1
|
da Silva GB, Braga GDC, Simões JLB, Bagatini MD, Kempka AP. Mitochondrial dysfunction and carcinogenesis: The engagement of ion channels in cancer development. Cell Calcium 2025; 128:103010. [PMID: 40043325 DOI: 10.1016/j.ceca.2025.103010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/27/2025] [Accepted: 02/20/2025] [Indexed: 05/11/2025]
Abstract
Mitochondria represent a fundamental structure for cellular homeostasis, controlling multiple conditions regarding energetic functions and cellular survival. To maintain these organelles functioning in ideal conditions, their membranes count with ion channels for different inorganic ions, which must be balanced to offer the proper function for both the organelle and the cell. However, studies have shown that other health conditions impair the activities of mitochondrial ion channels, including cancer. In this sense, the altered activities of potassium, calcium, and calcium-activated potassium channels are mainly linked with cancer development and cellular homeostasis alteration, demonstrating their role as pharmacological targets. With that in mind, scientists have found significant mitochondrial and cellular responses related to apoptosis and reduction of cellular survival from cells with modulated ion channels, indicating the potential of this possible therapy in carcinogenic contexts. Nonetheless, few studies still evaluate mitochondrial ion channel modulation as a treatment against cancer. Hence, more research must be conducted on this subject.
Collapse
|
2
|
Zhang L, Ye J, Qiu C. Twist2 knockdown alleviates renal ischemia-reperfusion injury by maintaining mitochondrial function and enhancing mitophagy through Bnip3. Hum Cell 2025; 38:50. [PMID: 39918659 DOI: 10.1007/s13577-025-01177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Renal ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury (AKI). Twist-related protein 2 (Twist2) is a basic helix/loop/helix transcription factor. However, the underlying effects of Twist2 in IRI remain to be elucidated. Herein, we found that the expression of Twist2 was significantly upregulated in renal tissues of mice induced by ischemia/reperfusion (I/R) and in human renal tubular epithelial cell HK-2 exposed to hypoxia-reoxygenation. We silenced Twist2 with RNAi technology. Twist2 knockdown alleviated renal pathological damage in mice. Twist2 depletion ameliorated IRI-induced mitochondrial dysfunction, such as increasing ATP content and mitochondrial DNA copy number and restoring mitochondrial membrane potential in the kidneys of mice, and similar results were observed in in vitro experiments. Twist2 interference increased the expression of LC3B and decreased the expression of p62 and mitochondrial membrane proteins TIMM23 and TOMM20 both in vivo and in vitro. Electron microscope and the co-localization of LC3B and mitotracker DsRed suggested the induction of autophagy and mitophagy after Twist2 knockdown in kidneys or cells. Mechanism studies revealed that Twist2 exerted a direct inhibitory effect on BCL2 interacting protein 3 (Bnip3) transcriptional activity by targeting the Bnip3 promoter. In hypoxia/reoxygenation-induced renal tubular epithelial cells, the interference of Bnip3 reversed the effect of Twist2 depletion on mitochondrial function and mitophagy. In conclusion, our findings suggest that the depletion of Twist2 exerts renoprotective effect in I/R-induced AKI. Twist2 regulates mitochondrial function and mitophagy in part by targeting and downregulating Bnip3. Our study provides new insights into the pathological mechanisms of I/R-induced AKI.
Collapse
Affiliation(s)
- Lexi Zhang
- Department of Kidney Transplantation, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Jianfeng Ye
- Department of Kidney Transplantation, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Cheng Qiu
- Department of Kidney Transplantation, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
3
|
Guo L. Mitochondrial permeability transition mediated by MTCH2 and F-ATP synthase contributes to ferroptosis defense. FEBS Lett 2025; 599:352-366. [PMID: 39227319 DOI: 10.1002/1873-3468.15008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/16/2024] [Accepted: 07/12/2024] [Indexed: 09/05/2024]
Abstract
The opening of the mitochondrial permeability transition pore (PTP), a Ca2+-dependent pore located in the inner mitochondrial membrane, triggers mitochondrial outer membrane permeabilization (MOMP) and induces organelle rupture. However, the underlying mechanism of PTP-induced MOMP remains unclear. Mitochondrial carrier homolog 2 (MTCH2) mediates MOMP process by facilitating the recruitment of tBID to mitochondria. Here, we show that MTCH2 binds to cyclophilin D (CyPD) and promotes the dimerization of F-ATP synthase via interaction with subunit j. The interplay between MTCH2 and subunit j coordinates MOMP and PTP to mediate the occurrence of mitochondrial permeability transition. Knockdown of CyPD, MTCH2 and subunit j markedly sensitizes cells to RSL3-induced ferroptosis, which is prevented by MitoTEMPO, suggesting that mitochondrial permeability transition mediates ferroptosis defense.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Tommasin L, Carrer A, Nata FB, Frigo E, Fogolari F, Lippe G, Carraro M, Bernardi P. Adenine nucleotide translocator and ATP synthase cooperate in mediating the mitochondrial permeability transition. J Physiol 2025. [PMID: 39808538 DOI: 10.1113/jp287147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
The permeability transition (PT) is a permeability increase of the mitochondrial inner membrane causing mitochondrial swelling in response to matrix Ca2+. The PT is mediated by regulated channel(s), the PT pore(s) (PTP), which can be generated by at least two components, adenine nucleotide translocator (ANT) and ATP synthase. Whether these provide independent permeation pathways remains to be established. Here, we assessed the contribution of ANT to the PT based on the effects of the selective ANT inhibitors atractylate (ATR) and bongkrekate (BKA), which trigger and inhibit channel formation by ANT, respectively. BKA partially inhibited Ca2+-dependent PT and did not prevent the inducing effect of phenylarsine oxide, which was still present in mouse embryonic fibroblasts deleted for all ANT isoforms. The contribution of ANT to the PT emerged at pH 6.5 (a condition that inhibits ATP synthase channel opening) in the presence of ATR, which triggered mitochondrial swelling and elicited currents in patch-clamped mitoplasts. Unexpectedly, ANT-dependent PT at pH 6.5 could also be stimulated by benzodiazepine-423 [a selective ligand of the oligomycin sensitivity conferral protein (OSCP) subunit of ATP synthase], suggesting that the ANT channel is regulated by the peripheral stalk of ATP synthase. In keeping with docking simulations, ANT could be co-immunoprecipitated with ATP synthase subunits c and g, and oligomycin (which binds adjacent c subunits) decreased the association of ANT with subunit c. These results reveal a close cooperation between ANT and ATP synthase in the PT and open new perspectives in the study of this process. KEY POINTS: We have assessed the relative role of adenine nucleotide translocator (ANT) and ATP synthase in generating the mitochondrial permeability transition (PT). At pH 7.4, bongkrekate had little effect on Ca2+-dependent PT, and did not prevent the inducing effect of phenylarsine oxide, which was still present in mouse embryonic fibroblasts deleted for all ANT isoforms. The contribution of ANT emerged at pH 6.5 (which inhibits ATP synthase channel opening) in the presence of atractylate, which triggered mitochondrial swelling and elicited currents in patch-clamped mitoplasts. Benzodiazepine-423, a selective ligand of the oligomycin sensitivity conferral protein subunit of ATP synthase, stimulated ANT-dependent PT at pH 6.5, suggesting that the ANT channel is regulated by the peripheral stalk of ATP synthase. ANT could be co-immunoprecipitated with ATP synthase subunits c and g; oligomycin, which binds adjacent c subunits, decreased the association with subunit c, in keeping with docking simulations.
