1
|
Sisalli MJ, D'Apolito E, Cuomo O, Lombardi G, Tufano M, Annunziato L, Scorziello A. The E3-ligase Siah2 activates mitochondrial quality control in neurons to maintain energy metabolism during ischemic brain tolerance. Cell Death Dis 2025; 16:52. [PMID: 39875361 PMCID: PMC11775118 DOI: 10.1038/s41419-025-07339-z] [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: 11/11/2023] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025]
Abstract
Mitochondrial quality control is crucial for the homeostasis of the mitochondrial network. The balance between mitophagy and biogenesis is needed to reduce cerebral ischemia-induced cell death. Ischemic preconditioning (IPC) represents an adaptation mechanism of CNS that increases tolerance to lethal cerebral ischemia. It has been demonstrated that hypoxia-induced Seven in absentia Homolog 2 (Siah2) E3-ligase activation influences mitochondrial dynamics promoting the degradation of mitochondrial proteins. Therefore, in the present study, we investigated the role of Siah2 in the IPC-induced neuroprotection in in vitro and in vivo models of IPC. To this aim, cortical neurons were exposed to 30-min oxygen and glucose deprivation (OGD, sublethal insult) followed by 3 h OGD plus reoxygenation (lethal insult). Our results revealed that the mitochondrial depolarization induced by hypoxia activates Siah2 at the mitochondrial level and increases LC3-II protein expression, a marker of mitophagy, an effect counteracted by the reoxygenation phase. By contrast, hypoxia reduced the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a marker of mitochondrial biogenesis, whereas its expression was increased after reoxygenation thus improving mitochondrial membrane potential, mitochondrial calcium content, and mitochondrial morphology, hence leading to neuroprotection in IPC. Furthermore, Siah2 silencing confirmed these results. Collectively, these findings indicate that the balance between mitophagy and mitochondrial biogenesis, due to the activation of the Siah2-E3-ligase, might play a role in IPC-induced neuroprotection.
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Elena D'Apolito
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Giovanna Lombardi
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Michele Tufano
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
2
|
Deutschmeyer VE, Schlaudraff NA, Walesch SK, Moyer J, Sokol AM, Graumann J, Meissner W, Schneider M, Muley T, Helmbold P, Schwinn M, Richter AM, Schmitz ML, Dammann RH. SIAH3 is frequently epigenetically silenced in cancer and regulates mitochondrial metabolism. Int J Cancer 2025; 156:353-367. [PMID: 39344659 DOI: 10.1002/ijc.35202] [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: 01/18/2024] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Of the seven in absentia homologue (SIAH) family, three members have been identified in the human genome. In contrast to the E3 ubiquitin ligase encoding SIAH1 and SIAH2, little is known on the regulation and function of SIAH3 in tumorigenesis. In this study, we reveal that SIAH3 is frequently epigenetically silenced in different cancer entities, including cutaneous melanoma, lung adenocarcinoma and head and neck cancer. Low SIAH3 levels correlate with an impaired survival of cancer patients. Additionally, induced expression of SIAH3 reduces cell proliferation and induces cell death. Functionally, SIAH3 negatively affects cellular metabolism by shifting cells form aerobic oxidative phosphorylation to glycolysis. SIAH3 is localized in the mitochondrion and interacts with proteins involved in mitochondrial ribosome biogenesis and translation. We also report that SIAH3 interacts with ubiquitin ligases, including SIAH1 or SIAH2, and is degraded by them. These results suggest that SIAH3 acts as an epigenetically controlled tumor suppressor by regulating cellular metabolism through the inhibition of oxidative phosphorylation.
Collapse
Affiliation(s)
| | - Nico A Schlaudraff
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sara K Walesch
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Janine Moyer
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Anna M Sokol
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Institute of Translational Proteomics, Department of Medicine, Philipps-University, Marburg, Germany
| | - Wolfgang Meissner
- Core Facility for Cellular Metabolism, Department of Medicine, Philipps-University, Marburg, Germany
| | - Marc Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- University of Giessen Marburg Lung Center (UGMLC) and Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Giessen, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- University of Giessen Marburg Lung Center (UGMLC) and Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Giessen, Germany
| | - Peter Helmbold
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Markus Schwinn
- Institute of Biochemistry, Medical Faculty of the University Giessen, Giessen, Germany
| | - Antje M Richter
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Medical Faculty of the University Giessen, Giessen, Germany
| | - Reinhard H Dammann
- Institute for Genetics, Justus-Liebig-University Giessen, Giessen, Germany
- University of Giessen Marburg Lung Center (UGMLC) and Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
3
|
Hsu CY, Faisal A, Jumaa SS, Gilmanova NS, Ubaid M, Athab AH, Mirzaei R, Karampoor S. Exploring the impact of circRNAs on cancer glycolysis: Insights into tumor progression and therapeutic strategies. Noncoding RNA Res 2024; 9:970-994. [PMID: 38770106 PMCID: PMC11103225 DOI: 10.1016/j.ncrna.2024.05.001] [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: 02/19/2024] [Revised: 04/18/2024] [Accepted: 05/04/2024] [Indexed: 05/22/2024] Open
Abstract
Cancer cells exhibit altered metabolic pathways, prominently featuring enhanced glycolytic activity to sustain their rapid growth and proliferation. Dysregulation of glycolysis is a well-established hallmark of cancer and contributes to tumor progression and resistance to therapy. Increased glycolysis supplies the energy necessary for increased proliferation and creates an acidic milieu, which in turn encourages tumor cells' infiltration, metastasis, and chemoresistance. Circular RNAs (circRNAs) have emerged as pivotal players in diverse biological processes, including cancer development and metabolic reprogramming. The interplay between circRNAs and glycolysis is explored, illuminating how circRNAs regulate key glycolysis-associated genes and enzymes, thereby influencing tumor metabolic profiles. In this overview, we highlight the mechanisms by which circRNAs regulate glycolytic enzymes and modulate glycolysis. In addition, we discuss the clinical implications of dysregulated circRNAs in cancer glycolysis, including their potential use as diagnostic and prognostic biomarkers. All in all, in this overview, we provide the most recent findings on how circRNAs operate at the molecular level to control glycolysis in various types of cancer, including hepatocellular carcinoma (HCC), prostate cancer (PCa), colorectal cancer (CRC), cervical cancer (CC), glioma, non-small cell lung cancer (NSCLC), breast cancer, and gastric cancer (GC). In conclusion, this review provides a comprehensive overview of the significance of circRNAs in cancer glycolysis, shedding light on their intricate roles in tumor development and presenting innovative therapeutic avenues.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City, 71710, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, 85004, USA
| | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | - Sally Salih Jumaa
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nataliya Sergeevna Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia, Moscow
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Aya H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom & Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
He Z, Xie L, Liu J, Wei X, Zhang W, Mei Z. Novel insight into the role of A-kinase anchoring proteins (AKAPs) in ischemic stroke and therapeutic potentials. Biomed Pharmacother 2024; 175:116715. [PMID: 38739993 DOI: 10.1016/j.biopha.2024.116715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Ischemic stroke, a devastating disease associated with high mortality and disability worldwide, has emerged as an urgent public health issue. A-kinase anchoring proteins (AKAPs) are a group of signal-organizing molecules that compartmentalize and anchor a wide range of receptors and effector proteins and have a major role in stabilizing mitochondrial function and promoting neurodevelopmental development in the central nervous system (CNS). Growing evidence suggests that dysregulation of AKAPs expression and activity is closely associated with oxidative stress, ion disorder, mitochondrial dysfunction, and blood-brain barrier (BBB) impairment in ischemic stroke. However, the underlying mechanisms remain inadequately understood. This review provides a comprehensive overview of the composition and structure of A-kinase anchoring protein (AKAP) family members, emphasizing their physiological functions in the CNS. We explored in depth the molecular and cellular mechanisms of AKAP complexes in the pathological progression and risk factors of ischemic stroke, including hypertension, hyperglycemia, lipid metabolism disorders, and atrial fibrillation. Herein, we highlight the potential of AKAP complexes as a pharmacological target against ischemic stroke in the hope of inspiring translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Ziyu He
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiyong Liu
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xuan Wei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China.
| |
Collapse
|
5
|
Borriello G, Buonincontri V, de Donato A, Della Corte M, Gravina I, Iulianiello P, Joshi R, Mone P, Cacciola G, Viggiano D. The interplay between sodium/glucose cotransporter type 2 and mitochondrial ionic environment. Mitochondrion 2024; 76:101878. [PMID: 38599300 DOI: 10.1016/j.mito.2024.101878] [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: 12/31/2023] [Revised: 03/04/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Mitochondrial volume is maintained through the permeability of the inner mitochondrial membrane by a specific aquaporin and the osmotic balance between the mitochondrial matrix and cellular cytoplasm. Various electrolytes, such as calcium and hydrogen ions, potassium, and sodium, as well as other osmotic substances, affect the swelling of mitochondria. Intracellular glucose levels may also affect mitochondrial swelling, although the relationship between mitochondrial ion homeostasis and intracellular glucose is poorly understood. This article reviews what is currently known about how the Sodium-Glucose transporter (SGLT) may impact mitochondrial sodium (Na+) homeostasis. SGLTs regulate intracellular glucose and sodium levels and, therefore, interfere with mitochondrial ion homeostasis because mitochondrial Na+ is closely linked to cytoplasmic calcium and sodium dynamics. Recently, a large amount of data has been available on the effects of SGLT2 inhibitors on mitochondria in different cell types, including renal proximal tubule cells, endothelial cells, mesangial cells, podocytes, neuronal cells, and cardiac cells. The current evidence suggests that SGLT inhibitors (SGLTi) may affect mitochondrial dynamics regarding intracellular Sodium and hydrogen ions. Although the regulation of mitochondrial ion channels by SGLTs is still in its infancy, the evidence accumulated thus far of the effect of SGLTi on mitochondrial functions certainly will foster further research in this direction.
Collapse
Affiliation(s)
- Gianmarco Borriello
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | | | - Antonio de Donato
- Biogem, Biology and Molecular Genetics Institute, Ariano Irpino, AV, Italy
| | - Michele Della Corte
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | - Ilenia Gravina
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | - Pietro Iulianiello
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | - Rashmi Joshi
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | - Pasquale Mone
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy; Casa di cura privata Montevergine, Mercogliano, Italy
| | - Giovanna Cacciola
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy
| | - Davide Viggiano
- Dept. Translational Medical Sciences, Univ. Campania, "L Vanvitelli", Naples, Italy.
| |
Collapse
|
6
|
Huang J, Zhu Z, Schlüter D, Lambertsen KL, Song W, Wang X. Ubiquitous regulation of cerebrovascular diseases by ubiquitin-modifying enzymes. Clin Transl Med 2024; 14:e1719. [PMID: 38778460 PMCID: PMC11111633 DOI: 10.1002/ctm2.1719] [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: 03/01/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Cerebrovascular diseases (CVDs) are a major threat to global health. Elucidation of the molecular mechanisms underlying the pathology of CVDs is critical for the development of efficacious preventative and therapeutic approaches. Accumulating studies have highlighted the significance of ubiquitin-modifying enzymes (UMEs) in the regulation of CVDs. UMEs are a group of enzymes that orchestrate ubiquitination, a post-translational modification tightly involved in CVDs. Functionally, UMEs regulate multiple pathological processes in ischemic and hemorrhagic stroke, moyamoya disease, and atherosclerosis. Considering the important roles of UMEs in CVDs, they may become novel druggable targets for these diseases. Besides, techniques applying UMEs, such as proteolysis-targeting chimera and deubiquitinase-targeting chimera, may also revolutionize the therapy of CVDs in the future.
