 0.05), enhanced mitochondrial function, and improved endoplasmic reticulum homeostasis. Specifically, BAM15 activated mitochondrial quality control (PINK1-ubiquitin binding and LC3II, P < 0.01), increased mitochondrial activity (citrate synthase and complex II activity, all P < 0.05), restricted endoplasmic reticulum (ER) misfolding (decreased oligomer A11 insoluble/soluble ratio, P < 0.0001) while limiting ER stress (decreased PERK signalling, P < 0.0001), apoptotic signalling (decreased cytochrome C release and Caspase-3/9 activation, all P < 0.001), and muscle protein degradation (decreased 14-kDa actin fragment insoluble/soluble ratio, P < 0.001). CONCLUSIONS Mitochondrial uncoupling by agents such as BAM15 may mitigate age-related decline in muscle mass and function by molecular and cellular bioenergetic adaptations that confer protection against sarcopenic obesity.
Collapse
Affiliation(s)
- Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Elizabeth R M Zunica
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Elizabeth C Heintz
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Z Elizabeth Floyd
- Ubiquitin Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Yongmei Yu
- Ubiquitin Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core, Case Western Reserve University, Cleveland, OH, USA.,Center for Mitochondrial Diseases, Case Western Reserve University of School of Medicine, Cleveland, OH, USA
| | - Charles L Hoppel
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.,Center for Mitochondrial Diseases, Case Western Reserve University of School of Medicine, Cleveland, OH, USA.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn P Belmont
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
39
|
Uchikado Y, Ikeda Y, Ohishi M. Current Understanding of the Pivotal Role of Mitochondrial Dynamics in Cardiovascular Diseases and Senescence. Front Cardiovasc Med 2022; 9:905072. [PMID: 35665261 PMCID: PMC9157625 DOI: 10.3389/fcvm.2022.905072] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 12/11/2022] Open
Abstract
The heart is dependent on ATP production in mitochondria, which is closely associated with cardiovascular disease because of the oxidative stress produced by mitochondria. Mitochondria are highly dynamic organelles that constantly change their morphology to elongated (fusion) or small and spherical (fission). These mitochondrial dynamics are regulated by various small GTPases, Drp1, Fis1, Mitofusin, and Opa1. Mitochondrial fission and fusion are essential to maintain a balance between mitochondrial biogenesis and mitochondrial turnover. Recent studies have demonstrated that mitochondrial dynamics play a crucial role in the development of cardiovascular diseases and senescence. Disruptions in mitochondrial dynamics affect mitochondrial dysfunction and cardiomyocyte survival leading to cardiac ischemia/reperfusion injury, cardiomyopathy, and heart failure. Mitochondrial dynamics and reactive oxygen species production have been associated with endothelial dysfunction, which in turn causes the development of atherosclerosis, hypertension, and even pulmonary hypertension, including pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Here, we review the association between cardiovascular diseases and mitochondrial dynamics, which may represent a potential therapeutic target.
Collapse
Affiliation(s)
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences Kagoshima University, Kagoshima, Japan
| | | |
Collapse
|
40
|
Kim JE, Lee DS, Kim TH, Kang TC. CDDO-Me Attenuates CA1 Neuronal Death by Facilitating RalBP1-Mediated Mitochondrial Fission and 4-HNE Efflux in the Rat Hippocampus Following Status Epilepticus. Antioxidants (Basel) 2022; 11:985. [PMID: 35624848 PMCID: PMC9137584 DOI: 10.3390/antiox11050985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Ras-related protein Ral-A (RalA)-binding protein 1 (RalBP1, also known as Ral-interacting protein of 76 kDa (RLIP76) or Ral-interacting protein 1 (RLIP1 or RIP1)) is involved in the efflux of 4-hydroxynonenal (4-HNE, an end product of lipid peroxidation), as well as mitochondrial fission. In the present study, we found that 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) attenuated CA1 neuronal death and aberrant mitochondrial elongations in these neurons coupled with enhanced RalBP1 expression and reduced 4-HNE levels following status epilepticus (SE). RalBP1 knockdown did not affect mitochondrial dynamics and CA1 neuronal death under physiological and post-SE conditions. Following SE, however, cotreatment of RalBP1 siRNA diminished the effect of CDDO-Me on 4-HNE levels, mitochondrial hyperfusion in CA1 neurons, and CA1 neuronal death. These findings indicate that CDDO-Me may ameliorate CA1 neuronal death by facilitating RalBP1-mediated 4-HNE efflux and mitochondrial fission following SE. Therefore, our findings suggest that increased RalBP1 expression/activity may be one of the considerable targets to protect neurons from SE.