Collapse
Affiliation(s)
- Ludovica Tommasin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Carrer
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Elena Frigo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Federico Fogolari
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Giovanna Lippe
- Department of Medicine, University of Udine, Udine, Italy
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Mastoor Y, Murphy E, Roman B. Mechanisms of postischemic cardiac death and protection following myocardial injury. J Clin Invest 2025; 135:e184134. [PMID: 39744953 DOI: 10.1172/jci184134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Acute myocardial infarction (MI) is a leading cause of death worldwide. Although with current treatment, acute mortality from MI is low, the damage and remodeling associated with MI are responsible for subsequent heart failure. Reducing cell death associated with acute MI would decrease the mortality associated with heart failure. Despite considerable study, the precise mechanism by which ischemia and reperfusion (I/R) trigger cell death is still not fully understood. In this Review, we summarize the changes that occur during I/R injury, with emphasis on those that might initiate cell death, such as calcium overload and oxidative stress. We review cell-death pathways and pathway crosstalk and discuss cardioprotective approaches in order to provide insight into mechanisms that could be targeted with therapeutic interventions. Finally, we review cardioprotective clinical trials, with a focus on possible reasons why they were not successful. Cardioprotection has largely focused on inhibiting a single cell-death pathway or one death-trigger mechanism (calcium or ROS). In treatment of other diseases, such as cancer, the benefit of targeting multiple pathways with a "drug cocktail" approach has been demonstrated. Given the crosstalk between cell-death pathways, targeting multiple cardiac death mechanisms should be considered.
Collapse
|
6
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
7
|
Coluccino G, Negro A, Filippi A, Bean C, Muraca VP, Gissi C, Canetti D, Mimmi MC, Zamprogno E, Ciscato F, Acquasaliente L, De Filippis V, Comelli M, Carraro M, Rasola A, Gerle C, Bernardi P, Corazza A, Lippe G. N-terminal cleavage of cyclophilin D boosts its ability to bind F-ATP synthase. Commun Biol 2024; 7:1486. [PMID: 39528709 PMCID: PMC11555324 DOI: 10.1038/s42003-024-07172-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Cyclophilin (CyP) D is a regulator of the mitochondrial F-ATP synthase. Here we report the discovery of a form of CyPD lacking the first 10 (mouse) or 13 (human) N-terminal residues (ΔN-CyPD), a protein region with species-specific features. NMR studies on recombinant human full-length CyPD (FL-CyPD) and ΔN-CyPD form revealed that the N-terminus is highly flexible, in contrast with the rigid globular part. We have studied the interactions of FL and ΔN-CyPD with F-ATP synthase at the OSCP subunit, a site where CyPD binding inhibits catalysis and favors the transition of the enzyme complex to the permeability transition pore. At variance from FL-CyPD, ΔN-CyPD binds OSCP in saline media, indicating that the N-terminus substantially decreases the binding affinity for OSCP. We also provide evidence that calpain 1 is responsible for generation of ΔN-CyPD in cells. Altogether, our work suggests the existence of a novel mechanism of modulation of CyPD through cleavage of its N-terminus that may have significant pathophysiological implications.
Collapse
Affiliation(s)
| | - Alessandro Negro
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Antonio Filippi
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Camilla Bean
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | | | - Clarissa Gissi
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Diana Canetti
- Centre for Amyloidosis, Division of Medicine, University College London, London, NW32PF, UK
| | - Maria Chiara Mimmi
- Centre for Inherited Cardiovascular Diseases, IRCCS San Matteo Hospital Foundation, 27100, Pavia, Italy
| | - Elisa Zamprogno
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Francesco Ciscato
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Institute of Neuroscience, National Research Council (CNR), 35131, Padova, Italy
| | - Laura Acquasaliente
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Vincenzo De Filippis
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Marina Comelli
- Department of Medicine, University of Udine, 33100, Udine, Italy
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Andrea Rasola
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Christoph Gerle
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Kouto, Hyogo, Japan
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | | | - Giovanna Lippe
- Department of Medicine, University of Udine, 33100, Udine, Italy.
| |
Collapse
|
8
|
Gutiérrez-Aguilar M, Klutho PJ, Aguayo-Ortiz R, Song L, Baines CP. Endogenous complement 1q binding protein (C1qbp) regulates mitochondrial permeability transition and post-myocardial infarction remodeling and dysfunction. J Mol Cell Cardiol 2024; 196:1-11. [PMID: 39209214 PMCID: PMC11534557 DOI: 10.1016/j.yjmcc.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The mitochondrial permeability transition (MPT) pore regulates necrotic cell death following diverse cardiac insults. While the componentry of the pore itself remains controversial, Cyclophilin D (CypD) has been well-established as a positive regulator of pore opening. We have previously identified Complement 1q-binding protein (C1qbp) as a novel CypD-interacting molecule and a negative regulator of MPT-dependent cell death in vitro. However, its effects on the MPT pore and sensitivity to cell death in the heart remain untested. We therefore hypothesized that C1qbp would inhibit MPT in cardiac mitochondria and protect cardiac myocytes against cell death in vivo. To investigate the effects of C1qbp in the myocardium we generated gain- and loss-of-function mice. Transgenic C1qbp overexpression resulted in decreased complex protein expression and reduced mitochondrial respiration and ATP production but MPT was unaffected. In contrast, while C1qbp+/- mice did not exhibit any changes in mitochondrial protein expression, respiration, or ATP, the MPT pore was markedly sensitized to Ca2+ in these animals. Neither overexpression nor depletion of C1qbp significantly affected baseline heart morphology or function at 3 months of age. When subjected to myocardial infarction, C1qbp transgenic mice exhibited similar infarct sizes and cardiac remodeling to non-transgenic mice, consistent with the lack of an effect on MPT. In contrast, cardiac scar formation and dysfunction were significantly increased in the C1qbp+/- mice compared to C1qbp+/+ controls. Our results suggest that C1qbp is required for normal regulation of the MPT pore and mitochondrial function, and influences cardiac remodeling following MI, the latter more likely being independent of C1qbp effects on the MPT pore.
Collapse
Affiliation(s)
- Manuel Gutiérrez-Aguilar
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Paula J Klutho
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Rodrigo Aguayo-Ortiz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Lihui Song
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA; Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
9
|
Ma Y, Sun X, Yao X. The role and mechanism of VDAC1 in type 2 diabetes: An underestimated target of environmental pollutants. Mitochondrion 2024; 78:101929. [PMID: 38986923 DOI: 10.1016/j.mito.2024.101929] [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: 02/26/2024] [Revised: 06/08/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disease that accounts for more than 90% of diabetic patients. Its main feature is hyperglycemia due to insulin resistance or insulin deficiency. With changes in diet and lifestyle habits, the incidence of T2D in adolescents has burst in recent decades. The deterioration in the exposure to the environmental pollutants further aggravates the prevalence of T2D, and consequently, it imposes a significant economic burden. Therefore, early prevention and symptomatic treatment are essential to prevent diabetic complications. Mitochondrial number and electron transport chain activity are decreased in the patients with T2D. Voltage-Dependent Anion Channel 1 (VDAC1), as a crucial channel protein on the outer membrane of mitochondria, regulates signal transduction between mitochondria and other cellular components, participating in various biological processes. When VDAC1 exists in oligomeric form, it additionally facilitates the entry and exit of macromolecules into and from mitochondria, modulating insulin secretion. We summarize and highlight the interplay between VDAC1 and T2D, especially in the environmental pollutants-related T2D, shed light on the potential therapeutic implications of targeting VDAC1 monomers and oligomers, providing a new possible target for the treatment of T2D.