Collapse
Affiliation(s)
- Jingyong Huang
- Department of Vascular SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhenhu Zhu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical SchoolHannoverGermany
| | - Kate Lykke Lambertsen
- Department of Neurobiology ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdense CDenmark
- BRIGDE—Brain Research—Inter‐Disciplinary Guided Excellence, Department of Clinical ResearchUniversity of Southern DenmarkOdense CDenmark
- Department of NeurologyOdense University HospitalOdense CDenmark
| | - Weihong Song
- Oujiang LaboratoryKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceZhejiang Provincial Clinical Research Center for Mental DisordersInstitute of AgingSchool of Mental HealthAffiliated Kangning HospitalThe Second Affiliated HospitalYuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| | - Xu Wang
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryKey Laboratory of Alzheimer's Disease of Zhejiang ProvinceZhejiang Provincial Clinical Research Center for Mental DisordersInstitute of AgingSchool of Mental HealthAffiliated Kangning HospitalThe Second Affiliated HospitalYuying Children's HospitalWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
7
|
Vardar Acar N, Özgül RK. A big picture of the mitochondria-mediated signals: From mitochondria to organism. Biochem Biophys Res Commun 2023; 678:45-61. [PMID: 37619311 DOI: 10.1016/j.bbrc.2023.08.032] [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: 06/06/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Mitochondria, well-known for years as the powerhouse and biosynthetic center of the cell, are dynamic signaling organelles beyond their energy production and biosynthesis functions. The metabolic functions of mitochondria, playing an important role in various biological events both in physiological and stress conditions, transform them into important cellular stress sensors. Mitochondria constantly communicate with the rest of the cell and even from other cells to the organism, transmitting stress signals including oxidative and reductive stress or adaptive signals such as mitohormesis. Mitochondrial signal transduction has a vital function in regulating integrity of human genome, organelles, cells, and ultimately organism.
Collapse
Affiliation(s)
- Neşe Vardar Acar
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - R Köksal Özgül
- Department of Pediatric Metabolism, Institute of Child Health, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
8
|
Hu D, Tan M, Lu D, Kleiboeker B, Liu X, Park H, Kravitz AV, Shoghi KI, Tseng YH, Razani B, Ikeda A, Lodhi IJ. TMEM135 links peroxisomes to the regulation of brown fat mitochondrial fission and energy homeostasis. Nat Commun 2023; 14:6099. [PMID: 37773161 PMCID: PMC10541902 DOI: 10.1038/s41467-023-41849-8] [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: 10/24/2022] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
Mitochondrial morphology, which is controlled by mitochondrial fission and fusion, is an important regulator of the thermogenic capacity of brown adipocytes. Adipose-specific peroxisome deficiency impairs thermogenesis by inhibiting cold-induced mitochondrial fission due to decreased mitochondrial membrane content of the peroxisome-derived lipids called plasmalogens. Here, we identify TMEM135 as a critical mediator of the peroxisomal regulation of mitochondrial fission and thermogenesis. Adipose-specific TMEM135 knockout in mice blocks mitochondrial fission, impairs thermogenesis, and increases diet-induced obesity and insulin resistance. Conversely, TMEM135 overexpression promotes mitochondrial division, counteracts obesity and insulin resistance, and rescues thermogenesis in peroxisome-deficient mice. Mechanistically, thermogenic stimuli promote association between peroxisomes and mitochondria and plasmalogen-dependent localization of TMEM135 in mitochondria, where it mediates PKA-dependent phosphorylation and mitochondrial retention of the fission factor Drp1. Together, these results reveal a previously unrecognized inter-organelle communication regulating mitochondrial fission and energy homeostasis and identify TMEM135 as a potential target for therapeutic activation of BAT.
Collapse
Affiliation(s)
- Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Tan
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuejing Liu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongsuk Park
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kooresh I Shoghi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
9
|
Saha S, Singh P, Dutta A, Vaidya H, Negi PC, Sengupta S, Seth S, Basak T. A Comprehensive Insight and Mechanistic Understanding of the Lipidomic Alterations Associated With DCM. JACC. ASIA 2023; 3:539-555. [PMID: 37614533 PMCID: PMC10442885 DOI: 10.1016/j.jacasi.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 08/25/2023]
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure characterized by the enlargement of the left ventricular cavity and contractile dysfunction of the myocardium. Lipids are the major sources of energy for the myocardium. Impairment of lipid homeostasis has a potential role in the pathogenesis of DCM. In this review, we have summarized the role of different lipids in the progression of DCM that can be considered as potential biomarkers. Further, we have also explained the mechanistic pathways followed by the lipid molecules in disease progression along with the cardioprotective role of certain lipids. As the global epidemiological status of DCM is alarming, it is high time to define some disease-specific biomarkers with greater prognostic value. We are proposing an adaptation of a system lipidomics-based approach to profile DCM patients in order to achieve a better diagnosis and prognosis of the disease.
Collapse
Affiliation(s)
- Shubham Saha
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Abhi Dutta
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| | - Hiteshi Vaidya
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Prakash Chand Negi
- Department of Cardiology, Indira Gandhi Medical College & Hospital, Shimla, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering. IIT-Mandi, Mandi, India
- BioX Center, Indian Institute of Technology-Mandi, Mandi, India
| |
Collapse
|
10
|
Das R, Pandey P, Maurya B, Pradhan P, Sinha D, Mukherjee A, Mutsuddi M. Spoonbill positively regulates JNK signalling mediated apoptosis in Drosophila melanogaster. Eur J Cell Biol 2023; 102:151300. [PMID: 36858008 DOI: 10.1016/j.ejcb.2023.151300] [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: 07/05/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
A-kinase anchoring protein (AKAP) comprises a family of scaffold proteins, which decides the subcellular localisation of a combination of signalling molecules. Spoonbill (Spoon) is a putative A-kinase anchoring protein in Drosophila. We have earlier reported that Spoon suppresses ribonuclear foci formed by trinucleotide repeat expanded transcripts associated with Spinocerebellar Ataxia 8 neurodegeneration in Drosophila. However, the role of Spoonbill in cellular signalling was unexplored. In this report, we have unravelled a novel function of Spoon protein in the regulation of the apoptotic pathway. The Drosophila TNFα homolog, Eiger, induces apoptosis via activation of the JNK pathway. We have shown here that Spoonbill is a positive regulator of the Eiger-induced JNK signalling. Further genetic interaction studies show that the spoon interacts with components of the JNK pathway, TGF-β activated kinase 1 (Tak1 - JNKKK), hemipterous (hep - JNKK) and basket (bsk - JNK). Interestingly, Spoonbill alone can also induce ectopic activation of the JNK pathway in a context-specific manner. To understand the molecular mechanism underlying Spoonbill-mediated modulation of the JNK pathway, the interaction between Spoon and Drosophila JNK was assessed. basket encodes the only known JNK in Drosophila. This serine/threonine-protein kinase phosphorylates Jra/Kay, which transcriptionally regulate downstream targets like Matrix metalloproteinase 1 (Mmp1), puckered (puc), and proapoptotic genes hid, reaper and grim. Interestingly, we found that Spoonbill colocalises and co-immunoprecipitates with the Basket protein in the developing photoreceptor neurons. Hence, we propose that Spoon plays a vital role in JNK-induced apoptosis. Furthermore, stress-induced JNK activation underlying Parkinson's Disease was also examined. In the Parkinson's Drosophila model of neurodegeneration, depletion of Spoonbill leads to a partial reduction of JNK pathway activation, along with improvement in adult motor activity. These observations suggest that the putative scaffold protein Spoonbill is a functional and physical interacting partner of the Drosophila JNK protein, Basket. Spoon protein is localised on the outer mitochondrial membrane (OMM), which may perhaps provide a suitable subcellular niche for activation of Drosophila Basket protein by its kinases which induce apoptosis.
Collapse
Affiliation(s)
- Rituparna Das
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Pranjali Pandey
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Bhawana Maurya
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | | | - Devanjan Sinha
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
11
|
Rinaldi L, Senatore E, Iannucci R, Chiuso F, Feliciello A. Control of Mitochondrial Activity by the Ubiquitin Code in Health and Cancer. Cells 2023; 12:234. [PMID: 36672167 PMCID: PMC9856579 DOI: 10.3390/cells12020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cellular homeostasis is tightly connected to the broad variety of mitochondrial functions. To stay healthy, cells need a constant supply of nutrients, energy production and antioxidants defenses, undergoing programmed death when a serious, irreversible damage occurs. The key element of a functional integration of all these processes is the correct crosstalk between cell signaling and mitochondrial activities. Once this crosstalk is interrupted, the cell is not able to communicate its needs to mitochondria, resulting in oxidative stress and development of pathological conditions. Conversely, dysfunctional mitochondria may affect cell viability, even in the presence of nutrients supply and energy production, indicating the existence of feed-back control mechanisms between mitochondria and other cellular compartments. The ubiquitin proteasome system (UPS) is a multi-step biochemical pathway that, through the conjugation of ubiquitin moieties to specific protein substrates, controls cellular proteostasis and signaling, removing damaged or aged proteins that might otherwise accumulate and affect cell viability. In response to specific needs or changed extracellular microenvironment, the UPS modulates the turnover of mitochondrial proteins, thus influencing the organelle shape, dynamics and function. Alterations of the dynamic and reciprocal regulation between mitochondria and UPS underpin genetic and proliferative disorders. This review focuses on the mitochondrial metabolism and activities supervised by UPS and examines how deregulation of this control mechanism results in proliferative disorders and cancer.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, 80131 Naples, Italy
| |
Collapse
|
12
|
Zhang WW, Li XL, Liu YL, Liu JY, Zhu XX, Li J, Zhao LL, Zhang C, Wang H, Xu DX, Gao L. 1-Nitropyrene disrupts testosterone biogenesis via AKAP1 degradation promoted mitochondrial fission in mouse Leydig cell. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119484. [PMID: 35613681 DOI: 10.1016/j.envpol.2022.119484] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
Previous study found 1-NP disrupted steroidogenesis in mouse testis, but the underlying mechanism remained elusive. The current work aims to explore the roles of ROS-promoted AKAP1 degradation and excessive mitochondrial fission in 1-NP-induced steroidogenesis disruption in MLTC-1 cells. Transmission electron microscope analysis found 1-NP promoted excessive mitochondrial fission. Further data showed 1-NP disrupted mitochondrial function. pDRP1 (Ser637), a negative regulator of mitochondrial fission, was reduced in 1-NP-treated MLTC-1 cells. Mechanistically, 1-NP caused degradation of AKAP1, an upstream regulator of pDRP1 (Ser637). MG132, a proteasome inhibitor, attenuated 1-NP-induced AKAP1 degradation and downstream pDRP1 (Ser637) reduction, thereby ameliorating 1-NP-downregulated steroidogenesis. Further analysis found that cellular ROS was elevated and NOX4, HO-1 and SOD2 were upregulated in 1-NP-exposed MLTC-1 cells. NAC, a well-known commercial antioxidant, alleviated 1-NP-induced excessive ROS and oxidative stress. 1-NP-induced AKAP1 degradation and subsequent downregulation of pDRP1 (Ser637) were prevented by NAC pretreatment. Moreover, NAC attenuated 1-NP-resulted T synthesis disturbance in MLTC-1 cells. The present study indicates that ROS mediated AKAP1 degradation and subsequent pDRP1 (Ser637) dependent mitochondrial fission is indispensable in 1-NP caused T synthesis disruption. This study provides a new insight into 1-NP-induced endocrine disruption, and offers theoretical basis in public health prevention.
Collapse
Affiliation(s)
- Wei-Wei Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Xiu-Liang Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Yu-Lin Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Jia-Yu Liu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Xin-Xin Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Jian Li
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Ling-Li Zhao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Cheng Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - De-Xiang Xu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China
| | - Lan Gao
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes & Department of Toxicology, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui Province, China.
| |
Collapse
|
13
|
Sifat AE, Nozohouri S, Archie SR, Chowdhury EA, Abbruscato TJ. Brain Energy Metabolism in Ischemic Stroke: Effects of Smoking and Diabetes. Int J Mol Sci 2022; 23:ijms23158512. [PMID: 35955647 PMCID: PMC9369264 DOI: 10.3390/ijms23158512] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/06/2023] Open
Abstract
Proper regulation of energy metabolism in the brain is crucial for maintaining brain activity in physiological and different pathophysiological conditions. Ischemic stroke has a complex pathophysiology which includes perturbations in the brain energy metabolism processes which can contribute to worsening of brain injury and stroke outcome. Smoking and diabetes are common risk factors and comorbid conditions for ischemic stroke which have also been associated with disruptions in brain energy metabolism. Simultaneous presence of these conditions may further alter energy metabolism in the brain leading to a poor clinical prognosis after an ischemic stroke event. In this review, we discuss the possible effects of smoking and/or diabetes on brain glucose utilization and mitochondrial energy metabolism which, when present concurrently, may exacerbate energy metabolism in the ischemic brain. More research is needed to investigate brain glucose utilization and mitochondrial oxidative metabolism in ischemic stroke in the presence of smoking and/or diabetes, which would provide further insights on the pathophysiology of these comorbid conditions and facilitate the development of therapeutic interventions.