Collapse
Affiliation(s)
| | | | | | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology and Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (D.-S.L.); (T.-H.K.)
| |
Collapse
|
41
|
Role of Mitochondrial Dynamics in Cocaine's Neurotoxicity. Int J Mol Sci 2022; 23:ijms23105418. [PMID: 35628228 PMCID: PMC9145816 DOI: 10.3390/ijms23105418] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 01/25/2023] Open
Abstract
The dynamic balance of mitochondrial fission and fusion maintains mitochondrial homeostasis and optimal function. It is indispensable for cells such as neurons, which rely on the finely tuned mitochondria to carry out their normal physiological activities. The potent psychostimulant cocaine impairs mitochondria as one way it exerts its neurotoxicity, wherein the disturbances in mitochondrial dynamics have been suggested to play an essential role. In this review, we summarize the neurotoxicity of cocaine and the role of mitochondrial dynamics in cellular physiology. Subsequently, we introduce current findings that link disturbed neuronal mitochondrial dynamics with cocaine exposure. Finally, the possible role and potential therapeutic value of mitochondrial dynamics in cocaine neurotoxicity are discussed.
Collapse
|
42
|
Bhatti GK, Gupta A, Pahwa P, Khullar N, Singh S, Navik U, Kumar S, Mastana SS, Reddy AP, Reddy PH, Bhatti JS. Targeting Mitochondrial bioenergetics as a promising therapeutic strategy in metabolic and neurodegenerative diseases. Biomed J 2022; 45:733-748. [PMID: 35568318 PMCID: PMC9661512 DOI: 10.1016/j.bj.2022.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 04/21/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Mitochondria are the organelles that generate energy for the cells and act as biosynthetic and bioenergetic factories, vital for normal cell functioning and human health. Mitochondrial bioenergetics is considered an important measure to assess the pathogenesis of various diseases. Dysfunctional mitochondria affect or cause several conditions involving the most energy-intensive organs, including the brain, muscles, heart, and liver. This dysfunction may be attributed to an alteration in mitochondrial enzymes, increased oxidative stress, impairment of electron transport chain and oxidative phosphorylation, or mutations in mitochondrial DNA that leads to the pathophysiology of various pathological conditions, including neurological and metabolic disorders. The drugs or compounds targeting mitochondria are considered more effective and safer for treating these diseases. In this review, we make an effort to concise the available literature on mitochondrial bioenergetics in various conditions and the therapeutic potential of various drugs/compounds targeting mitochondrial bioenergetics in metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali Punjab, India.
| | - Anshika Gupta
- Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India.
| | - Paras Pahwa
- Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India.
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India.
| | - Satwinder Singh
- Department of Computer Science and Technology, Central University of Punjab, Bathinda, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| | - Shashank Kumar
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, India.
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Arubala P Reddy
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
43
|
Schizosaccharomyces pombe Fzo1 is subjected to the ubiquitin-proteasome-mediated degradation during the stationary phase. INTERNATIONAL MICROBIOLOGY : THE OFFICIAL JOURNAL OF THE SPANISH SOCIETY FOR MICROBIOLOGY 2022; 25:397-404. [PMID: 35075549 DOI: 10.1007/s10123-022-00231-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 02/01/2023]
Abstract
Mitochondria are highly dynamic organelles that undergo fission and fusion to adapt to the metabolic needs of the cell. Mitofusins are dynamin-like GTPases that play a key role in the regulation of mitochondrial fusion and metabolism. In Saccharomyces cerevisiae, mitofusin Fzo1 levels are controlled by post-translational ubiquitination and degradation. However, it is not clear whether the levels of the Schizosaccharomyces pombe mitofusin Fzo1 are similarly regulated. In this study, we examined the expression S. pombe Fzo1 during normal growth. We showed that Fzo1 protein levels but not mRNA expression levels were reduced during the stationary phase. The protein was stabilized by the proteasome inhibitor bortezomib. Disruption of ubc8 encoding a ubiquitin-conjugating enzyme and rsv2 encoding an S. pombe homolog of S. cerevisiae RPN4 known for activating the expression of genes required for proteasomal biogenesis suppresses the proteasomal degradation of Fzo1 during the stationary phase. Overexpression of fzo1 prevents its degradation. Our results suggest that like S. pombe Fzo1 expression is not regulated by transcription but rather by proteolytic degradation during the stationary phase. Our findings also suggest that although S. cerevisiae and S. pombe Fzo1 proteins are regulated by ubiquitin-proteasomal degradation, different ubiquitin-conjugating enzymes (E2) and ubiquitin ligases (E3) are involved in their degradation.