Collapse
Affiliation(s)
- Yu Ma
- Environmental and Occupational Health Department, Dalian Medical University, 9 West Lushun South Road, Dalian, China
| | - Xiance Sun
- Environmental and Occupational Health Department, Dalian Medical University, 9 West Lushun South Road, Dalian, China
| | - Xiaofeng Yao
- Environmental and Occupational Health Department, Dalian Medical University, 9 West Lushun South Road, Dalian, China.
| |
Collapse
|
10
|
O’Brien JT, Jalilvand SP, Suji NA, Jupelly RK, Phensy A, Mwirigi JM, Elahi H, Price TJ, Kroener S. Elevations in the Mitochondrial Matrix Protein Cyclophilin D Correlate With Reduced Parvalbumin Expression in the Prefrontal Cortex of Patients With Schizophrenia. Schizophr Bull 2024; 50:1197-1207. [PMID: 38412332 PMCID: PMC11349014 DOI: 10.1093/schbul/sbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS Cognitive deficits in schizophrenia are linked to dysfunctions of the dorsolateral prefrontal cortex (DLPFC), including alterations in parvalbumin (PV)-expressing interneurons (PVIs). Redox dysregulation and oxidative stress may represent convergence points in the pathology of schizophrenia, causing dysfunction of GABAergic interneurons and loss of PV. Here, we show that the mitochondrial matrix protein cyclophilin D (CypD), a critical initiator of the mitochondrial permeability transition pore (mPTP) and modulator of the intracellular redox state, is altered in PVIs in schizophrenia. STUDY DESIGN Western blotting was used to measure CypD protein levels in postmortem DLPFC specimens of schizophrenic patients (n = 27) and matched comparison subjects with no known history of psychiatric or neurological disorders (n = 26). In a subset of this cohort, multilabel immunofluorescent confocal microscopy with unbiased stereological sampling methods were used to quantify (1) numbers of PVI across the cortical mantle (20 unaffected comparison, 14 schizophrenia) and (2) PV and CypD protein levels from PVIs in the cortical layers 2-4 (23 unaffected comparison, 18 schizophrenia). STUDY RESULTS In schizophrenic patients, the overall number of PVIs in the DLPFC was not significantly altered, but in individual PVIs of layers 2-4 PV protein levels decreased along a superficial-to-deep gradient when compared to unaffected comparison subjects. These laminar-specific PVI alterations were reciprocally linked to significant CypD elevations both in PVIs and total DLPFC gray matter. CONCLUSIONS Our findings support previously reported PVI anomalies in schizophrenia and suggest that CypD-mediated mPTP formation could be a potential contributor to PVI dysfunction in schizophrenia.
Collapse
Affiliation(s)
- John T O’Brien
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sophia P Jalilvand
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Neha A Suji
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Rohan K Jupelly
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Aarron Phensy
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Juliet M Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Hajira Elahi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Sven Kroener
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
11
|
Noh MR, Padanilam BJ. Cell death induced by acute renal injury: a perspective on the contributions of accidental and programmed cell death. Am J Physiol Renal Physiol 2024; 327:F4-F20. [PMID: 38660714 PMCID: PMC11390133 DOI: 10.1152/ajprenal.00275.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
The involvement of cell death in acute kidney injury (AKI) is linked to multiple factors including energy depletion, electrolyte imbalance, reactive oxygen species, inflammation, mitochondrial dysfunction, and activation of several cell death pathway components. Since our review in 2003, discussing the relative contributions of apoptosis and necrosis, several other forms of cell death have been identified and are shown to contribute to AKI. Currently, these various forms of cell death can be fundamentally divided into accidental cell death and regulated or programmed cell death based on functional aspects. Several death initiator and effector molecules switch molecules that may act as signaling components triggering either death or protective mechanisms or alternate cell death pathways have been identified as part of the machinery. Intriguingly, several of these cell death pathways share components and signaling pathways suggesting complementary or compensatory functions. Thus, defining the cross talk between distinct cell death pathways and identifying the unique molecular effectors for each type of cell death may be required to develop novel strategies to prevent cell death. Furthermore, depending on the multiple forms of cell death simultaneously induced in different AKI settings, strategies for combination therapies that block multiple cell death pathways need to be developed to completely prevent injury, cell death, and renal function. This review highlights the various cell death pathways, cross talk, and interactions between different cell death modalities in AKI.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Babu J Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
12
|
Meng L, Ouyang Z, Chen Y, Huang C, Yu Y, Fan R. Low-dose BPA-induced neuronal energy metabolism dysfunction and apoptosis mediated by PINK1/parkin mitophagy pathway in juvenile rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172655. [PMID: 38653419 DOI: 10.1016/j.scitotenv.2024.172655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Bisphenol A (BPA) is related to neurological disorders involving mitochondrial dysfunction, while the mechanism remains elusive. Therefore, we explored it through in vitro and in vivo experiments. In vitro, hippocampal neurons derived from neonatal rats of different genders were exposed to 1-100 nM and 100 μM BPA, autophagy activator Rapa and inhibitor 3-MA for 7 d. The results suggested that even nanomolar BPA (1-100 nM) disturbed Ca2+ homeostasis and damaged the integrity of mitochondrial cristae in neurons (p < 0.05). Furthermore, BPA increased the number of autophagic lysosomes, LC3II/LC3I ratio, and p62 expression, and decreased parkin expression (p < 0.05), suggesting that the entry of damaged mitochondria into autophagic pathway was prompted, while the autophagic degradation pathway was blocked. This further disrupts neuronal energy metabolism and promotes neuronal apoptosis. However, Rapa attenuated the adverse effects caused by BPA, while 3-MA exacerbated these reactions. In vivo, exposure of juvenile rats to 0.5, 50, 5000 μg/kg‧bw/day BPA during PND 7-21 markedly impaired the structure of hippocampal mitochondria, increased the number of autophagosomes, the rate of neuronal apoptosis, and the expression levels of pro-apoptotic proteins Cyt C, Bax, Bak1, and Caspase3, and decreased the expression of anti-apoptotic protein Bcl2 (p < 0.05). Particularly, male rats are more sensitive to low-dose BPA than females. Overall, environmental-doses BPA can induce the imbalance of energy metabolism in hippocampal neurons via PINK1/parkin mitophagy, thereby inducing their apoptosis. Importantly, this study provides a theoretical basis for attenuating BPA-related neurological diseases.
Collapse
Affiliation(s)
- Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China; State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yuxin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Chengmeng Huang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yunjiang Yu
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
13
|
Bansal R, Torres M, Hunt M, Wang N, Chatzopoulou M, Manchanda M, Taddeo EP, Shu C, Shirihai OS, Bachar-Wikstrom E, Wikstrom JD. Role of the mitochondrial protein cyclophilin D in skin wound healing and collagen secretion. JCI Insight 2024; 9:e169213. [PMID: 38564292 PMCID: PMC11141914 DOI: 10.1172/jci.insight.169213] [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: 01/26/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Central for wound healing is the formation of granulation tissue, which largely consists of collagen and whose importance stretches past wound healing, including being implicated in both fibrosis and skin aging. Cyclophilin D (CyD) is a mitochondrial protein that regulates the permeability transition pore, known for its role in apoptosis and ischemia-reperfusion. To date, the role of CyD in human wound healing and collagen generation has been largely unexplored. Here, we show that CyD was upregulated in normal wounds and venous ulcers, likely adaptive as CyD inhibition impaired reepithelialization, granulation tissue formation, and wound closure in both human and pig models. Overexpression of CyD increased keratinocyte migration and fibroblast proliferation, while its inhibition reduced migration. Independent of wound healing, CyD inhibition in fibroblasts reduced collagen secretion and caused endoplasmic reticulum collagen accumulation, while its overexpression increased collagen secretion. This was confirmed in a Ppif-KO mouse model, which showed a reduction in skin collagen. Overall, this study revealed previously unreported roles of CyD in skin, with implications for wound healing and beyond.