Collapse
|
14
|
Mitochondrial a Kinase Anchor Proteins in Cardiovascular Health and Disease: A Review Article on Behalf of the Working Group on Cellular and Molecular Biology of the Heart of the Italian Society of Cardiology. Int J Mol Sci 2022; 23:ijms23147691. [PMID: 35887048 PMCID: PMC9322728 DOI: 10.3390/ijms23147691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Second messenger cyclic adenosine monophosphate (cAMP) has been found to regulate multiple mitochondrial functions, including respiration, dynamics, reactive oxygen species production, cell survival and death through the activation of cAMP-dependent protein kinase A (PKA) and other effectors. Several members of the large family of A kinase anchor proteins (AKAPs) have been previously shown to locally amplify cAMP/PKA signaling to mitochondria, promoting the assembly of signalosomes, regulating multiple cardiac functions under both physiological and pathological conditions. In this review, we will discuss roles and regulation of major mitochondria-targeted AKAPs, along with opportunities and challenges to modulate their functions for translational purposes in the cardiovascular system.
Collapse
|
15
|
Dixit P, Suratkal SS, Kokate SB, Chakraborty D, Poirah I, Samal S, Rout N, Singh SP, Sarkar A, Bhattacharyya A. Siah2-GRP78 interaction regulates ROS and provides a proliferative advantage to Helicobacter pylori-infected gastric epithelial cancer cells. Cell Mol Life Sci 2022; 79:414. [PMID: 35816252 PMCID: PMC11072387 DOI: 10.1007/s00018-022-04437-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Helicobacter pylori-mediated gastric carcinogenesis involves upregulation of the E3 ubiquitin ligase Siah2 and its phosphorylation-mediated stabilization. This study elucidates a novel mechanism of oxidative stress regulation by phosphorylated Siah2 in H. pylori-infected gastric epithelial cancer cells (GECs). We identify that H. pylori-mediated Siah2 phosphorylation at the 6th serine residue (P-S6-Siah2) enhances proteasomal degradation of the 78-kDa glucose-regulated protein (GRP78) possessing antioxidant functions. S6 phosphorylation stabilizes Siah2 and P-S6-Siah2 potentiates H. pylori-mediated reactive oxygen species (ROS) generation. However, infected S6A phospho-null Siah2-expressing cells have decreased cellular GRP78 level as surprisingly these cells release GRP78 to a higher extent and accumulate significantly higher ROS than the wild type (WT) Siah2 construct-expressing cells. Ectopic expression of GRP78 prevents the loss of mitochondrial membrane potential and cellular ROS accumulation caused by H. pylori. H. pylori-induced mitochondrial damage and mitochondrial membrane potential loss are potentiated in Siah2-overexpressing cells but these effects are further enhanced in S6A-expressing cells. This study also confirms that while phosphorylation-mediated Siah2 stabilization optimally upregulates aggresome accumulation, it suppresses autophagosome formation, thus decreasing the dependency on the latter mechanism in regulating cellular protein abundance. Disruption of the phospho-Siah2-mediated aggresome formation impairs proliferation of infected GECs. Thus, Siah2 phosphorylation has diagnostic and therapeutic significance in H. pylori-mediated gastric cancer (GC).
Collapse
Affiliation(s)
- Pragyesh Dixit
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
| | - Swathi Shivaram Suratkal
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Shrikant Babanrao Kokate
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
- HiLIFE Institute of Biotechnology, University of Helsinki, PO Box 56, 00014, Helsinki, Finland
| | - Debashish Chakraborty
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
| | - Indrajit Poirah
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
| | - Supriya Samal
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India
| | - Niranjan Rout
- Department of Pathology, Acharya Harihar Post Graduate Institute of Cancer, Cuttack, Odisha, 753007, India
| | - Shivaram P Singh
- Department of Gastroenterology, SCB Medical College, Cuttack, Odisha, 753007, India
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, 751024, India
| | - Asima Bhattacharyya
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, An OCC of Homi Bhabha National Institute, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, Jatni, Odisha, 752050, India.
| |
Collapse
|
16
|
The Role of Mitochondrial Dynamin in Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2504798. [PMID: 35571256 PMCID: PMC9106451 DOI: 10.1155/2022/2504798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/17/2022] [Indexed: 11/25/2022]
Abstract
Stroke is one of the leading causes of death and disability in the world. However, the pathophysiological process of stroke is still not fully clarified. Mitochondria play an important role in promoting nerve survival and are an important drug target for the treatment of stroke. Mitochondrial dysfunction is one of the hallmarks of stroke. Mitochondria are in a state of continuous fission and fusion, which are termed as mitochondrial dynamics. Mitochondrial dynamics are very important for maintaining various functions of mitochondria. In this review, we will introduce the structure and functions of mitochondrial fission and fusion related proteins and discuss their role in the pathophysiologic process of stroke. A better understanding of mitochondrial dynamin in stroke will pave way for the development of new therapeutic options.
Collapse
|
17
|
Sidramagowda Patil S, Soundararajan R, Fukumoto J, Breitzig M, Hernández-Cuervo H, Alleyn M, Lin M, Narala VR, Lockey R, Kolliputi N, Galam L. Mitochondrial Protein Akap1 Deletion Exacerbates Endoplasmic Reticulum Stress in Mice Exposed to Hyperoxia. Front Pharmacol 2022; 13:762840. [PMID: 35370705 PMCID: PMC8964370 DOI: 10.3389/fphar.2022.762840] [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] [Received: 08/23/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Acute lung injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), are treated with high concentrations of supplementary oxygen. However, prolonged exposure to high oxygen concentrations stimulates the production of reactive oxygen species (ROS), which damages the mitochondria and accumulates misfolded proteins in the endoplasmic reticulum (ER). The mitochondrial protein A-kinase anchoring protein 1 (Akap1) is critical for mitochondrial homeostasis. It is known that Akap1 deficiency results in heart damage, neuronal development impairment, and mitochondrial malfunction in preclinical studies. Our laboratory recently revealed that deleting Akap1 increases the severity of hyperoxia-induced ALI in mice. To assess the role of Akap1 deletion in ER stress in lung injury, wild-type and Akap1−/− mice were exposed to hyperoxia for 48 h. This study indicates that Akap1−/− mice exposed to hyperoxia undergo ER stress, which is associated with an increased expression of BiP, JNK phosphorylation, eIF2α phosphorylation, ER stress-induced cell death, and autophagy. This work demonstrates that deleting Akap1 results in increased ER stress in the lungs of mice and that hyperoxia exacerbates ER stress-related consequences.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Ramani Soundararajan
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Jutaro Fukumoto
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Mason Breitzig
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,Washington University in St. Louis, Brown School, St. Louis, MO, United States
| | - Helena Hernández-Cuervo
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Matthew Alleyn
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Muling Lin
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | | | - Richard Lockey
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Narasaiah Kolliputi
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Lakshmi Galam
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| |
Collapse
|
18
|
Jaroslawska J, Gospodarska E, Korytko A. Increasing energy expenditure through exercise and low ambient temperature offers oxidative protection to the hypothalamus after high-fat feeding to mice. J Neuroendocrinol 2022; 34:e13095. [PMID: 35138671 DOI: 10.1111/jne.13095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 11/30/2022]
Abstract
The effects of weight loss produced by increased energy expenditure on measures of oxidative stress and mitochondrial damage have not been investigated in the hypothalamus of diet-induced obese mice. The present study aimed to characterize the effects of either a low housing temperature of 17°C or daily exercise on a treadmill on high-fat diet (HFD)-induced abnormalities in the hypothalamic tissue of mice. Exercise and low ambient temperature protocols were designed to produce energy deficit through increased energy expenditure. Forty mice aged 8 weeks were assigned to one of four conditions: chow diet (n = 10), HFD (n = 10), HFD and 5 weeks of either exercise training (ET; n = 10) or an ambient temperature of 17°C (n = 10). Mice were killed at the age of 31 weeks. In comparison with HFD treatment alone, both interventions reduced body adiposity (14.6% and 27.6% reduction for the ET and 17°C groups, respectively). Moreover, exposing obese mice to ET and 17°C restored mitochondrial DNA content (41.3% and 32.6% increase for the ET and 17°C groups, respectively), decreased level of lipid peroxidation as assessed by the detection of 4-hydroxy-nonenal protein adducts (12.8% and 29.4% reduction for the ET and 17°C groups, respectively) and normalized the expression levels of proinflammatory cytokines (Tnfα: 73.9% and 62%; Il1β: 54.5% and 39.6%; Il6: 33.1% and 35.6% reduction for the ET and 17°C groups, respectively), as well as several proteins associated with mitochondrial respiratory chain (OxPhos Complex I: 75.7% and 53.9%; Complex III: 33% and 36%; Complex V: 42% and 36.9% reduction for the ET and 17°C groups, respectively) in hypothalamic cells. Negative energy balance induced through either lower ambient temperature or exercise resulted in substantial and similar improvements in markers of inflammation and mitochondrial damage in the hypothalamus of mice with diet-induced obesity, potentially by reducing oxidative stress.
Collapse
Affiliation(s)
- Julia Jaroslawska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Emilia Gospodarska
- Department of Biological Functions of Food, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Agnieszka Korytko
- Collegium Medicum, Department of Physiology and Pathophysiology, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
19
|
Nguyen V, Ameri K, Huynh K, Fredkin M, Grona R, Larpthaveesarp A, Gonzalez F, Yeghiazarians Y. Interleukin-15 modulates the response of cortical neurons to ischemia. Mol Cell Neurosci 2021; 115:103658. [PMID: 34343628 DOI: 10.1016/j.mcn.2021.103658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Stroke is a major cause of death and disability in the United States. Current acute stroke therapy consists of clot-dissolving drugs, catheter-based interventions and physical rehabilitation. To date, there are no therapies that directly enhance neuronal survival after a stroke. Previous work from our lab demonstrated that Interleukin-15 (IL-15) peptide could rescue cardiomyocytes subjected to hypoxia. We sought to extend these findings to cortical neurons since IL-15 has been implicated to have an important role in neuronal homeostasis. METHODS We have evaluated the effect of IL-15 peptide on primary cortical neurons derived from embryonic rats in vitro under conditions of anoxia and glucose deprivation, and in vivo following middle cerebral artery occlusion. RESULTS IL-15 administration rescued neuronal cells subjected to anoxia coupled with glucose deprivation (AGD), as well as with reoxygenation. A hallmark of stroke is the ischemic microenvironment and associated oxidative stress, which results in DNA damage and ER stress, both of which contribute to neuronal cell damage and death. The expression of anoxia, ER stress, and DNA damage factors/markers was evaluated via western blot and correlated with the cellular survival effects of IL-15 in vitro. In addition, IL-15 effect of alleviating ER stress and increasing cell survival was also observed in vivo. INTERPRETATION Our data indicate, for the first time, that administration of the pleiotropic factor IL-15 reduces neuronal cell death during AGD, which correlates with modulation of multiple cellular stress pathways.
Collapse
Affiliation(s)
- Vien Nguyen
- Department of Medicine, University of California San Francisco (UCSF), USA.
| | - Kurosh Ameri
- Department of Medicine, University of California San Francisco (UCSF), USA
| | - Kevin Huynh
- Department of Medicine, University of California San Francisco (UCSF), USA
| | - Maxwell Fredkin
- Department of Medicine, University of California San Francisco (UCSF), USA
| | - Reinier Grona
- Department of Medicine, University of California San Francisco (UCSF), USA
| | | | | | - Yerem Yeghiazarians
- Department of Medicine, University of California San Francisco (UCSF), USA; Helen Diller Family Comprehensive Cancer Center, UCSF, USA
| |
Collapse
|
20
|
Ross JA, Vissers JPC, Nanda J, Stewart GD, Husi H, Habib FK, Hammond DE, Gethings LA. The influence of hypoxia on the prostate cancer proteome. Clin Chem Lab Med 2021; 58:980-993. [PMID: 31940282 DOI: 10.1515/cclm-2019-0626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022]
Abstract
Prostate cancer accounts for around 15% of male deaths in Western Europe and is the second leading cause of cancer death in men after lung cancer. Mounting evidence suggests that prostate cancer deposits exist within a hypoxic environment and this contributes to radio-resistance thus hampering one of the major therapies for this cancer. Recent reports have shown that nitric oxide (NO) donating non-steroidal anti-inflammatory drugs (NSAIDs) reduced tumour hypoxia as well as maintaining a radio-sensitising/therapeutic effect on prostate cancer cells. The aim of this study was to evaluate the impact of hypoxia on the proteome of the prostate and to establish whether NO-NSAID treatment reverted the protein profiles back to their normoxic status. To this end an established hormone insensitive prostate cancer cell line, PC-3, was cultured under hypoxic and normoxic conditions before and following exposure to NO-NSAID in combination with selected other common prostate cancer treatment types. The extracted proteins were analysed by ion mobility-assisted data independent acquisition mass spectrometry (MS), combined with multivariate statistical analyses, to measure hypoxia-induced alterations in the proteome of these cells. The analyses demonstrated that under hypoxic conditions there were well-defined, significantly regulated/differentially expressed proteins primarily involved with structural and binding processes including, for example, TUBB4A, CIRP and PLOD1. Additionally, the exposure of hypoxic cells to NSAID and NO-NSAID agents, resulted in some of these proteins being differentially expressed; for example, both PCNA and HNRNPA1L were down-regulated, corresponding with disruption in the nucleocytoplasmic shuttling process.