Collapse
|
44
|
Wongkitikamjorn W, Hosomichi J, Wada E, Maeda H, Satrawaha S, Hong H, Hayashi YK, Yoshida KI, Ono T. Gestational Intermittent Hypoxia Induces Mitochondrial Impairment in the Geniohyoid Muscle of Offspring Rats. Cureus 2022; 14:e25088. [PMID: 35600069 PMCID: PMC9117862 DOI: 10.7759/cureus.25088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Gestational intermittent hypoxia (IH), a hallmark of obstructive sleep apnea during gestation, alters respiratory neural control and diaphragm muscle contractile function in the offspring. The geniohyoid (GH) muscle is innervated by the respiratory-related hypoglossal nerve and plays a role in tongue traction and suckling, motor behaviors that then give way to chewing. Here, we aimed to investigate the effects of gestational exposure to IH on the muscle development and metabolism of GH and masseter muscles in male offspring rats. Materials and methods Pregnant Sprague-Dawley rats were exposed to IH (3-min periods of 4-21% O2) for eight hours/day during gestational days 7-20. The GH and masseter muscles from 35-day-old male offspring (n = 6 in each group) were analyzed. Results Gestational IH induction reduced type IIA fiber size in the GH muscle of the offspring but not in the masseter muscle. Western blot analysis showed that gestational IH-induced significant downregulation of peroxisome proliferator-activated receptor (PPAR)-gamma coactivator 1-alpha (PGC1α) protein in the GH muscle but not in the masseter muscle. Moreover, optic atrophy 1 and mitofusin-2 proteins were decreased and mitochondrial fission 1 protein levels were increased in the GH muscle of the offspring exposed to gestational IH. Mitochondrial adenosine triphosphate (ATP) synthase subunit alpha and transcriptional factor A (TFAM) were decreased in the GH muscle post-gestational IH. Conclusion These findings suggest that gestational IH-induced impaired mitochondrial metabolism and alteration of oxidative myofibers of the GH muscle in the pre-adolescent offspring, but not the masseter muscle, owing to the susceptibility of GH muscular mitochondria to gestational IH.
Collapse
Affiliation(s)
- Wirongrong Wongkitikamjorn
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, THA
| | - Jun Hosomichi
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| | - Eiji Wada
- Department of Pathophysiology, Tokyo Medical University, Tokyo, JPN
| | - Hideyuki Maeda
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, JPN
| | - Sirichom Satrawaha
- Department of Orthodontics, Faculty of Dentistry, Chulalongkorn University, Bangkok, THA
| | - Haixin Hong
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen, CHN
| | - Yukiko K Hayashi
- Department of Pathophysiology, Tokyo Medical University, Tokyo, JPN
| | - Ken-Ichi Yoshida
- Department of Forensic Medicine, Tokyo Medical University, Tokyo, JPN
| | - Takashi Ono
- Department of Orthodontic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, JPN
| |
Collapse
|
45
|
Gregorio JD, Petricca S, Iorio R, Toniato E, Flati V. MITOCHONDRIAL AND METABOLIC ALTERATIONS IN CANCER CELLS. Eur J Cell Biol 2022; 101:151225. [DOI: 10.1016/j.ejcb.2022.151225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
|
46
|
Mihajlovic M, Vinken M. Mitochondria as the Target of Hepatotoxicity and Drug-Induced Liver Injury: Molecular Mechanisms and Detection Methods. Int J Mol Sci 2022; 23:ijms23063315. [PMID: 35328737 PMCID: PMC8951158 DOI: 10.3390/ijms23063315] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
One of the major mechanisms of drug-induced liver injury includes mitochondrial perturbation and dysfunction. This is not a surprise, given that mitochondria are essential organelles in most cells, which are responsible for energy homeostasis and the regulation of cellular metabolism. Drug-induced mitochondrial dysfunction can be influenced by various factors and conditions, such as genetic predisposition, the presence of metabolic disorders and obesity, viral infections, as well as drugs. Despite the fact that many methods have been developed for studying mitochondrial function, there is still a need for advanced and integrative models and approaches more closely resembling liver physiology, which would take into account predisposing factors. This could reduce the costs of drug development by the early prediction of potential mitochondrial toxicity during pre-clinical tests and, especially, prevent serious complications observed in clinical settings.