Collapse
Affiliation(s)
- Ritu Bansal
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Nuoqi Wang
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Margarita Chatzopoulou
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Mansi Manchanda
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Evan P. Taddeo
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Cynthia Shu
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Orian S. Shirihai
- Metabolism Theme
- Department of Molecular and Medical Pharmacology, and
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D. Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Kamińska D, Skrzycki M. Lipid droplets, autophagy, and ER stress as key (survival) pathways during ischemia-reperfusion of transplanted grafts. Cell Biol Int 2024; 48:253-279. [PMID: 38178581 DOI: 10.1002/cbin.12114] [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: 07/13/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024]
Abstract
Ischemia-reperfusion injury is an event concerning any organ under a procedure of transplantation. The early result of ischemia is hypoxia, which causes malfunction of mitochondria and decrease in cellular ATP. This leads to disruption of cellular metabolism. Reperfusion also results in cell damage due to reoxygenation and increased production of reactive oxygen species, and later by induced inflammation. In damaged and hypoxic cells, the endoplasmic reticulum (ER) stress pathway is activated by increased amount of damaged or misfolded proteins, accumulation of free fatty acids and other lipids due to inability of their oxidation (lipotoxicity). ER stress is an adaptive response and a survival pathway, however, its prolonged activity eventually lead to induction of apoptosis. Sustaining cell functionality in stress conditions is a great challenge for transplant surgeons as it is crucial for maintaining a desired level of graft vitality. Pathways counteracting negative consequences of ischemia-reperfusion are autophagy and lipid droplets (LD) metabolism. Autophagy remove damaged organelles and molecules driving them to lysosomes, digested simpler compounds are energy source for the cell. Mitophagy and ER-phagy results in improvement of cell energetic balance and alleviation of ER stress. This is important in sustaining metabolic homeostasis and thus cell survival. LD metabolism is connected with autophagy as LD are degraded by lipophagy, a source of free fatty acids and glycerol-thus autophagy and LD can readily remove lipotoxic compounds in the cell. In conclusion, monitoring and pharmaceutic regulation of those pathways during transplantation procedure might result in increased/improved vitality of transplanted organ.
Collapse
Affiliation(s)
- Daria Kamińska
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Michał Skrzycki
- Chair and Department of Biochemistry, Medical University of Warsaw, Warszawa, Poland
| |
Collapse
|
15
|
Zhang M, Luo X, Zhang B, Luo D, Huang L, Long Q. Unveiling OSCP as the potential therapeutic target for mitochondrial dysfunction-related diseases. Life Sci 2024; 336:122293. [PMID: 38030056 DOI: 10.1016/j.lfs.2023.122293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Mitochondria are important organelles in cells responsible for energy production and regulation. Mitochondrial dysfunction has been implicated in the pathogenesis of many diseases. Oligomycin sensitivity-conferring protein (OSCP), a component of the inner mitochondrial membrane, has been studied for a long time. OSCP is a component of the F1Fo-ATP synthase in mitochondria and is closely related to the regulation of the mitochondrial permeability transition pore (mPTP). Studies have shown that OSCP plays an important role in cardiovascular disease, neurological disorders, and tumor development. This review summarizes the localization, structure, function, and regulatory mechanisms of OSCP and outlines its role in cardiovascular disease, neurological disease, and tumor development. In addition, this article reviews the research on the interaction between OSCP and mPTP. Finally, the article suggests future research directions, including further exploration of the mechanism of action of OSCP, the interaction between OSCP and other proteins and signaling pathways, and the development of new treatment strategies for mitochondrial dysfunction. In conclusion, in-depth research on OSCP will help to elucidate its importance in cell function and disease and provide new ideas for the treatment and prevention of related diseases.
Collapse
Affiliation(s)
- Mingyue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binzhi Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Harris AC, Sun J, Jacobs KM. Concussive Head Trauma Deranges Axon Initial Segment Function in Axotomized and Intact Layer 5 Pyramidal Neurons. J Neurotrauma 2024; 41:244-270. [PMID: 37650832 PMCID: PMC11074420 DOI: 10.1089/neu.2022.0469] [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] [Indexed: 09/01/2023] Open
Abstract
The axon initial segment (AIS) is a critical locus of control of action potential (AP) generation and neuronal information synthesis. Concussive traumatic brain injury gives rise to diffuse axotomy, and the majority of neocortical axonal injury arises at the AIS. Consequently, concussive traumatic brain injury might profoundly disrupt the functional specialization of this region. To investigate this hypothesis, one and two days after mild central fluid percussion injury in Thy1-YFP-H mice, we recorded high-resolution APs from axotomized and adjacent intact layer 5 pyramidal neurons and applied a second derivative (2o) analysis to measure the AIS- and soma-regional contributions to the AP upstroke. All layer 5 pyramidal neurons recorded from sham animals manifested two stark 2o peaks separated by a negative intervening slope. In contrast, within injured mice, we discovered a subset of axotomized layer 5 pyramidal neurons in which the AIS-regional 2o peak was abolished, a functional perturbation associated with diminished excitability, axonal sprouting and distention of the AIS as assessed by staining for ankyrin-G. Our analysis revealed an additional subpopulation of both axotomized and intact layer 5 pyramidal neurons that manifested a melding together of the AIS- and soma-regional 2o peaks, suggesting a more subtle aberration of sodium channel function and/or translocation of the AIS initiation zone closer to the soma. When these experiments were repeated in animals in which cyclophilin-D was knocked out, these effects were ameliorated, suggesting that trauma-induced AIS functional perturbation is associated with mitochondrial calcium dysregulation.
Collapse
Affiliation(s)
- Alan C. Harris
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jianli Sun
- Delaware Center for Neuroscience Research, Delaware State University, Dover, Delaware, USA
| | - Kimberle M. Jacobs
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
17
|
Bortoluzzi VT, Ribeiro RT, Zemniaçak ÂB, Cunha SDA, Sass JO, Castilho RF, Amaral AU, Wajner M. Disturbance of mitochondrial functions caused by N-acetylglutamate and N-acetylmethionine in brain of adolescent rats: Potential relevance in aminoacylase 1 deficiency. Neurochem Int 2023; 171:105631. [PMID: 37852579 DOI: 10.1016/j.neuint.2023.105631] [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: 07/10/2023] [Revised: 09/24/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Aminoacylase 1 (ACY1) deficiency is a rare genetic disorder that affects the breakdown of short-chain aliphatic N-acetylated amino acids, leading to the accumulation of these amino acid derivatives in the urine of patients. Some of the affected individuals have presented with heterogeneous neurological symptoms such as psychomotor delay, seizures, and intellectual disability. Considering that the pathological mechanisms of brain damage in this disorder remain mostly unknown, here we investigated whether major metabolites accumulating in ACY1 deficiency, namely N-acetylglutamate (NAG) and N-acetylmethionine (NAM), could be toxic to the brain by examining their in vitro effects on important mitochondrial properties. We assessed the effects of NAG and NAM on membrane potential, swelling, reducing equivalents, and Ca2+ retention capacity in purified mitochondrial preparations obtained from the brain of adolescent rats. NAG and NAM decreased mitochondrial membrane potential, reducing equivalents, and calcium retention capacity, and induced swelling in Ca2+-loaded brain mitochondria supported by glutamate plus malate. Notably, these changes were completely prevented by the classical inhibitors of mitochondrial permeability transition (MPT) pore cyclosporin A plus ADP and by ruthenium red, implying the participation of MPT and Ca2+ in these effects. Our findings suggest that NAG- and NAM-induced disruption of mitochondrial functions involving MPT may represent relevant mechanisms of neuropathology in ACY1 deficiency.
Collapse
Affiliation(s)
- Vanessa Trindade Bortoluzzi
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Rafael Teixeira Ribeiro
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Ângela Beatris Zemniaçak
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Sâmela de Azevedo Cunha
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Jörn Oliver Sass
- Research Group Inborn Errors of Metabolism, Department of Natural Sciences & Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany.
| | - Roger Frigério Castilho
- Departamento de Patologia, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, Brazil.
| | - Alexandre Umpierrez Amaral
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; PPG Atenção Integral à Saúde, Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, Brazil.
| | - Moacir Wajner
- PPG Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
18
|
Ren K, Pei J, Guo Y, Jiao Y, Xing H, Xie Y, Yang Y, Feng Q, Yang J. Regulated necrosis pathways: a potential target for ischemic stroke. BURNS & TRAUMA 2023; 11:tkad016. [PMID: 38026442 PMCID: PMC10656754 DOI: 10.1093/burnst/tkad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/24/2022] [Indexed: 12/01/2023]
Abstract
Globally, ischemic stroke causes millions of deaths per year. The outcomes of ischemic stroke are largely determined by the amount of ischemia-related and reperfusion-related neuronal death in the infarct region. In the infarct region, cell injuries follow either the regulated pathway involving precise signaling cascades, such as apoptosis and autophagy, or the nonregulated pathway, which is uncontrolled by any molecularly defined effector mechanisms such as necrosis. However, numerous studies have recently found that a certain type of necrosis can be regulated and potentially modified by drugs and is nonapoptotic; this type of necrosis is referred to as regulated necrosis. Depending on the signaling pathway, various elements of regulated necrosis contribute to the development of ischemic stroke, such as necroptosis, pyroptosis, ferroptosis, pathanatos, mitochondrial permeability transition pore-mediated necrosis and oncosis. In this review, we aim to summarize the underlying molecular mechanisms of regulated necrosis in ischemic stroke and explore the crosstalk and interplay among the diverse types of regulated necrosis. We believe that targeting these regulated necrosis pathways both pharmacologically and genetically in ischemia-induced neuronal death and protection could be an efficient strategy to increase neuronal survival and regeneration in ischemic stroke.