Collapse
Affiliation(s)
- James A Ross
- Tissue Injury and Repair Group, University of Edinburgh, Edinburgh, UK
| | | | - Jyoti Nanda
- Tissue Injury and Repair Group, University of Edinburgh, Edinburgh, UK.,Prostate Research Group, University of Edinburgh, Edinburgh, UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Fouad K Habib
- Tissue Injury and Repair Group, University of Edinburgh, Edinburgh, UK.,Prostate Research Group, University of Edinburgh, Edinburgh, UK
| | - Dean E Hammond
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Lee A Gethings
- Waters Corporation, Wilmslow, UK.,Manchester Institute of Biotechnology, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Piedade WP, Famulski JK. E3 ubiquitin ligase-mediated regulation of vertebrate ocular development; new insights into the function of SIAH enzymes. Biochem Soc Trans 2021; 49:327-340. [PMID: 33616626 PMCID: PMC7924998 DOI: 10.1042/bst20200613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/18/2023]
Abstract
Developmental regulation of the vertebrate visual system has been a focus of investigation for generations as understanding this critical time period has direct implications on our understanding of congenital blinding disease. The majority of studies to date have focused on transcriptional regulation mediated by morphogen gradients and signaling pathways. However, recent studies of post translational regulation during ocular development have shed light on the role of the ubiquitin proteasome system (UPS). This rather ubiquitous yet highly diverse system is well known for regulating protein function and localization as well as stability via targeting for degradation by the 26S proteasome. Work from many model organisms has recently identified UPS activity during various milestones of ocular development including retinal morphogenesis, retinal ganglion cell function as well as photoreceptor homeostasis. In particular work from flies and zebrafish has highlighted the role of the E3 ligase enzyme family, Seven in Absentia Homologue (Siah) during these events. In this review, we summarize the current understanding of UPS activity during Drosophila and vertebrate ocular development, with a major focus on recent findings correlating Siah E3 ligase activity with two major developmental stages of vertebrate ocular development, retinal morphogenesis and photoreceptor specification and survival.
Collapse
|
22
|
Onyango IG, Bennett JP, Stokin GB. Regulation of neuronal bioenergetics as a therapeutic strategy in neurodegenerative diseases. Neural Regen Res 2021; 16:1467-1482. [PMID: 33433460 PMCID: PMC8323696 DOI: 10.4103/1673-5374.303007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are a heterogeneous group of debilitating disorders with multifactorial etiologies and pathogeneses that manifest distinct molecular mechanisms and clinical manifestations with abnormal protein dynamics and impaired bioenergetics. Mitochondrial dysfunction is emerging as an important feature in the etiopathogenesis of these age-related neurodegenerative diseases. The prevalence and incidence of these diseases is on the rise with the increasing global population and average lifespan. Although many therapeutic approaches have been tested, there are currently no effective treatment routes for the prevention or cure of these diseases. We present the current status of our knowledge and understanding of the involvement of mitochondrial dysfunction in these diseases and highlight recent advances in novel therapeutic strategies targeting neuronal bioenergetics as potential approach for treating these diseases.
Collapse
Affiliation(s)
- Isaac G Onyango
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| | - James P Bennett
- Neurodegeneration Therapeutics, 3050A Berkmar Drive, Charlottesville, VA, USA
| | - Gorazd B Stokin
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
23
|
Marin W, Marin D, Ao X, Liu Y. Mitochondria as a therapeutic target for cardiac ischemia‑reperfusion injury (Review). Int J Mol Med 2020; 47:485-499. [PMID: 33416090 PMCID: PMC7797474 DOI: 10.3892/ijmm.2020.4823] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
Acute myocardial infarction is the leading cause of cardiovascular-related mortality and chronic heart failure worldwide. As regards treatment, the reperfusion of ischemic tissue generates irreversible damage to the myocardium, which is termed 'cardiac ischemia-reperfusion (IR) injury'. Due to the large number of mitochondria in cardiomyocytes, an increasing number of studies have focused on the roles of mitochondria in IR injury. The primary causes of IR injury are reduced oxidative phosphorylation during hypoxia and the increased production of reactive oxygen species (ROS), together with the insufficient elimination of these oxidative species following reperfusion. IR injury includes the oxidation of DNA, incorrect modifications of proteins, the disruption of the mitochondrial membrane and respiratory chain, the loss of mitochondrial membrane potential (∆Ψm), Ca2+ over-load, mitochondrial permeability transition pore formation, swelling of the mitochondria, and ultimately, cardiomyocyte necrosis. The present review article discusses the molecular mechanisms of IR injury, and summarizes the metabolic and dynamic changes occurring in the mitochondria in response to IR stress. The mitochondria are strongly recommended as a target for the development of therapeutic agents; however, the appropriate use of agents remains a challenge.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Dennis Marin
- Qingdao University of Science and Technology, Qingdao, Shandong 266061, P.R. China
| | - Xiang Ao
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
24
|
Du Q, Han J, Gao S, Zhang S, Pan Y. Hypoxia-induced circular RNA hsa_circ_0008450 accelerates hepatocellular cancer progression via the miR-431/AKAP1 axis. Oncol Lett 2020; 20:388. [PMID: 33193848 PMCID: PMC7656113 DOI: 10.3892/ol.2020.12251] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Hypoxia facilitates the progression of numerous cancers. Circular RNAs (circRNA) have been revealed to be involved in the process of tumors mediated by hypoxia. However, the role and molecular mechanism of circular RNA hsa_circ_0008450 (circ_0008450) in hepatocellular cancer (HCC) under hypoxic conditions has been rarely reported. Expression levels of circ_0008450, microRNA(miR)-431 and A-kinase anchor protein 1 (AKAP1) were examined using reverse transcription-quantitative PCR. Cell viability, apoptosis and glycolysis were assessed via Cell Counting Kit-8, flow cytometry and glycolysis assays, respectively. The association between circ_0008450 or AKAP1 and miR-431 was verified via dual-luciferase reporter assays. Protein levels of AKAP1 were detected by western blotting. Effect of hsa_circ_0008450 on tumor growth in vivo was confirmed by xenograft assays. Circ_0008450 was upregulated in HCC tissues and hypoxia-disposed HCC cells. Depletion of circ_0008450 suppressed tumor growth in vivo and reversed the repression of apoptosis and the acceleration of viability and glycolysis of HCC cells induced by hypoxia treatment in vitro. Notably, circ_0008450 regulated AKAP1 expression by sponging miR-431. Furthermore, miR-431 inhibition reversed the circ_0008450 silencing-mediated effects on viability, apoptosis and glycolysis in hypoxia-treated HCC cells. Additionally, AKAP1 enhancement abolished the effects of miR-431 upregulation on the viability, apoptosis and glycolysis in hypoxia-treated HCC cells. In conclusion, circ_0008450 repression mitigated the progression of HCC under hypoxia by downregulating AKAP1 via miR-431, providing a potential target for HCC treatment.
Collapse
Affiliation(s)
- Qiajun Du
- Department of Clinical Laboratory Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Jie Han
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Shan Gao
- Department of Clinical Laboratory Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Shangdi Zhang
- Department of Clinical Laboratory Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Yunyan Pan
- Department of Clinical Laboratory Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
25
|
Scorziello A, Borzacchiello D, Sisalli MJ, Di Martino R, Morelli M, Feliciello A. Mitochondrial Homeostasis and Signaling in Parkinson's Disease. Front Aging Neurosci 2020; 12:100. [PMID: 32372945 PMCID: PMC7186467 DOI: 10.3389/fnagi.2020.00100] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
The loss of dopaminergic (DA) neurons in the substantia nigra leads to a progressive, long-term decline of movement and other non-motor deficits. The symptoms of Parkinson's disease (PD) often appear later in the course of the disease, when most of the functional dopaminergic neurons have been lost. The late onset of the disease, the severity of the illness, and its impact on the global health system demand earlier diagnosis and better targeted therapy. PD etiology and pathogenesis are largely unknown. There are mutations in genes that have been linked to PD and, from these complex phenotypes, mitochondrial dysfunction emerged as central in the pathogenesis and evolution of PD. In fact, several PD-associated genes negatively impact on mitochondria physiology, supporting the notion that dysregulation of mitochondrial signaling and homeostasis is pathogenically relevant. Derangement of mitochondrial homeostatic controls can lead to oxidative stress and neuronal cell death. Restoring deranged signaling cascades to and from mitochondria in PD neurons may then represent a viable opportunity to reset energy metabolism and delay the death of dopaminergic neurons. Here, we will highlight the relevance of dysfunctional mitochondrial homeostasis and signaling in PD, the molecular mechanisms involved, and potential therapeutic approaches to restore mitochondrial activities in damaged neurons.
Collapse
Affiliation(s)
- Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maria Jose Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Rossana Di Martino
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, University of Naples Federico II, Naples, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, University of Cagliari, Cagliari, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Sisalli MJ, Ianniello G, Savoia C, Cuomo O, Annunziato L, Scorziello A. Knocking-out the Siah2 E3 ubiquitin ligase prevents mitochondrial NCX3 degradation, regulates mitochondrial fission and fusion, and restores mitochondrial function in hypoxic neurons. Cell Commun Signal 2020; 18:42. [PMID: 32164721 PMCID: PMC7066748 DOI: 10.1186/s12964-020-0529-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 02/05/2020] [Indexed: 11/10/2022] Open
Abstract
Background Na+/Ca2+ exchanger isoform 3 (NCX3) regulates mitochondrial Ca2+ handling through the outer mitochondrial membrane (OMM) and promotes neuronal survival during oxygen and glucose deprivation (OGD). Conversely, Seven In-Absentia Homolog 2 (Siah2), an E3-ubiquitin ligase, which is activated under hypoxic conditions, causes proteolysis of mitochondrial and cellular proteins. In the present study, we investigated whether siah2, upon its activation during hypoxia, interacts with NCX3 and whether such interaction could regulate the molecular events underlying changes in mitochondrial morphology, i.e., fusion and fission, and function, in neurons exposed to anoxia and anoxia/reoxygenation. Methods To answer these questions, after exposing cortical neurons from siah2 KO mice (siah2 −/−) to OGD and OGD/Reoxygenation, we monitored the changes in mitochondrial fusion and fission protein expression, mitochondrial membrane potential (ΔΨm), and mitochondrial calcium concentration ([Ca2+]m) by using specific fluorescent probes, confocal microscopy, and Western Blot analysis. Results As opposed to congenic wild-type neurons, in neurons from siah2−/− mice exposed to OGD, form factor (FF), an index of the complexity and branching aspect of mitochondria, and aspect ratio (AR), an index reflecting the “length-to-width ratio” of mitochondria, maintained low expression. In KO siah2 neurons exposed to OGD, downregulation of mitofusin 1 (Mfn1), a protein involved in mitochondrial fusion and upregulation of dynamin-related protein 1 (Drp1), a protein involved in the mitochondrial fission, were prevented. Furthermore, under OGD conditions, whereas [Ca2+]m was reduced, ΔΨm, mitochondrial oxidative capacity and ATP production were improved. Interestingly, our immunoprecipitation assay revealed that Siah2 interacted with NCX3. Indeed, siah2 knock-out prevented NCX3 degradation in neurons exposed to OGD. Finally, when siah2−/− neurons were exposed to OGD/reoxygenation, FF, AR, and Mfn1 expression increased, and mitochondrial function improved compared to siah2+/+ neurons. Conclusions Collectively, these findings indicate that hypoxia-induced SIAH2-E3 ligase activation influences mitochondrial fusion and fission, as well as function, by inducing NCX3 degradation. Video Abstract
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Gaetano Ianniello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Claudia Savoia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via S. Pansini 5, 80131, Naples, Italy
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
27
|
Sisalli MJ, Feliciello A, Della Notte S, Di Martino R, Borzacchiello D, Annunziato L, Scorziello A. Nuclear-encoded NCX3 and AKAP121: Two novel modulators of mitochondrial calcium efflux in normoxic and hypoxic neurons. Cell Calcium 2020; 87:102193. [PMID: 32193001 DOI: 10.1016/j.ceca.2020.102193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/30/2022]
Abstract
Mitochondria are highly dynamic organelles extremely important for cell survival. Their structure resembles that of prokaryotic cells since they are composed with two membranes, the inner (IMM) and the outer mitochondrial membrane (OMM) delimitating the intermembrane space (IMS) and the matrix which contains mitochondrial DNA (mtDNA). This structure is strictly related to mitochondrial function since they produce the most of the cellular ATP through the oxidative phosphorylation which generate the electrochemical gradient at the two sides of the inner mitochondrial membrane an essential requirement for mitochondrial function. Cells of highly metabolic demand like those composing muscle, liver and brain, are particularly dependent on mitochondria for their activities. Mitochondria undergo to continual changes in morphology since, they fuse and divide, branch and fragment, swell and extend. Importantly, they move throughout the cell to deliver ATP and other metabolites where they are mostly required. Along with the capability to control energy metabolism, mitochondria play a critical role in the regulation of many physiological processes such as programmed cell death, autophagy, redox signalling, and stem cells reprogramming. All these phenomena are regulated by Ca2+ ions within this organelle. This review will discuss the molecular mechanisms regulating mitochondrial calcium cycling in physiological and pathological conditions with particular regard to their impact on mitochondrial dynamics and function during ischemia. Particular emphasis will be devoted to the role played by NCX3 and AKAP121 as new molecular targets for mitochondrial function and dysfunction.