Collapse
|
47
|
Gautam M, Gunay A, Chandel NS, Ozdinler PH. Mitochondrial dysregulation occurs early in ALS motor cortex with TDP-43 pathology and suggests maintaining NAD + balance as a therapeutic strategy. Sci Rep 2022; 12:4287. [PMID: 35277554 PMCID: PMC8917163 DOI: 10.1038/s41598-022-08068-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial defects result in dysregulation of metabolomics and energy homeostasis that are detected in upper motor neurons (UMNs) with TDP-43 pathology, a pathology that is predominantly present in both familial and sporadic cases of amyotrophic lateral sclerosis (ALS). While same mitochondrial problems are present in the UMNs of ALS patients with TDP-43 pathology and UMNs of TDP-43 mouse models, and since pathologies are shared at a cellular level, regardless of species, we first analyzed the metabolite profile of both healthy and diseased motor cortex to investigate whether metabolomic changes occur with respect to TDP-43 pathology. High-performance liquid chromatography, high-resolution mass spectrometry and tandem mass spectrometry (HPLC-MS/MS) for metabolite profiling began to suggest that reduced levels of NAD+ is one of the underlying causes of metabolomic problems. Since nicotinamide mononucleotide (NMN) was reported to restore NAD+ levels, we next investigated whether NMN treatment would improve the health of diseased corticospinal motor neurons (CSMN, a.k.a. UMN in mice). prpTDP-43A315T-UeGFP mice, the CSMN reporter line with TDP-43 pathology, allowed cell-type specific responses of CSMN to NMN treatment to be assessed in vitro. Our results show that metabolomic defects occur early in ALS motor cortex and establishing NAD+ balance could offer therapeutic benefit to UMNs with TDP-43 pathology.
Collapse
Affiliation(s)
- Mukesh Gautam
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Aksu Gunay
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - P Hande Ozdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA. .,Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60611, USA. .,Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA. .,Feinberg School of Medicine, Les Turner ALS Center at Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
48
|
Benaroya H. Understanding mitochondria and the utility of optimization as a canonical framework for identifying and modeling mitochondrial pathways. Rev Neurosci 2022; 33:657-690. [PMID: 35219282 DOI: 10.1515/revneuro-2021-0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/25/2022] [Indexed: 11/15/2022]
Abstract
The goal of this paper is to provide an overview of our current understanding of mitochondrial function as a framework to motivate the hypothesis that mitochondrial behavior is governed by optimization principles that are constrained by the laws of the physical and biological sciences. Then, mathematical optimization tools can generally be useful to model some of these processes under reasonable assumptions and limitations. We are specifically interested in optimizations via variational methods, which are briefly summarized. Within such an optimization framework, we suggest that the numerous mechanical instigators of cell and intracellular functioning can be modeled utilizing some of the principles of mechanics that govern engineered systems, as well as by the frequently observed feedback and feedforward mechanisms that coordinate the multitude of processes within cells. These mechanical aspects would need to be coupled to governing biochemical rules. Of course, biological systems are significantly more complex than engineered systems, and require considerably more experimentation to ascertain and characterize parameters and subsequent behavior. That complexity requires well-defined limitations and assumptions for any derived models. Optimality is being motivated as a framework to help us understand how cellular decisions are made, especially those that transition between physiological behaviors and dysfunctions along pathophysiological pathways. We elaborate on our interpretation of optimality and cellular decision making within the body of this paper, as we revisit these ideas in the numerous different contexts of mitochondrial functions.