Collapse
Affiliation(s)
- Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Yuanyuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yuxue Jiao
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Research Center for Clinical System Biology, Translational Medicine Center, No. 1 Jianshe Dong Road, ErQi District, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
19
|
Nikiforova AB, Baburina YL, Borisova MP, Surin AK, Kharechkina ES, Krestinina OV, Suvorina MY, Kruglova SA, Kruglov AG. Mitochondrial F-ATP Synthase Co-Migrating Proteins and Ca 2+-Dependent Formation of Large Channels. Cells 2023; 12:2414. [PMID: 37830628 PMCID: PMC10572550 DOI: 10.3390/cells12192414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023] Open
Abstract
Monomers, dimers, and individual FOF1-ATP synthase subunits are, presumably, involved in the formation of the mitochondrial permeability transition pore (PTP), whose molecular structure, however, is still unknown. We hypothesized that, during the Ca2+-dependent assembly of a PTP complex, the F-ATP synthase (subunits) recruits mitochondrial proteins that do not interact or weakly interact with the F-ATP synthase under normal conditions. Therefore, we examined whether the PTP opening in mitochondria before the separation of supercomplexes via BN-PAGE will increase the channel stability and channel-forming capacity of isolated F-ATP synthase dimers and monomers in planar lipid membranes. Additionally, we studied the specific activity and the protein composition of F-ATP synthase dimers and monomers from rat liver and heart mitochondria before and after PTP opening. Against our expectations, preliminary PTP opening dramatically suppressed the high-conductance channel activity of F-ATP synthase dimers and monomers and decreased their specific "in-gel" activity. The decline in the channel-forming activity correlated with the reduced levels of as few as two proteins in the bands: methylmalonate-semialdehyde dehydrogenase and prohibitin 2. These results indicate that proteins co-migrating with the F-ATP synthase may be important players in PTP formation and stabilization.
Collapse
Affiliation(s)
- Anna B. Nikiforova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Yulia L. Baburina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Marina P. Borisova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Alexey K. Surin
- Branch of the Shemyakin—Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki 6, 142290 Pushchino, Russia;
- State Research Centre for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, 142290 Pushchino, Russia;
| | - Ekaterina S. Kharechkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Olga V. Krestinina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| | - Maria Y. Suvorina
- Institute of Protein Research, Russian Academy of Sciences, Institutskaya 4, 142290 Pushchino, Russia;
| | - Svetlana A. Kruglova
- Institute of Basic Biological Problems, Russian Academy of Sciences, Institutskaya 2, 142290 Pushchino, Russia;
| | - Alexey G. Kruglov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (A.B.N.); (Y.L.B.); (M.P.B.); (E.S.K.); (O.V.K.)
| |
Collapse
|
20
|
Wang Y, Li J, Han H, Huang H, Du H, Cheng L, Ma C, Cai Y, Li G, Tao J, Cheng P. Application of locally responsive design of biomaterials based on microenvironmental changes in myocardial infarction. iScience 2023; 26:107662. [PMID: 37670787 PMCID: PMC10475519 DOI: 10.1016/j.isci.2023.107662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Morbidity and mortality caused by acute myocardial infarction (AMI) are on the rise, posing a grave threat to the health of the general population. Up to now, interventional, surgical, and pharmaceutical therapies have been the main treatment methods for AMI. Effective and timely reperfusion therapy decreases mortality, but it cannot stimulate myocardial cell regeneration or reverse ventricular remodeling. Cell therapy, gene therapy, immunotherapy, anti-inflammatory therapy, and several other techniques are utilized by researchers to improve patients' prognosis. In recent years, biomaterials for AMI therapy have become a hot spot in medical care. Biomaterials furnish a microenvironment conducive to cell growth and deliver therapeutic factors that stimulate cell regeneration and differentiation. Biomaterials adapt to the complex microenvironment and respond to changes in local physical and biochemical conditions. Therefore, environmental factors and material properties must be taken into account when designing biomaterials for the treatment of AMI. This article will review the factors that need to be fully considered in the design of biological materials.
Collapse
Affiliation(s)
- Yiren Wang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Junlin Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hukui Han
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huihui Huang
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Huan Du
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lianying Cheng
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing 400038, China
| | - Yongxiang Cai
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Gang Li
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jianhong Tao
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu 610072, China
| |
Collapse
|
21
|
Quader M, Akande O, Cholyway R, Lesnefsky EJ, Toldo S, Chen Q. Infarct Size With Incremental Global Myocardial Ischemia Times: Cyclosporine A in Donation After Circulatory Death Rat Hearts. Transplant Proc 2023; 55:1495-1503. [PMID: 37422374 DOI: 10.1016/j.transproceed.2023.03.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/03/2023] [Accepted: 03/30/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND We quantified the myocardial infarct size with varying global ischemia durations and studied the benefits of Cyclosporine A (CyA) in reducing cardiac injury in ex vivo and transplanted rat hearts. METHODS Infarct size was measured after 15, 20, 25, 30, and 35 minutes of in vivo global ischemia (n = 34) and compared with control beating-heart donor (CBD) hearts (n = 10). For heart function assessment, donation after circulatory death (DCD) rat hearts (n = 20) were procured after 25 minutes of in vivo ischemia and reanimated ex vivo for 90 minutes. Half of the DCD hearts received CyA (0.5 mM) at reanimation. The CBD hearts (n = 10) served as controls. A separate group of CBD and DCD (with or without CyA treatment) hearts underwent heterotopic heart transplantation; heart function was measured at 48 hours. RESULTS Infarct size was 25% with 25 minutes of ischemia and increased significantly with 30 and 35 minutes to 32% and 41%, respectively. CyA treatment decreased infarct size in DCD hearts (15% vs 25%). Heart function in the transplanted DCD hearts was significantly better with CyA treatment and was comparable to CBD hearts. CONCLUSIONS CyA administered at reperfusion limited infarct size in DCD hearts and improved their function in transplanted hearts.
Collapse
Affiliation(s)
- Mohammed Quader
- Division of Cardio-Thoracic Surgery, Virginia Commonwealth University, Richmond, Virginia; Department of Surgery, McGuire Veterans Administration Medical Center and Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Oluwatoyin Akande
- Division of Cardio-Thoracic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Renee Cholyway
- Division of Cardio-Thoracic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Surgery, McGuire Veterans Administration Medical Center and Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Stefano Toldo
- Department of Surgery, McGuire Veterans Administration Medical Center and Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Qun Chen
- Department of Surgery, McGuire Veterans Administration Medical Center and Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
22
|
Bround MJ, Havens JR, York AJ, Sargent MA, Karch J, Molkentin JD. ANT-dependent MPTP underlies necrotic myofiber death in muscular dystrophy. SCIENCE ADVANCES 2023; 9:eadi2767. [PMID: 37624892 PMCID: PMC10456852 DOI: 10.1126/sciadv.adi2767] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023]
Abstract
Mitochondrial permeability transition pore (MPTP) formation contributes to ischemia-reperfusion injury in the heart and several degenerative diseases, including muscular dystrophy (MD). MD is a family of genetic disorders characterized by progressive muscle necrosis and premature death. It has been proposed that the MPTP has two molecular components, the adenine nucleotide translocase (ANT) family of proteins and an unknown component that requires the chaperone cyclophilin D (CypD) to activate. This model was examined in vivo by deleting the gene encoding ANT1 (Slc25a4) or CypD (Ppif) in a δ-sarcoglycan (Sgcd) gene-deleted mouse model of MD, revealing that dystrophic mice lacking Slc25a4 were partially protected from cell death and MD pathology. Dystrophic mice lacking both Slc25a4 and Ppif together were almost completely protected from necrotic cell death and MD disease. This study provides direct evidence that ANT1 and CypD are required MPTP components governing in vivo cell death, suggesting a previously unrecognized therapeutic approach in MD and other necrotic diseases.