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Italy
| | - Salvatore Della Notte
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Italy
| | - Rossana Di Martino
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Italy
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Italy.
| |
Collapse
|
28
|
Lucia K, Wu Y, Garcia JM, Barlier A, Buchfelder M, Saeger W, Renner U, Stalla GK, Theodoropoulou M. Hypoxia and the hypoxia inducible factor 1α activate protein kinase A by repressing RII beta subunit transcription. Oncogene 2020; 39:3367-3380. [PMID: 32111982 PMCID: PMC7160059 DOI: 10.1038/s41388-020-1223-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022]
Abstract
Overactivation of the cAMP signal transduction pathway plays a central role in the pathogenesis of endocrine tumors. Genetic aberrations leading to increased intracellular cAMP or directly affecting PKA subunit expression have been identified in inherited and sporadic endocrine tumors, but are rare indicating the presence of nongenomic pathological PKA activation. In the present study, we examined the impact of hypoxia on PKA activation using human growth hormone (GH)-secreting pituitary tumors as a model of an endocrine disease displaying PKA-CREB overactivation. We show that hypoxia activates PKA and enhances CREB transcriptional activity and subsequently GH oversecretion. This is due to a previously uncharacterized ability of HIF-1α to suppress the transcription of the PKA regulatory subunit 2B (PRKAR2B) by sequestering Sp1 from the PRKAR2B promoter. The present study reveals a novel mechanism through which the transcription factor HIF-1α transduces environmental signals directly onto PKA activity, without affecting intracellular cAMP concentrations. By identifying a point of interaction between the cellular microenvironment and intracellular enzyme activation, neoplastic, and nonneoplastic diseases involving overactivated PKA pathway may be more efficiently targeted.
Collapse
Affiliation(s)
- Kristin Lucia
- Department of Endocrinology, Max Planck Institute of Psychiatry, Munich, Germany.,Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Neurosurgery, Charité-Universitätsmedizin, Berlin, Germany.,Division of Molecular Genetics, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Yonghe Wu
- Division of Molecular Genetics, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | - Anne Barlier
- Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Marseille, France
| | - Michael Buchfelder
- Department of Neurosurgery, Klinikum der Universität Erlangen, Erlangen, Germany
| | - Wolfgang Saeger
- Department of Neuropathology, Universität Hamburg, Hamburg, Germany
| | - Ulrich Renner
- Department of Endocrinology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Günter K Stalla
- Department of Endocrinology, Max Planck Institute of Psychiatry, Munich, Germany.,Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marily Theodoropoulou
- Department of Endocrinology, Max Planck Institute of Psychiatry, Munich, Germany. .,Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
29
|
Chen Z, Ma Y, Yang Q, Hu J, Feng J, Liang W, Ding G. AKAP1 mediates high glucose-induced mitochondrial fission through the phosphorylation of Drp1 in podocytes. J Cell Physiol 2020; 235:7433-7448. [PMID: 32108342 DOI: 10.1002/jcp.29646] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that mitochondrial dysfunction plays a critical role in the development of diabetic kidney disease (DKD), however, its specific pathomechanism remains unclear. A-kinase anchoring protein (AKAP) 1 is a scaffold protein in the AKAP family that is involved in mitochondrial fission and fusion. Here, we show that rats with streptozotocin (STZ)-induced diabetes developed podocyte damage accompanied by AKAP1 overexpression and that AKAP1 closely interacted with the mitochondrial fission enzyme dynamin-related protein 1 (Drp1). At the molecular level, high glucose (HG) promoted podocyte injury and Drp1 phosphorylation at Ser637 as proven by decreased mitochondrial membrane potential, elevated reactive oxygen species generation, reduced adenosine triphosphate synthesis, and increased podocyte apoptosis. Furthermore, the AKAP1 knockdown protected HG-induced podocyte injury and suppressed HG-induced Drp1 phosphorylation at Ser637. AKAP1 overexpression aggravated HG-induced mitochondrial fragmentation and podocyte apoptosis. The coimmunoprecipitation assay showed that HG-induced Drp1 interacted with AKAP1, revealing that AKAP1 could recruit Drp1 from the cytoplasm under HG stimulation. Subsequently, we detected the effect of drp1 phosphorylation on Ser637 by transferring several different Drp1 mutants. We demonstrated that activated AKAP1 promoted Drp1 phosphorylation at Ser637, which promoted the transposition of Drp1 to the surface of the mitochondria and accounts for mitochondrial dysfunction events. These findings indicate that AKAP1 is the main pathogenic factor in the development and progression of HG-induced podocyte injury through the destruction of mitochondrial dynamic homeostasis by regulating Drp1 phosphorylation in human podocytes.
Collapse
Affiliation(s)
- Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqiong Ma
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
30
|
A-Kinase Anchoring Protein 1: Emerging Roles in Regulating Mitochondrial Form and Function in Health and Disease. Cells 2020; 9:cells9020298. [PMID: 31991888 PMCID: PMC7072574 DOI: 10.3390/cells9020298] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023] Open
Abstract
Best known as the powerhouse of the cell, mitochondria have many other important functions such as buffering intracellular calcium and reactive oxygen species levels, initiating apoptosis and supporting cell proliferation and survival. Mitochondria are also dynamic organelles that are constantly undergoing fission and fusion to meet specific functional needs. These processes and functions are regulated by intracellular signaling at the mitochondria. A-kinase anchoring protein 1 (AKAP1) is a scaffold protein that recruits protein kinase A (PKA), other signaling proteins, as well as RNA to the outer mitochondrial membrane. Hence, AKAP1 can be considered a mitochondrial signaling hub. In this review, we discuss what is currently known about AKAP1's function in health and diseases. We focus on the recent literature on AKAP1's roles in metabolic homeostasis, cancer and cardiovascular and neurodegenerative diseases. In healthy tissues, AKAP1 has been shown to be important for driving mitochondrial respiration during exercise and for mitochondrial DNA replication and quality control. Several recent in vivo studies using AKAP1 knockout mice have elucidated the role of AKAP1 in supporting cardiovascular, lung and neuronal cell survival in the stressful post-ischemic environment. In addition, we discuss the unique involvement of AKAP1 in cancer tumor growth, metastasis and resistance to chemotherapy. Collectively, the data indicate that AKAP1 promotes cell survival throug regulating mitochondrial form and function. Lastly, we discuss the potential of targeting of AKAP1 for therapy of various disorders.
Collapse
|
31
|
Haushalter KJ, Schilling JM, Song Y, Sastri M, Perkins GA, Strack S, Taylor SS, Patel HH. Cardiac ischemia-reperfusion injury induces ROS-dependent loss of PKA regulatory subunit RIα. Am J Physiol Heart Circ Physiol 2019; 317:H1231-H1242. [PMID: 31674811 PMCID: PMC6962616 DOI: 10.1152/ajpheart.00237.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Type I PKA regulatory α-subunit (RIα; encoded by the Prkar1a gene) serves as the predominant inhibitor protein of the catalytic subunit of cAMP-dependent protein kinase (PKAc). However, recent evidence suggests that PKA signaling can be initiated by cAMP-independent events, especially within the context of cellular oxidative stress such as ischemia-reperfusion (I/R) injury. We determined whether RIα is actively involved in the regulation of PKA activity via reactive oxygen species (ROS)-dependent mechanisms during I/R stress in the heart. Induction of ex vivo global I/R injury in mouse hearts selectively downregulated RIα protein expression, whereas RII subunit expression appears to remain unaltered. Cardiac myocyte cell culture models were used to determine that oxidant stimulus (i.e., H2O2) alone is sufficient to induce RIα protein downregulation. Transient increase of RIα expression (via adenoviral overexpression) negatively affects cell survival and function upon oxidative stress as measured by increased induction of apoptosis and decreased mitochondrial respiration. Furthermore, analysis of mitochondrial subcellular fractions in heart tissue showed that PKA-associated proteins are enriched in subsarcolemmal mitochondria (SSM) fractions and that loss of RIα is most pronounced at SSM upon I/R injury. These data were supported via electron microscopy in A-kinase anchoring protein 1 (AKAP1)-knockout mice, where loss of AKAP1 expression leads to aberrant mitochondrial morphology manifested in SSM but not interfibrillar mitochondria. Thus, we conclude that modification of RIα via ROS-dependent mechanisms induced by I/R injury has the potential to sensitize PKA signaling in the cell without the direct use of the canonical cAMP-dependent activation pathway.NEW & NOTEWORTHY We uncovered a previously undescribed phenomenon involving oxidation-induced activation of PKA signaling in the progression of cardiac ischemia-reperfusion injury. Type I PKA regulatory subunit RIα, but not type II PKA regulatory subunits, is dynamically regulated by oxidative stress to trigger the activation of the catalytic subunit of PKA in cardiac myocytes. This effect may play a critical role in the regulation of subsarcolemmal mitochondria function upon the induction of ischemic injury in the heart.
Collapse
Affiliation(s)
- Kristofer J Haushalter
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Jan M Schilling
- Veterans Affairs San Diego Healthcare System, San Diego, California
- Department of Anesthesiology, University of California, San Diego, La Jolla, California
| | - Young Song
- Veterans Affairs San Diego Healthcare System, San Diego, California
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Mira Sastri
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, California
| | - Stefan Strack
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Susan S Taylor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, California
- Department of Anesthesiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
32
|
Marin W. A-kinase anchoring protein 1 (AKAP1) and its role in some cardiovascular diseases. J Mol Cell Cardiol 2019; 138:99-109. [PMID: 31783032 DOI: 10.1016/j.yjmcc.2019.11.154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 01/09/2023]
Abstract
A-kinase anchoring proteins (AKAPs) play crucial roles in regulating compartmentalized multi-protein signaling networks related to PKA-mediated phosphorylation. The mitochondrial AKAP - AKAP1 proteins are enriched in heart and play cardiac protective roles. This review aims to thoroughly summarize AKAP1 variants from their sequence features to the structure-function relationships between AKAP1 and its binding partners, as well as the molecular mechanisms of AKAP1 in cardiac hypertrophy, hypoxia-induced myocardial infarction and endothelial cells dysfunction, suggesting AKAP1 as a candidate for cardiovascular therapy.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, Medical Faculty of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
33
|
Lucero M, Suarez AE, Chambers JW. Phosphoregulation on mitochondria: Integration of cell and organelle responses. CNS Neurosci Ther 2019; 25:837-858. [PMID: 31025544 PMCID: PMC6566066 DOI: 10.1111/cns.13141] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are highly integrated organelles that are crucial to cell adaptation and mitigating adverse physiology. Recent studies demonstrate that fundamental signal transduction pathways incorporate mitochondrial substrates into their biological programs. Reversible phosphorylation is emerging as a useful mechanism to modulate mitochondrial function in accordance with cellular changes. Critical serine/threonine protein kinases, such as the c-Jun N-terminal kinase (JNK), protein kinase A (PKA), PTEN-induced kinase-1 (PINK1), and AMP-dependent protein kinase (AMPK), readily translocate to the outer mitochondrial membrane (OMM), the interface of mitochondria-cell communication. OMM protein kinases phosphorylate diverse mitochondrial substrates that have discrete effects on organelle dynamics, protein import, respiratory complex activity, antioxidant capacity, and apoptosis. OMM phosphorylation events can be tempered through the actions of local protein phosphatases, such as mitogen-activated protein kinase phosphatase-1 (MKP-1) and protein phosphatase 2A (PP2A), to regulate the extent and duration of signaling. The central mediators of OMM signal transduction are the scaffold proteins because the relative abundance of these accessory proteins determines the magnitude and duration of a signaling event on the mitochondrial surface, which dictates the biological outcome of a local signal transduction pathway. The concentrations of scaffold proteins, such as A-kinase anchoring proteins (AKAPs) and Sab (or SH3 binding protein 5-SH3BP5), have been shown to influence neuronal survival and vulnerability, respectively, in models of Parkinson's disease (PD), highlighting the importance of OMM signaling to health and disease. Despite recent progress, much remains to be discovered concerning the mechanisms of OMM signaling. Nonetheless, enhancing beneficial OMM signaling events and inhibiting detrimental protein-protein interactions on the mitochondrial surface may represent highly selective approaches to restore mitochondrial health and homeostasis and mitigate organelle dysfunction in conditions such as PD.