Collapse
Affiliation(s)
- Haym Benaroya
- Department of Mechanical and Aerospace Engineering, Rutgers University, 98 Brett Road, Piscataway, NJ 08901, USA
| |
Collapse
|
49
|
Dussouchaud A, Jacob J, Secq C, Verbavatz JM, Moras M, Larghero J, Fader CM, Ostuni MA, Lefevre SD. Transmission Electron Microscopy to Follow Ultrastructural Modifications of Erythroblasts Upon ex vivo Human Erythropoiesis. Front Physiol 2022; 12:791691. [PMID: 35222062 PMCID: PMC8864112 DOI: 10.3389/fphys.2021.791691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 01/02/2023] Open
Abstract
Throughout mammal erythroid differentiation, erythroblasts undergo enucleation and organelle clearance becoming mature red blood cell. Organelles are cleared by autophagic pathways non-specifically targeting organelles and cytosolic content or by specific mitophagy targeting mitochondria. Mitochondrial functions are essential to coordinate metabolism reprogramming, cell death, and differentiation balance, and also synthesis of heme, the prosthetic group needed in hemoglobin assembly. In mammals, mitochondria subcellular localization and mitochondria interaction with other structures as endoplasmic reticulum and nucleus might be of importance for the removal of the nucleus, that is, the enucleation. Here, we aim to characterize by electron microscopy the changes in ultrastructure of cells over successive stages of human erythroblast differentiation. We focus on mitochondria to gain insights into intracellular localization, ultrastructure, and contact with other organelles. We found that mitochondria are progressively cleared with a significant switch between PolyE and OrthoE stages, acquiring a rounded shape and losing contact sites with both ER (MAM) and nucleus (NAM). We studied intracellular vesicle trafficking and found that endosomes and MVBs, known to be involved in iron traffic and heme synthesis, are increased during BasoE to PolyE transition; autophagic structures such as autophagosomes increase from ProE to OrthoE stages. Finally, consistent with metabolic switch, glycogen accumulation was observed in OrthoE stage.
Collapse
Affiliation(s)
- Alice Dussouchaud
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Julieta Jacob
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Charles Secq
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | | | - Martina Moras
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Jérôme Larghero
- CNRS, UMR 7592, Institut Jacques Monod, Université de Paris, Paris, France
| | - Claudio M. Fader
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mariano A. Ostuni
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Sophie D. Lefevre
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
- *Correspondence: Sophie D. Lefevre,
| |
Collapse
|
50
|
Valdés-Aguayo JJ, Garza-Veloz I, Vargas-Rodríguez JR, Martinez-Vazquez MC, Avila-Carrasco L, Bernal-Silva S, González-Fuentes C, Comas-García A, Alvarado-Hernández DE, Centeno-Ramirez ASH, Rodriguez-Sánchez IP, Delgado-Enciso I, Martinez-Fierro ML. Peripheral Blood Mitochondrial DNA Levels Were Modulated by SARS-CoV-2 Infection Severity and Its Lessening Was Associated With Mortality Among Hospitalized Patients With COVID-19. Front Cell Infect Microbiol 2022; 11:754708. [PMID: 34976854 PMCID: PMC8716733 DOI: 10.3389/fcimb.2021.754708] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Introduction During severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the virus hijacks the mitochondria causing damage of its membrane and release of mt-DNA into the circulation which can trigger innate immunity and generate an inflammatory state. In this study, we explored the importance of peripheral blood mt-DNA as an early predictor of evolution in patients with COVID-19 and to evaluate the association between the concentration of mt-DNA and the severity of the disease and the patient’s outcome. Methods A total 102 patients (51 COVID-19 cases and 51 controls) were included in the study. mt-DNA obtained from peripheral blood was quantified by qRT-PCR using the NADH mitochondrial gene. Results There were differences in peripheral blood mt-DNA between patients with COVID-19 (4.25 ng/μl ± 0.30) and controls (3.3 ng/μl ± 0.16) (p = 0.007). Lower mt-DNA concentrations were observed in patients with severe COVID-19 when compared with mild (p= 0.005) and moderate (p= 0.011) cases of COVID-19. In comparison with patients with severe COVID-19 who survived (3.74 ± 0.26 ng/μl) decreased levels of mt-DNA in patients with severe COVID-19 who died (2.4 ± 0.65 ng/μl) were also observed (p = 0.037). Conclusion High levels of mt-DNA were associated with COVID-19 and its decrease could be used as a potential biomarker to establish a prognosis of severity and mortality of patients with COVID-19.
Collapse
Affiliation(s)
- José J Valdés-Aguayo
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - José R Vargas-Rodríguez
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - María C Martinez-Vazquez
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Lorena Avila-Carrasco
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| | - Sofia Bernal-Silva
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Andreu Comas-García
- Departamento de Microbiología, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Diana E Alvarado-Hernández
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | | - Iram P Rodriguez-Sánchez
- Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, Universidad Autónoma de Nuevo León, Nuevo León, Mexico
| | | | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y C.S, Universidad Autónoma de Zacatecas, Zacatecas, Mexico
| |
Collapse
|