Collapse
Affiliation(s)
- Michael J. Bround
- Department of Pediatrics, Cincinnati Children's Hospital and the University of Cincinnati, Cincinnati, OH, USA
| | - Julian R. Havens
- Department of Pediatrics, Cincinnati Children's Hospital and the University of Cincinnati, Cincinnati, OH, USA
| | - Allen J. York
- Department of Pediatrics, Cincinnati Children's Hospital and the University of Cincinnati, Cincinnati, OH, USA
| | - Michelle A. Sargent
- Department of Pediatrics, Cincinnati Children's Hospital and the University of Cincinnati, Cincinnati, OH, USA
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jeffery D. Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital and the University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
23
|
Coluccino G, Muraca VP, Corazza A, Lippe G. Cyclophilin D in Mitochondrial Dysfunction: A Key Player in Neurodegeneration? Biomolecules 2023; 13:1265. [PMID: 37627330 PMCID: PMC10452829 DOI: 10.3390/biom13081265] [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: 07/05/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in numerous complex diseases. Understanding the molecular mechanisms by which the "powerhouse of the cell" turns into the "factory of death" is an exciting yet challenging task that can unveil new therapeutic targets. The mitochondrial matrix protein CyPD is a peptidylprolyl cis-trans isomerase involved in the regulation of the permeability transition pore (mPTP). The mPTP is a multi-conductance channel in the inner mitochondrial membrane whose dysregulated opening can ultimately lead to cell death and whose involvement in pathology has been extensively documented over the past few decades. Moreover, several mPTP-independent CyPD interactions have been identified, indicating that CyPD could be involved in the fine regulation of several biochemical pathways. To further enrich the picture, CyPD undergoes several post-translational modifications that regulate both its activity and interaction with its clients. Here, we will dissect what is currently known about CyPD and critically review the most recent literature about its involvement in neurodegenerative disorders, focusing on Alzheimer's Disease and Parkinson's Disease, supporting the notion that CyPD could serve as a promising therapeutic target for the treatment of such conditions. Notably, significant efforts have been made to develop CyPD-specific inhibitors, which hold promise for the treatment of such complex disorders.
Collapse
Affiliation(s)
- Gabriele Coluccino
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (V.P.M.); (A.C.)
| | | | | | - Giovanna Lippe
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy; (V.P.M.); (A.C.)
| |
Collapse
|
24
|
Ashok D, Papanicolaou K, Sidor A, Wang M, Solhjoo S, Liu T, O'Rourke B. Mitochondrial membrane potential instability on reperfusion after ischemia does not depend on mitochondrial Ca 2+ uptake. J Biol Chem 2023; 299:104708. [PMID: 37061004 PMCID: PMC10206190 DOI: 10.1016/j.jbc.2023.104708] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/21/2023] [Accepted: 04/09/2023] [Indexed: 04/17/2023] Open
Abstract
Physiologic Ca2+ entry via the Mitochondrial Calcium Uniporter (MCU) participates in energetic adaption to workload but may also contribute to cell death during ischemia/reperfusion (I/R) injury. The MCU has been identified as the primary mode of Ca2+ import into mitochondria. Several groups have tested the hypothesis that Ca2+ import via MCU is detrimental during I/R injury using genetically-engineered mouse models, yet the results from these studies are inconclusive. Furthermore, mitochondria exhibit unstable or oscillatory membrane potentials (ΔΨm) when subjected to stress, such as during I/R, but it is unclear if the primary trigger is an excess influx of mitochondrial Ca2+ (mCa2+), reactive oxygen species (ROS) accumulation, or other factors. Here, we critically examine whether MCU-mediated mitochondrial Ca2+ uptake during I/R is involved in ΔΨm instability, or sustained mitochondrial depolarization, during reperfusion by acutely knocking out MCU in neonatal mouse ventricular myocyte (NMVM) monolayers subjected to simulated I/R. Unexpectedly, we find that MCU knockout does not significantly alter mCa2+ import during I/R, nor does it affect ΔΨm recovery during reperfusion. In contrast, blocking the mitochondrial sodium-calcium exchanger (mNCE) suppressed the mCa2+ increase during Ischemia but did not affect ΔΨm recovery or the frequency of ΔΨm oscillations during reperfusion, indicating that mitochondrial ΔΨm instability on reperfusion is not triggered by mCa2+. Interestingly, inhibition of mitochondrial electron transport or supplementation with antioxidants stabilized I/R-induced ΔΨm oscillations. The findings are consistent with mCa2+ overload being mediated by reverse-mode mNCE activity and supporting ROS-induced ROS release as the primary trigger of ΔΨm instability during reperfusion injury.
Collapse
Affiliation(s)
- Deepthi Ashok
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Kyriakos Papanicolaou
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Agnieszka Sidor
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Michelle Wang
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Soroosh Solhjoo
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Ting Liu
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA
| | - Brian O'Rourke
- Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
25
|
Murphy E, Liu JC. Mitochondrial calcium and reactive oxygen species in cardiovascular disease. Cardiovasc Res 2023; 119:1105-1116. [PMID: 35986915 PMCID: PMC10411964 DOI: 10.1093/cvr/cvac134] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 08/11/2023] Open
Abstract
Cardiomyocytes are one of the most mitochondria-rich cell types in the body, with ∼30-40% of the cell volume being composed of mitochondria. Mitochondria are well established as the primary site of adenosine triphosphate (ATP) generation in a beating cardiomyocyte, generating up to 90% of its ATP. Mitochondria have many functions in the cell, which could contribute to susceptibility to and development of cardiovascular disease (CVD). Mitochondria are key players in cell metabolism, ATP production, reactive oxygen species (ROS) production, and cell death. Mitochondrial calcium (Ca2+) plays a critical role in many of these pathways, and thus the dynamics of mitochondrial Ca2+ are important in regulating mitochondrial processes. Alterations in these varied and in many cases interrelated functions play an important role in CVD. This review will focus on the interrelationship of mitochondrial energetics, Ca2+, and ROS and their roles in CVD. Recent insights into the regulation and dysregulation of these pathways have led to some novel therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth Murphy
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Julia C Liu
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
26
|
Frigo E, Tommasin L, Lippe G, Carraro M, Bernardi P. The Haves and Have-Nots: The Mitochondrial Permeability Transition Pore across Species. Cells 2023; 12:1409. [PMID: 37408243 PMCID: PMC10216546 DOI: 10.3390/cells12101409] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 07/07/2023] Open
Abstract
The demonstration that F1FO (F)-ATP synthase and adenine nucleotide translocase (ANT) can form Ca2+-activated, high-conductance channels in the inner membrane of mitochondria from a variety of eukaryotes led to renewed interest in the permeability transition (PT), a permeability increase mediated by the PT pore (PTP). The PT is a Ca2+-dependent permeability increase in the inner mitochondrial membrane whose function and underlying molecular mechanisms have challenged scientists for the last 70 years. Although most of our knowledge about the PTP comes from studies in mammals, recent data obtained in other species highlighted substantial differences that could be perhaps attributed to specific features of F-ATP synthase and/or ANT. Strikingly, the anoxia and salt-tolerant brine shrimp Artemia franciscana does not undergo a PT in spite of its ability to take up and store Ca2+ in mitochondria, and the anoxia-resistant Drosophila melanogaster displays a low-conductance, selective Ca2+-induced Ca2+ release channel rather than a PTP. In mammals, the PT provides a mechanism for the release of cytochrome c and other proapoptotic proteins and mediates various forms of cell death. In this review, we cover the features of the PT (or lack thereof) in mammals, yeast, Drosophila melanogaster, Artemia franciscana and Caenorhabditis elegans, and we discuss the presence of the intrinsic pathway of apoptosis and of other forms of cell death. We hope that this exercise may help elucidate the function(s) of the PT and its possible role in evolution and inspire further tests to define its molecular nature.