Collapse
Affiliation(s)
- Maribel Lucero
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Ana E Suarez
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Jeremy W Chambers
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| |
Collapse
|
34
|
Ma B, Cheng H, Mu C, Geng G, Zhao T, Luo Q, Ma K, Chang R, Liu Q, Gao R, Nie J, Xie J, Han J, Chen L, Ma G, Zhu Y, Chen Q. The SIAH2-NRF1 axis spatially regulates tumor microenvironment remodeling for tumor progression. Nat Commun 2019; 10:1034. [PMID: 30833558 PMCID: PMC6399320 DOI: 10.1038/s41467-019-08618-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023] Open
Abstract
The interactions between tumor cells with their microenvironments, including hypoxia, acidosis and immune cells, lead to the tumor heterogeneity which promotes tumor progression. Here, we show that SIAH2-NRF1 axis remodels tumor microenvironment through regulating tumor mitochondrial function, tumor-associated macrophages (TAMs) polarization and cell death for tumor maintenance and progression. Mechanistically, low mitochondrial gene expression in breast cancers is associated with a poor clinical outcome. The hypoxia-activated E3 ligase SIAH2 spatially downregulates nuclear-encoded mitochondrial gene expression including pyruvate dehydrogenase beta via degrading NRF1 (Nuclear Respiratory Factor 1) through ubiquitination on lysine 230, resulting in enhanced Warburg effect, metabolic reprogramming and pro-tumor immune response. Dampening NRF1 degradation under hypoxia not only impairs the polarization of TAMs, but also promotes tumor cells to become more susceptible to apoptosis in a FADD-dependent fashion, resulting in secondary necrosis due to the impairment of efferocytosis. These data represent that inhibition of NRF1 degradation is a potential therapeutic strategy against cancer.
Collapse
Affiliation(s)
- Biao Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Hongcheng Cheng
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Chenglong Mu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Guangfeng Geng
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tian Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qian Luo
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kaili Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Rui Chang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiangqiang Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ruize Gao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Junli Nie
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jiaying Xie
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jinxue Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Linbo Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Gui Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yushan Zhu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Quan Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, 300071, China. .,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
35
|
Aggarwal S, Gabrovsek L, Langeberg LK, Golkowski M, Ong SE, Smith FD, Scott JD. Depletion of dAKAP1-protein kinase A signaling islands from the outer mitochondrial membrane alters breast cancer cell metabolism and motility. J Biol Chem 2019; 294:3152-3168. [PMID: 30598507 PMCID: PMC6398132 DOI: 10.1074/jbc.ra118.006741] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/21/2018] [Indexed: 01/23/2023] Open
Abstract
Breast cancer screening and new precision therapies have led to improved patient outcomes. Yet, a positive prognosis is less certain when primary tumors metastasize. Metastasis requires a coordinated program of cellular changes that promote increased survival, migration, and energy consumption. These pathways converge on mitochondrial function, where distinct signaling networks of kinases, phosphatases, and metabolic enzymes regulate these processes. The protein kinase A-anchoring protein dAKAP1 compartmentalizes protein kinase A (PKA) and other signaling enzymes at the outer mitochondrial membrane and thereby controls mitochondrial function and dynamics. Modulation of these processes occurs in part through regulation of dynamin-related protein 1 (Drp1). Here, we report an inverse relationship between the expression of dAKAP1 and mesenchymal markers in breast cancer. Molecular, cellular, and in silico analyses of breast cancer cell lines confirmed that dAKAP1 depletion is associated with impaired mitochondrial function and dynamics, as well as with increased glycolytic potential and invasiveness. Furthermore, disruption of dAKAP1-PKA complexes affected cell motility and mitochondrial movement toward the leading edge in invasive breast cancer cells. We therefore propose that depletion of dAKAP1-PKA "signaling islands" from the outer mitochondrial membrane augments progression toward metastatic breast cancer.
Collapse
Affiliation(s)
- Stacey Aggarwal
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Laura Gabrovsek
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Lorene K Langeberg
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Martin Golkowski
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - Shao-En Ong
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - F Donelson Smith
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| | - John D Scott
- From the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195
| |
Collapse
|
36
|
Zhang J, Feng J, Ma D, Wang F, Wang Y, Li C, Wang X, Yin X, Zhang M, Dagda RK, Zhang Y. Neuroprotective Mitochondrial Remodeling by AKAP121/PKA Protects HT22 Cell from Glutamate-Induced Oxidative Stress. Mol Neurobiol 2019; 56:5586-5607. [PMID: 30652267 DOI: 10.1007/s12035-018-1464-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Protein kinase A (PKA) is a ser/thr kinase that is critical for maintaining essential neuronal functions including mitochondrial homeostasis, bioenergetics, neuronal development, and neurotransmission. The endogenous pool of PKA is targeted to the mitochondrion by forming a complex with the mitochondrial scaffold A-kinase anchoring protein 121 (AKAP121). Enhanced PKA signaling via AKAP121 leads to PKA-mediated phosphorylation of the fission modulator Drp1, leading to enhanced mitochondrial networks and thereby blocking apoptosis against different toxic insults. In this study, we show for the first time that AKAP121/PKA confers neuroprotection in an in vitro model of oxidative stress induced by exposure to excess glutamate. Unexpectedly, treating mouse hippocampal progenitor neuronal HT22 cells with an acute dose or chronic exposure of glutamate robustly elevates PKA signaling, a beneficial compensatory response that is phenocopied in HT22 cells conditioned to thrive in the presence of excess glutamate but not in parental HT22 cells. Secondly, redirecting the endogenous pool of PKA by transiently transfecting AKAP121 or transfecting a constitutively active mutant of PKA targeted to the mitochondrion (OMM-PKA) or of an isoform of AKAP121 that lacks the KH and Tudor domains (S-AKAP84) are sufficient to significantly block cell death induced by glutamate toxicity but not in an oxygen deprivation/reperfusion model. Conversely, transient transfection of HT22 neuronal cells with a PKA-binding-deficient mutant of AKAP121 is unable to protect against oxidative stress induced by glutamate toxicity suggesting that the catalytic activity of PKA is required for AKAP121's protective effects. Mechanistically, AKAP121 promotes neuroprotection by enhancing PKA-mediated phosphorylation of Drp1 to increase mitochondrial fusion, elevates ATP levels, and elicits an increase in the levels of antioxidants GSH and superoxide dismutase 2 leading to a reduction in the level of mitochondrial superoxide. Overall, our data supports AKAP121/PKA as a new molecular target that confers neuroprotection against glutamate toxicity by phosphorylating Drp1, to stabilize mitochondrial networks and mitochondrial function and to elicit antioxidant responses.
Collapse
Affiliation(s)
- Jingdian Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Feng Wang
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yumeng Wang
- Department of Physiology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Xinmin Street No. 126, Changchun, 130000, China
| | - Chunxiao Li
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Xiang Yin
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, 126 Xin Min Street, Changchun, 130021, Jilin, China
| | - Ruben K Dagda
- Department of Pharmacology, Reno School of Medicine, University of Nevada, Mailstop 318, Howard Medical Sciences Building 148A (Office), Reno, NV, 89557,, USA
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No. 71, Changchun, 130000, China.
| |
Collapse
|
37
|
AKAP1 Protects from Cerebral Ischemic Stroke by Inhibiting Drp1-Dependent Mitochondrial Fission. J Neurosci 2018; 38:8233-8242. [PMID: 30093535 PMCID: PMC6146498 DOI: 10.1523/jneurosci.0649-18.2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/29/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial fission and fusion impact numerous cellular functions and neurons are particularly sensitive to perturbations in mitochondrial dynamics. Here we describe that male mice lacking the mitochondrial A-kinase anchoring protein 1 (AKAP1) exhibit increased sensitivity in the transient middle cerebral artery occlusion model of focal ischemia. At the ultrastructural level, AKAP1-/- mice have smaller mitochondria and increased contacts between mitochondria and the endoplasmic reticulum in the brain. Mechanistically, deletion of AKAP1 dysregulates complex II of the electron transport chain, increases superoxide production, and impairs Ca2+ homeostasis in neurons subjected to excitotoxic glutamate. Ca2+ deregulation in neurons lacking AKAP1 can be attributed to loss of inhibitory phosphorylation of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) at the protein kinase A (PKA) site Ser637. Our results indicate that inhibition of Drp1-dependent mitochondrial fission by the outer mitochondrial AKAP1/PKA complex protects neurons from ischemic stroke by maintaining respiratory chain activity, inhibiting superoxide production, and delaying Ca2+ deregulation. They also provide the first genetic evidence that Drp1 inhibition may be of therapeutic relevance for the treatment of stroke and neurodegeneration.SIGNIFICANCE STATEMENT Previous work suggests that activation of dynamin-related protein 1 (Drp1) and mitochondrial fission contribute to ischemic injury in the brain. However, the specificity and efficacy of the pharmacological Drp1 inhibitor mdivi-1 that was used has now been discredited by several high-profile studies. Our report is timely and highly impactful because it provides the first evidence that genetic disinhibition of Drp1 via knock-out of the mitochondrial protein kinase A (PKA) scaffold AKAP1 exacerbates stroke injury in mice. Mechanistically, we show that electron transport deficiency, increased superoxide production, and Ca2+ overload result from genetic disinhibition of Drp1. In summary, our work settles current controversies regarding the role of mitochondrial fission in neuronal injury, provides mechanisms, and suggests that fission inhibitors hold promise as future therapeutic agents.
Collapse
|
38
|
Schiattarella GG, Boccella N, Paolillo R, Cattaneo F, Trimarco V, Franzone A, D’Apice S, Giugliano G, Rinaldi L, Borzacchiello D, Gentile A, Lombardi A, Feliciello A, Esposito G, Perrino C. Loss of Akap1 Exacerbates Pressure Overload-Induced Cardiac Hypertrophy and Heart Failure. Front Physiol 2018; 9:558. [PMID: 29892230 PMCID: PMC5985454 DOI: 10.3389/fphys.2018.00558] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/30/2018] [Indexed: 01/05/2023] Open
Abstract
Left ventricular hypertrophy (LVH) is a major contributor to the development of heart failure (HF). Alterations in cyclic adenosine monophosphate (cAMP)-dependent signaling pathways participate in cardiomyocyte hypertrophy and mitochondrial dysfunction occurring in LVH and HF. cAMP signals are received and integrated by a family of cAMP-dependent protein kinase A (PKA) anchor proteins (AKAPs), tethering PKA to discrete cellular locations. AKAPs encoded by the Akap1 gene (mitoAKAPs) promote PKA mitochondrial targeting, regulating mitochondrial structure and function, reactive oxygen species production, and cell survival. To determine the role of mitoAKAPs in LVH development, in the present investigation, mice with global genetic deletion of Akap1 (Akap1-/-), Akap1 heterozygous (Akap1+/-), and their wild-type (wt) littermates underwent transverse aortic constriction (TAC) or SHAM procedure for 1 week. In wt mice, pressure overload induced the downregulation of AKAP121, the major cardiac mitoAKAP. Compared to wt, Akap1-/- mice did not display basal alterations in cardiac structure or function and cardiomyocyte size or fibrosis. However, loss of Akap1 exacerbated LVH and cardiomyocyte hypertrophy induced by pressure overload and accelerated the progression toward HF in TAC mice, and these changes were not observed upon prevention of AKAP121 degradation in seven in absentia homolog 2 (Siah2) knockout mice (Siah2-/-). Loss of Akap1 was also associated to a significant increase in cardiac apoptosis as well as lack of activation of Akt signaling after pressure overload. Taken together, these results demonstrate that in vivo genetic deletion of Akap1 enhances LVH development and accelerates pressure overload-induced cardiac dysfunction, pointing at Akap1 as a novel repressor of pathological LVH. These results confirm and extend the important role of mitoAKAPs in cardiac response to stress.