Collapse
Affiliation(s)
- Elena Frigo
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Via Ugo Bassi 58/B, I-35131 Padova, Italy; (E.F.); (L.T.); (M.C.)
| | - Ludovica Tommasin
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Via Ugo Bassi 58/B, I-35131 Padova, Italy; (E.F.); (L.T.); (M.C.)
| | - Giovanna Lippe
- Department of Medicine, University of Udine, Piazzale Kolbe 4, I-33100 Udine, Italy;
| | - Michela Carraro
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Via Ugo Bassi 58/B, I-35131 Padova, Italy; (E.F.); (L.T.); (M.C.)
| | - Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Via Ugo Bassi 58/B, I-35131 Padova, Italy; (E.F.); (L.T.); (M.C.)
| |
Collapse
|
27
|
Endlicher R, Drahota Z, Štefková K, Červinková Z, Kučera O. The Mitochondrial Permeability Transition Pore-Current Knowledge of Its Structure, Function, and Regulation, and Optimized Methods for Evaluating Its Functional State. Cells 2023; 12:1273. [PMID: 37174672 PMCID: PMC10177258 DOI: 10.3390/cells12091273] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The mitochondrial permeability transition pore (MPTP) is a calcium-dependent, ion non-selective membrane pore with a wide range of functions. Although the MPTP has been studied for more than 50 years, its molecular structure remains unclear. Short-term (reversible) opening of the MPTP protects cells from oxidative damage and enables the efflux of Ca2+ ions from the mitochondrial matrix and cell signaling. However, long-term (irreversible) opening induces processes leading to cell death. Ca2+ ions, reactive oxygen species, and changes in mitochondrial membrane potential regulate pore opening. The sensitivity of the pore to Ca2+ ions changes as an organism ages, and MPTP opening plays a key role in the pathogenesis of many diseases. Most studies of the MPTP have focused on elucidating its molecular structure. However, understanding the mechanisms that will inhibit the MPTP may improve the treatment of diseases associated with its opening. To evaluate the functional state of the MPTP and its inhibitors, it is therefore necessary to use appropriate methods that provide reproducible results across laboratories. This review summarizes our current knowledge of the function and regulation of the MPTP. The latter part of the review introduces two optimized methods for evaluating the functional state of the pore under standardized conditions.
Collapse
Affiliation(s)
- René Endlicher
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic; (R.E.); (Z.Č.)
- Department of Anatomy, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic;
| | - Zdeněk Drahota
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic; (R.E.); (Z.Č.)
- Institute of Physiology, Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Kateřina Štefková
- Department of Anatomy, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic;
| | - Zuzana Červinková
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic; (R.E.); (Z.Č.)
| | - Otto Kučera
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, 500 03 Hradec Králové, Czech Republic; (R.E.); (Z.Č.)
| |
Collapse
|
28
|
Lu P, Liang F, Dong Y, Xie Z, Zhang Y. Sevoflurane Induces a Cyclophilin D-Dependent Decrease of Neural Progenitor Cells Migration. Int J Mol Sci 2023; 24:ijms24076746. [PMID: 37047719 PMCID: PMC10095407 DOI: 10.3390/ijms24076746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
Clinical studies have suggested that repeated exposure to anesthesia and surgery at a young age may increase the risk of cognitive impairment. Our previous research has shown that sevoflurane can affect neurogenesis and cognitive function in young animals by altering cyclophilin D (CypD) levels and mitochondrial function. Neural progenitor cells (NPCs) migration is associated with cognitive function in developing brains. However, it is unclear whether sevoflurane can regulate NPCs migration via changes in CypD. To address this question, we treated NPCs harvested from wild-type (WT) and CypD knockout (KO) mice and young WT and CypD KO mice with sevoflurane. We used immunofluorescence staining, wound healing assay, transwell assay, mass spectrometry, and Western blot to assess the effects of sevoflurane on CypD, reactive oxygen species (ROS), doublecortin levels, and NPCs migration. We showed that sevoflurane increased levels of CypD and ROS, decreased levels of doublecortin, and reduced migration of NPCs harvested from WT mice in vitro and in WT young mice. KO of CypD attenuated these effects, suggesting that a sevoflurane-induced decrease in NPCs migration is dependent on CypD. Our findings have established a system for future studies aimed at exploring the impacts of sevoflurane anesthesia on the impairment of NPCs migration.
Collapse
Affiliation(s)
- Pan Lu
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Feng Liang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Yiying Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
29
|
Próchnicki T, Vasconcelos MB, Robinson KS, Mangan MSJ, De Graaf D, Shkarina K, Lovotti M, Standke L, Kaiser R, Stahl R, Duthie FG, Rothe M, Antonova K, Jenster LM, Lau ZH, Rösing S, Mirza N, Gottschild C, Wachten D, Günther C, Kufer TA, Schmidt FI, Zhong FL, Latz E. Mitochondrial damage activates the NLRP10 inflammasome. Nat Immunol 2023; 24:595-603. [PMID: 36941400 DOI: 10.1038/s41590-023-01451-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
Upon detecting pathogens or cell stress, several NOD-like receptors (NLRs) form inflammasome complexes with the adapter ASC and caspase-1, inducing gasdermin D (GSDMD)-dependent cell death and maturation and release of IL-1β and IL-18. The triggers and activation mechanisms of several inflammasome-forming sensors are not well understood. Here we show that mitochondrial damage activates the NLRP10 inflammasome, leading to ASC speck formation and caspase-1-dependent cytokine release. While the AIM2 inflammasome can also sense mitochondrial demise by detecting mitochondrial DNA (mtDNA) in the cytosol, NLRP10 monitors mitochondrial integrity in an mtDNA-independent manner, suggesting the recognition of distinct molecular entities displayed by the damaged organelles. NLRP10 is highly expressed in differentiated human keratinocytes, in which it can also assemble an inflammasome. Our study shows that this inflammasome surveils mitochondrial integrity. These findings might also lead to a better understanding of mitochondria-linked inflammatory diseases.