Collapse
Affiliation(s)
| | - Nicola Boccella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Fabio Cattaneo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Anna Franzone
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
- Department of Cardiology, Inselspital, Universitätsspital Bern, Bern, Switzerland
| | - Stefania D’Apice
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Giugliano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | | | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
39
|
Yu J, Zhang Y, Zhou D, Wu J, Luo R. Higher expression of A-kinase anchoring-protein 1 predicts poor prognosis in human hepatocellular carcinoma. Oncol Lett 2018; 16:131-136. [PMID: 29928393 PMCID: PMC6006472 DOI: 10.3892/ol.2018.8685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
A-kinase anchoring protein 1 (AKAP1) plays important regulatory roles in the regulation of mitochondrial function, oxidative metabolism, and cell survival. However, the expression pattern and prognostic value of AKAP1 in hepatocellular carcinoma (HCC) remains unclear. AKAP1 expression levels in tumor and matched non-tumor tissues were evaluated using reverse transcription-quantitative polymerase chain reaction and immunohistochemical staining. Kaplan-Meier and Cox regression analyses were used to analyze the survival rates. We found that AKAP1 protein expression was increased in HCC tissues, and high AKAP1 expression was associated with tumor size (P=0.024), Tumor-Node-Metastasis stage (P=0.0296) and portal vein thrombosis (P=0.00498). Kaplan-Meier survival analyses further revealed that high AKAP1 expression was associated with poor overall (P=0.004) and disease-free survival (DFS) (P=0.002) rates in patients with HCC. Multivariate survival analysis revealed that AKAP1 served as an independent poor prognostic factor for DFS rates. The findings of the present study indicated that AKAP1 expression may contribute to HCC progression. High AKAP1 expression could serve as a valuable prognostic biomarker in predicting the survival of patients with HCC following radical resection.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrinology, CHC International Hospital, Cixi, Zhejiang 315315, P.R. China
| | - Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Dongxun Zhou
- Department of Endoscopy, Eastern Hepato-biliary Surgery Hospital, Shanghai 200438, P.R. China
| | - Jun Wu
- Department of Endoscopy, Eastern Hepato-biliary Surgery Hospital, Shanghai 200438, P.R. China
| | - Rong Luo
- Department of Endocrinology, CHC International Hospital, Cixi, Zhejiang 315315, P.R. China
| |
Collapse
|
40
|
Mitochondrial cAMP-PKA signaling: What do we really know? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:868-877. [PMID: 29694829 DOI: 10.1016/j.bbabio.2018.04.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
Mitochondria are key organelles for cellular homeostasis. They generate the most part of ATP that is used by cells through oxidative phosphorylation. They also produce reactive oxygen species, neurotransmitters and other signaling molecules. They are important for calcium homeostasis and apoptosis. Considering the role of this organelle, it is not surprising that most mitochondrial dysfunctions are linked to the development of pathologies. Various mechanisms adjust mitochondrial activity according to physiological needs. The cAMP-PKA signaling emerged in recent years as a direct and powerful mean to regulate mitochondrial functions. Multiple evidence demonstrates that such pathway can be triggered from cytosol or directly within mitochondria. Notably, specific anchor proteins target PKA to mitochondria whereas enzymes necessary for generation and degradation of cAMP are found directly in these organelles. Mitochondrial PKA targets proteins localized in different compartments of mitochondria, and related to various functions. Alterations of mitochondrial cAMP-PKA signaling affect the development of several physiopathological conditions, including neurodegenerative diseases. It is however difficult to discriminate between the effects of cAMP-PKA signaling triggered from cytosol or directly in mitochondria. The specific roles of PKA localized in different mitochondrial compartments are also not completely understood. The aim of this work is to review the role of cAMP-PKA signaling in mitochondrial (patho)physiology.
Collapse
|
41
|
The role of compartmentalized signaling pathways in the control of mitochondrial activities in cancer cells. Biochim Biophys Acta Rev Cancer 2018; 1869:293-302. [PMID: 29673970 DOI: 10.1016/j.bbcan.2018.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 02/06/2023]
Abstract
Mitochondria are the powerhouse organelles present in all eukaryotic cells. They play a fundamental role in cell respiration, survival and metabolism. Stimulation of G-protein coupled receptors (GPCRs) by dedicated ligands and consequent activation of the cAMP·PKA pathway finely couple energy production and metabolism to cell growth and survival. Compartmentalization of PKA signaling at mitochondria by A-Kinase Anchor Proteins (AKAPs) ensures efficient transduction of signals generated at the cell membrane to the organelles, controlling important aspects of mitochondrial biology. Emerging evidence implicates mitochondria as essential bioenergetic elements of cancer cells that promote and support tumor growth and metastasis. In this context, mitochondria provide the building blocks for cellular organelles, cytoskeleton and membranes, and supply all the metabolic needs for the expansion and dissemination of actively replicating cancer cells. Functional interference with mitochondrial activity deeply impacts on cancer cell survival and proliferation. Therefore, mitochondria represent valuable targets of novel therapeutic approaches for the treatment of cancer patients. Understanding the biology of mitochondria, uncovering the molecular mechanisms regulating mitochondrial activity andmapping the relevant metabolic and signaling networks operating in cancer cells will undoubtly contribute to create a molecular platform to be used for the treatment of proliferative disorders. Here, we will highlight the emerging roles of signaling pathways acting downstream to GPCRs and their intersection with the ubiquitin proteasome system in the control of mitochondrial activity in different aspects of cancer cell biology.
Collapse
|
42
|
Schiattarella GG, Cattaneo F, Carrizzo A, Paolillo R, Boccella N, Ambrosio M, Damato A, Pironti G, Franzone A, Russo G, Magliulo F, Pirozzi M, Storto M, Madonna M, Gargiulo G, Trimarco V, Rinaldi L, De Lucia M, Garbi C, Feliciello A, Esposito G, Vecchione C, Perrino C. Akap1
Regulates Vascular Function and Endothelial Cells Behavior. Hypertension 2018; 71:507-517. [DOI: 10.1161/hypertensionaha.117.10185] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 08/29/2017] [Accepted: 12/14/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Gabriele Giacomo Schiattarella
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Fabio Cattaneo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Albino Carrizzo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Roberta Paolillo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Nicola Boccella
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Mariateresa Ambrosio
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Antonio Damato
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Gianluigi Pironti
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Anna Franzone
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giusi Russo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Fabio Magliulo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Marinella Pirozzi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Marianna Storto
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Michele Madonna
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giuseppe Gargiulo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Valentina Trimarco
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Laura Rinaldi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Massimiliano De Lucia
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Corrado Garbi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Antonio Feliciello
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giovanni Esposito
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Carmine Vecchione
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Cinzia Perrino
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| |
Collapse
|
43
|
Gaignard P, Fréchou M, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Sex differences in brain mitochondrial metabolism: influence of endogenous steroids and stroke. J Neuroendocrinol 2018. [PMID: 28650095 DOI: 10.1111/jne.12497] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Steroids are neuroprotective and a growing body of evidence indicates that mitochondria are a potential target of their effects. The mitochondria are the site of cellular energy synthesis, regulate oxidative stress and play a key role in cell death after brain injury and neurodegenerative diseases. After providing a summary of the literature on the general functions of mitochondria and the effects of sex steroid administrations on mitochondrial metabolism, we summarise and discuss our recent findings concerning sex differences in brain mitochondrial function under physiological and pathological conditions. To analyse the influence of endogenous sex steroids, the oxidative phosphorylation system, mitochondrial oxidative stress and brain steroid levels were compared between male and female mice, either intact or gonadectomised. The results obtained show that females have higher a mitochondrial respiration and lower oxidative stress compared to males and also that these differences were suppressed by ovariectomy but not orchidectomy. We have also shown that the decrease in brain mitochondrial respiration induced by ischaemia/reperfusion is different according to sex. In both sexes, treatment with progesterone reduced the ischaemia/reperfusion-induced mitochondrial alterations. Our findings indicate sex differences in brain mitochondrial function under physiological conditions, as well as after stroke, and identify mitochondria as a target of the neuroprotective properties of progesterone. Thus, it is necessary to investigate sex specificity in brain physiopathological mechanisms, especially when mitochondria impairment is involved.
Collapse
Affiliation(s)
- P Gaignard
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - M Fréchou
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - P Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - P Thérond
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - M Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - A Slama
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - R Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
44
|
How AMPK and PKA Interplay to Regulate Mitochondrial Function and Survival in Models of Ischemia and Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4353510. [PMID: 29391924 PMCID: PMC5748092 DOI: 10.1155/2017/4353510] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a conserved, redox-activated master regulator of cell metabolism. In the presence of oxidative stress, AMPK promotes cytoprotection by enhancing the conservation of energy by suppressing protein translation and by stimulating autophagy. AMPK interplays with protein kinase A (PKA) to regulate oxidative stress, mitochondrial function, and cell survival. AMPK and dual-specificity A-kinase anchoring protein 1 (D-AKAP1), a mitochondrial-directed scaffold of PKA, interact to regulate mitochondrial function and oxidative stress in cardiac and endothelial cells. Ischemia and diabetes, a chronic disease that increases the onset of cardiovascular diseases, suppress the cardioprotective effects of AMPK and PKA. Here, we review the molecular mechanisms by which AMPK and D-AKAP1/PKA interplay to regulate mitochondrial function, oxidative stress, and signaling pathways that prime endothelial cells, cardiac cells, and neurons for cytoprotection against oxidative stress. We discuss recent literature showing how temporal dynamics and localization of activated AMPK and PKA holoenzymes play a crucial role in governing cellular bioenergetics and cell survival in models of ischemia, cardiovascular diseases, and diabetes. Finally, we propose therapeutic strategies that tout localized PKA and AMPK signaling to reverse mitochondrial dysfunction, oxidative stress, and death of neurons and cardiac and endothelial cells during ischemia and diabetes.
Collapse
|
45
|
Li B, Wang W, Li Z, Chen Z, Zhi X, Xu J, Li Q, Wang L, Huang X, Wang L, Wei S, Sun G, Zhang X, He Z, Zhang L, Zhang D, Xu H, El-Rifai W, Xu Z. MicroRNA-148a-3p enhances cisplatin cytotoxicity in gastric cancer through mitochondrial fission induction and cyto-protective autophagy suppression. Cancer Lett 2017; 410:212-227. [PMID: 28965855 PMCID: PMC5675767 DOI: 10.1016/j.canlet.2017.09.035] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022]
Abstract
Cisplatin (CDDP) resistance is a major clinical problem associated with poor prognosis in gastric cancer (GC) patients. In this study, we performed integrated analysis of TCGA data from microRNAs (miRNAs) expression matrix of GC patients who received CDDP-based chemotherapy with GEO dataset which contains differential miRNAs expression profiles in CDDP-resistant and -sensitive cell lines. We identified miR-148a-3p downregulation as a key step involved in CDDP resistance. Using a cohort consisting 105 GC patients who received CDDP-based therapy, we found that miR-148a-3p downregulation was associated with a decrease in patients' disease-free survival (DFS, P = 0.0077). A series of experiment data demonstrated that: 1) miR-148a-3p was downregulated in CDDP-resistant GC cell lines; 2) miR-148a-3p reconstitution sensitized CDDP-resistant cells to CDDP treatment through promoting mitochondrial fission and decreasing AKAP1 expression level; 3) AKAP1 played a novel role in CDDP resistance by inhibiting P53-mediated DRP1 dephosphorylation; 4) miR-148a-3p reconstitution in CDDP-resistant cells inhibits the cyto-protective autophagy by suppressing RAB12 expression and mTOR1 activation. Taken together, our study demonstrates that miR-148a-3p could be a promising prognostic marker or therapeutic candidate for overcoming CDDP resistance in GC.
Collapse
Affiliation(s)
- Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zheng Chen
- Department of Surgery and Cancer Biology, Vanderbilt University Medical Center, Nashville, 37232, TN, USA
| | - Xiaofei Zhi
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu province, China
| | - Jianghao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Qing Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Xiaoxu Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Linjun Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Song Wei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Guangli Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Xuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Lu Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Wael El-Rifai
- Department of Surgery and Cancer Biology, Vanderbilt University Medical Center, Nashville, 37232, TN, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, 37232, TN, USA.
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China.
| |
Collapse
|
46
|
Tsushima K, Bugger H, Wende AR, Soto J, Jenson GA, Tor AR, McGlauflin R, Kenny HC, Zhang Y, Souvenir R, Hu XX, Sloan CL, Pereira RO, Lira VA, Spitzer KW, Sharp TL, Shoghi KI, Sparagna GC, Rog-Zielinska EA, Kohl P, Khalimonchuk O, Schaffer JE, Abel ED. Mitochondrial Reactive Oxygen Species in Lipotoxic Hearts Induce Post-Translational Modifications of AKAP121, DRP1, and OPA1 That Promote Mitochondrial Fission. Circ Res 2017; 122:58-73. [PMID: 29092894 DOI: 10.1161/circresaha.117.311307] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022]
Abstract
RATIONALE Cardiac lipotoxicity, characterized by increased uptake, oxidation, and accumulation of lipid intermediates, contributes to cardiac dysfunction in obesity and diabetes mellitus. However, mechanisms linking lipid overload and mitochondrial dysfunction are incompletely understood. OBJECTIVE To elucidate the mechanisms for mitochondrial adaptations to lipid overload in postnatal hearts in vivo. METHODS AND RESULTS Using a transgenic mouse model of cardiac lipotoxicity overexpressing ACSL1 (long-chain acyl-CoA synthetase 1) in cardiomyocytes, we show that modestly increased myocardial fatty acid uptake leads to mitochondrial structural remodeling with significant reduction in minimum diameter. This is associated with increased palmitoyl-carnitine oxidation and increased reactive oxygen species (ROS) generation in isolated mitochondria. Mitochondrial morphological changes and elevated ROS generation are also observed in palmitate-treated neonatal rat ventricular cardiomyocytes. Palmitate exposure to neonatal rat ventricular cardiomyocytes initially activates mitochondrial respiration, coupled with increased mitochondrial polarization and ATP synthesis. However, long-term exposure to palmitate (>8 hours) enhances ROS generation, which is accompanied by loss of the mitochondrial reticulum and a pattern suggesting increased mitochondrial fission. Mechanistically, lipid-induced changes in mitochondrial redox status increased mitochondrial fission by increased ubiquitination of AKAP121 (A-kinase anchor protein 121) leading to reduced phosphorylation of DRP1 (dynamin-related protein 1) at Ser637 and altered proteolytic processing of OPA1 (optic atrophy 1). Scavenging mitochondrial ROS restored mitochondrial morphology in vivo and in vitro. CONCLUSIONS Our results reveal a molecular mechanism by which lipid overload-induced mitochondrial ROS generation causes mitochondrial dysfunction by inducing post-translational modifications of mitochondrial proteins that regulate mitochondrial dynamics. These findings provide a novel mechanism for mitochondrial dysfunction in lipotoxic cardiomyopathy.
Collapse
Affiliation(s)
- Kensuke Tsushima
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Heiko Bugger
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Adam R Wende
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Jamie Soto
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Gregory A Jenson
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Austin R Tor
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Rose McGlauflin
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Helena C Kenny
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Yuan Zhang
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Rhonda Souvenir
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Xiao X Hu
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Crystal L Sloan
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Renata O Pereira
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Vitor A Lira
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Kenneth W Spitzer
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Terry L Sharp
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Kooresh I Shoghi
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Genevieve C Sparagna
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Eva A Rog-Zielinska
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Peter Kohl
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Oleh Khalimonchuk
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - Jean E Schaffer
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.)
| | - E Dale Abel
- From the Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine (K.T., J.S., G.A.J., A.R.T., R.M., H.C.K., Y.Z., R.S., R.O.P., E.D.A.) and Department of Health and Human Physiology (V.A.L.), University of Iowa, Iowa City; Division of Endocrinology, Metabolism, and Diabetes and Program in Molecular Medicine (K.T., H.B., A.R.W., J.S., X.X.H., C.L.S., E.D.A.), Nora Eccles Harrison Cardiovascular Research and Training Institute (K.W.S.), and Department of Biochemistry (O.K.), University of Utah School of Medicine, Salt Lake City; Cardiology and Angiology I (H.B.) and Institute for Experimental Cardiovascular Medicine (E.A.R.-Z., P.K.), Heart Center Freiburg University, and Faculty of Medicine (H.B., E.A.R.-Z., P.K.), University of Freiburg, Germany; Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.); Department of Radiology (T.L.S., K.I.S.) and Diabetic Cardiovascular Disease Center, Cardiovascular Division (J.E.S.), Washington University School of Medicine, St. Louis, MO; Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora (G.C.S.); and Department of Biochemistry and Nebraska Redox Biology Center, University of Nebraska, Lincoln (O.K.).
| |
Collapse
|
47
|
Ong T, Solecki DJ. Seven in Absentia E3 Ubiquitin Ligases: Central Regulators of Neural Cell Fate and Neuronal Polarity. Front Cell Neurosci 2017; 11:322. [PMID: 29081737 PMCID: PMC5646344 DOI: 10.3389/fncel.2017.00322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/31/2022] Open
Abstract
During neural development, neural precursors transition from a proliferative state within their germinal niches to a migratory state as they relocate to their final laminar positions. Transitions across these states are coupled with dynamic alterations in cellular polarity. This key feature can be seen throughout the developing vertebrate brain, in which neural stem cells give rise to multipolar or unpolarized transit-amplifying progenitors. These transit-amplifying progenitors then expand to give rise to mature neuronal lineages that become polarized as they initiate radial migration to their final laminar positions. The conventional understanding of the cellular polarity regulatory program has revolved around signaling cascades and transcriptional networks. In this review, we discuss recent discoveries concerning the role of the Siah2 ubiquitin ligase in initiating neuronal polarity during cerebellar development. Given the unique features of Siah ubiquitin ligases, we highlight some of the key substrates that play important roles in cellular polarity and propose a function for the Siah ubiquitin proteasome pathway in mediating a post-translational regulatory network to control the onset of polarization.
Collapse
Affiliation(s)
- Taren Ong
- Cancer and Developmental Biology Track, Integrated Biomedical Sciences Graduate Program, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
48
|
Das Banerjee T, Dagda RY, Dagda M, Chu CT, Rice M, Vazquez-Mayorga E, Dagda RK. PINK1 regulates mitochondrial trafficking in dendrites of cortical neurons through mitochondrial PKA. J Neurochem 2017; 142:545-559. [PMID: 28556983 DOI: 10.1111/jnc.14083] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022]
Abstract
Mitochondrial Protein Kinase A (PKA) and PTEN-induced kinase 1 (PINK1), which is linked to Parkinson's disease, are two neuroprotective serine/threonine kinases that regulate dendrite remodeling and mitochondrial function. We have previously shown that PINK1 regulates dendrite morphology by enhancing PKA activity. Here, we show the molecular mechanisms by which PINK1 and PKA in the mitochondrion interact to regulate dendrite remodeling, mitochondrial morphology, content, and trafficking in dendrites. PINK1-deficient cortical neurons exhibit impaired mitochondrial trafficking, reduced mitochondrial content, fragmented mitochondria, and a reduction in dendrite outgrowth compared to wild-type neurons. Transient expression of wild-type, but not a PKA-binding-deficient mutant of the PKA-mitochondrial scaffold dual-specificity A Kinase Anchoring Protein 1 (D-AKAP1), restores mitochondrial trafficking, morphology, and content in dendrites of PINK1-deficient cortical neurons suggesting that recruiting PKA to the mitochondrion reverses mitochondrial pathology in dendrites induced by loss of PINK1. Mechanistically, full-length and cleaved forms of PINK1 increase the binding of the regulatory subunit β of PKA (PKA/RIIβ) to D-AKAP1 to enhance the autocatalytic-mediated phosphorylation of PKA/RIIβ and PKA activity. D-AKAP1/PKA governs mitochondrial trafficking in dendrites via the Miro-2/TRAK2 complex and by increasing the phosphorylation of Miro-2. Our study identifies a new role of D-AKAP1 in regulating mitochondrial trafficking through Miro-2, and supports a model in which PINK1 and mitochondrial PKA participate in a similar neuroprotective signaling pathway to maintain dendrite connectivity.
Collapse
Affiliation(s)
- Tania Das Banerjee
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Raul Y Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Charleen T Chu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Monica Rice
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Emmanuel Vazquez-Mayorga
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA.,Department of Biomedical Sciences, Universidad Autonoma de Ciudad Juarez, Cd. Juarez, Mexico
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| |
Collapse
|
49
|
Mitochondrial AKAP1 supports mTOR pathway and tumor growth. Cell Death Dis 2017; 8:e2842. [PMID: 28569781 PMCID: PMC5520900 DOI: 10.1038/cddis.2017.241] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/06/2017] [Accepted: 04/28/2017] [Indexed: 12/23/2022]
Abstract
Mitochondria are the powerhouses of energy production and the sites where metabolic pathway and survival signals integrate and focus, promoting adaptive responses to hormone stimulation and nutrient availability. Increasing evidence suggests that mitochondrial bioenergetics, metabolism and signaling are linked to tumorigenesis. AKAP1 scaffolding protein integrates cAMP and src signaling on mitochondria, regulating organelle biogenesis, oxidative metabolism and cell survival. Here, we provide evidence that AKAP1 is a transcriptional target of Myc and supports the growth of cancer cells. We identify Sestrin2, a leucine sensor and inhibitor of the mammalian target of rapamycin (mTOR), as a novel component of the complex assembled by AKAP1 on mitochondria. Downregulation of AKAP1 impaired mTOR pathway and inhibited glioblastoma growth. Both effects were reversed by concomitant depletion of AKAP1 and sestrin2. High levels of AKAP1 were found in a wide variety of high-grade cancer tissues. In lung cancer, AKAP1 expression correlates with high levels of Myc, mTOR phosphorylation and reduced patient survival. Collectively, these data disclose a previously unrecognized role of AKAP1 in mTOR pathway regulation and cancer growth. AKAP1/mTOR signal integration on mitochondria may provide a new target for cancer therapy.
Collapse
|
50
|
Torres-Quesada O, Mayrhofer JE, Stefan E. The many faces of compartmentalized PKA signalosomes. Cell Signal 2017; 37:1-11. [PMID: 28528970 DOI: 10.1016/j.cellsig.2017.05.012] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 01/03/2023]
Abstract
Cellular signal transmission requires the dynamic formation of spatiotemporally controlled molecular interactions. At the cell surface information is received by receptor complexes and relayed through intracellular signaling platforms which organize the actions of functionally interacting signaling enzymes and substrates. The list of hormone or neurotransmitter pathways that utilize the ubiquitous cAMP-sensing protein kinase A (PKA) system is expansive. This requires that the specificity, duration, and intensity of PKA responses are spatially and temporally restricted. Hereby, scaffolding proteins take the center stage for ensuring proper signal transmission. They unite second messenger sensors, activators, effectors, and kinase substrates within cellular micro-domains to precisely control and route signal propagation. A-kinase anchoring proteins (AKAPs) organize such subcellular signalosomes by tethering the PKA holoenzyme to distinct cell compartments. AKAPs differ in their modular organization showing pathway specific arrangements of interaction motifs or domains. This enables the cell- and compartment- guided assembly of signalosomes with unique enzyme composition and function. The AKAP-mediated clustering of cAMP and other second messenger sensing and interacting signaling components along with functional successive enzymes facilitates the rapid and precise dissemination of incoming signals. This review article delineates examples for different means of PKA regulation and for snapshots of compartmentalized PKA signalosomes.
Collapse
Affiliation(s)
- Omar Torres-Quesada
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Johanna E Mayrhofer
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Eduard Stefan
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| |
Collapse
|