Collapse
Affiliation(s)
- Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Kim S Robinson
- A*STAR Skin Research Labs (A*SRL), Clinical Sciences Building, Singapore, Singapore
- Skin Research Institute of Singapore (SRIS), Clinical Sciences Building, Singapore, Singapore
| | - Matthew S J Mangan
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Dennis De Graaf
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kateryna Shkarina
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Marta Lovotti
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lena Standke
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Romina Kaiser
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Rainer Stahl
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Fraser G Duthie
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kateryna Antonova
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Zhi Heng Lau
- A*STAR Skin Research Labs (A*SRL), Clinical Sciences Building, Singapore, Singapore
- Skin Research Institute of Singapore (SRIS), Clinical Sciences Building, Singapore, Singapore
| | - Sarah Rösing
- Department of Dermatology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nora Mirza
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Stuttgart, Stuttgart, Germany
| | - Clarissa Gottschild
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Stuttgart, Stuttgart, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Claudia Günther
- Department of Dermatology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thomas A Kufer
- Institute of Nutritional Medicine, Department of Immunology, University of Hohenheim, Stuttgart, Stuttgart, Germany
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Franklin L Zhong
- Skin Research Institute of Singapore (SRIS), Immunos, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
- German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
30
|
Flores-Romero H, Dadsena S, García-Sáez AJ. Mitochondrial pores at the crossroad between cell death and inflammatory signaling. Mol Cell 2023; 83:843-856. [PMID: 36931255 DOI: 10.1016/j.molcel.2023.02.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 03/18/2023]
Abstract
Mitochondria are cellular organelles with a major role in many cellular processes, including not only energy production, metabolism, and calcium homeostasis but also regulated cell death and innate immunity. Their proteobacterial origin makes them a rich source of potent immune agonists, normally hidden within the mitochondrial membrane barriers. Alteration of mitochondrial permeability through mitochondrial pores thus provides efficient mechanisms not only to communicate mitochondrial stress to the cell but also as a key event in the integration of cellular responses. In this regard, eukaryotic cells have developed diverse signaling networks that sense and respond to the release of mitochondrial components into the cytosol and play a key role in controlling cell death and inflammatory pathways. Modulating pore formation at mitochondria through direct or indirect mechanisms may thus open new opportunities for therapy. In this review, we discuss the current understanding of the structure and molecular mechanisms of mitochondrial pores and how they function at the interface between cell death and inflammatory signaling to regulate cellular outcomes.
Collapse
Affiliation(s)
- Hector Flores-Romero
- Institute for Genetics, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Shashank Dadsena
- Institute for Genetics, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD Research Center, University of Cologne, Cologne, Germany.
| |
Collapse
|
31
|
Protasoni M, Serrano M. Targeting Mitochondria to Control Ageing and Senescence. Pharmaceutics 2023; 15:352. [PMID: 36839673 PMCID: PMC9960816 DOI: 10.3390/pharmaceutics15020352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/24/2023] Open
Abstract
Ageing is accompanied by a progressive impairment of cellular function and a systemic deterioration of tissues and organs, resulting in increased vulnerability to multiple diseases. Here, we review the interplay between two hallmarks of ageing, namely, mitochondrial dysfunction and cellular senescence. The targeting of specific mitochondrial features in senescent cells has the potential of delaying or even reverting the ageing process. A deeper and more comprehensive understanding of mitochondrial biology in senescent cells is necessary to effectively face this challenge. Here, we discuss the main alterations in mitochondrial functions and structure in both ageing and cellular senescence, highlighting the differences and similarities between the two processes. Moreover, we describe the treatments available to target these pathways and speculate on possible future directions of anti-ageing and anti-senescence therapies targeting mitochondria.
Collapse
Affiliation(s)
- Margherita Protasoni
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Cambridge Institute of Science, Altos Labs, Granta Park, Cambridge CB21 6GP, UK
| |
Collapse
|
32
|
Yoon Y, Lee H, Federico M, Sheu SS. Non-conventional mitochondrial permeability transition: Its regulation by mitochondrial dynamics. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2023; 1864:148914. [PMID: 36063902 PMCID: PMC9729414 DOI: 10.1016/j.bbabio.2022.148914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022]
Abstract
Mitochondrial permeability transition (MPT) is a phenomenon that the inner mitochondrial membrane (IMM) loses its selective permeability, leading to mitochondrial dysfunction and cell injury. Electrophysiological evidence indicates the presence of a mega-channel commonly called permeability transition pore (PTP) whose opening is responsible for MPT. However, the molecular identity of the PTP is still under intensive investigations and debates, although cyclophilin D that is inhibited by cyclosporine A (CsA) is the established regulatory component of the PTP. PTP can also open transiently and functions as a rapid mitochondrial Ca2+ releasing mechanism. Mitochondrial fission and fusion, the main components of mitochondrial dynamics, control the number and size of mitochondria, and have been shown to play a role in regulating MPT directly or indirectly. Studies by us and others have indicated the potential existence of a form of transient MPT that is insensitive to CsA. This "non-conventional" MPT is regulated by mitochondrial dynamics and may serve a protective role possibly by decreasing the susceptibility for a frequent or sustained PTP opening; hence, it may have a therapeutic value in many disease conditions involving MPT.
Collapse
Affiliation(s)
- Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta 30912, GA, USA.
| | - Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta 30912, GA, USA
| | - Marilen Federico
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
33
|
Robichaux DJ, Harata M, Murphy E, Karch J. Mitochondrial permeability transition pore-dependent necrosis. J Mol Cell Cardiol 2023; 174:47-55. [PMID: 36410526 PMCID: PMC9868081 DOI: 10.1016/j.yjmcc.2022.11.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
Mitochondrial permeability transition pore (mPTP)-dependent cell death is a form of necrotic cell death that is driven by mitochondrial dysfunction by the opening of the mPTP and is triggered by increases in matrix levels of Ca2+ and reactive oxygen species. This form of cell death has been implicated in ischemic injuries of the heart and brain as well as numerous degenerative diseases in the brain and skeletal muscle. This review focuses on the molecular triggers and regulators of mPTP-dependent necrosis in the context of myocardial ischemia reperfusion injury. Research over the past 50 years has led to the identity of regulators and putative pore-forming components of the mPTP. Finally, downstream consequences of activation of the mPTP as well as ongoing questions and areas of research are discussed. These questions pose a particular interest as targeting the mPTP could potentially represent an efficacious therapeutic strategy to reduce infarct size following an ischemic event.
Collapse
Affiliation(s)
- Dexter J Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mikako Harata
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Elizabeth Murphy
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
34
|
Bernardi P, Carraro M, Lippe G. The mitochondrial permeability transition: Recent progress and open questions. FEBS J 2022; 289:7051-7074. [PMID: 34710270 PMCID: PMC9787756 DOI: 10.1111/febs.16254] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 01/13/2023]
Abstract
Major progress has been made in defining the basis of the mitochondrial permeability transition, a Ca2+ -dependent permeability increase of the inner membrane that has puzzled mitochondrial research for almost 70 years. Initially considered an artefact of limited biological interest by most, over the years the permeability transition has raised to the status of regulator of mitochondrial ion homeostasis and of druggable effector mechanism of cell death. The permeability transition is mediated by opening of channel(s) modulated by matrix cyclophilin D, the permeability transition pore(s) (PTP). The field has received new impulse (a) from the hypothesis that the PTP may originate from a Ca2+ -dependent conformational change of F-ATP synthase and (b) from the reevaluation of the long-standing hypothesis that it originates from the adenine nucleotide translocator (ANT). Here, we provide a synthetic account of the structure of ANT and F-ATP synthase to discuss potential and controversial mechanisms through which they may form high-conductance channels; and review some intriguing findings from the wealth of early studies of PTP modulation that still await an explanation. We hope that this review will stimulate new experiments addressing the many outstanding problems, and thus contribute to the eventual solution of the puzzle of the permeability transition.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience InstituteUniversity of PadovaItaly
| | - Michela Carraro
- Department of Biomedical Sciences and CNR Neuroscience InstituteUniversity of PadovaItaly
| | | |
Collapse
|
35
|
Li A, Yi J, Li X, Dong L, Ostrow LW, Ma J, Zhou J. Deficient Sarcolemma Repair in ALS: A Novel Mechanism with Therapeutic Potential. Cells 2022; 11:cells11203263. [PMID: 36291129 PMCID: PMC9600524 DOI: 10.3390/cells11203263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
The plasma membrane (sarcolemma) of skeletal muscle myofibers is susceptible to injury caused by physical and chemical stresses during normal daily movement and/or under disease conditions. These acute plasma membrane disruptions are normally compensated by an intrinsic membrane resealing process involving interactions of multiple intracellular proteins including dysferlin, annexin, caveolin, and Mitsugumin 53 (MG53)/TRIM72. There is new evidence for compromised muscle sarcolemma repair mechanisms in Amyotrophic Lateral Sclerosis (ALS). Mitochondrial dysfunction in proximity to neuromuscular junctions (NMJs) increases oxidative stress, triggering MG53 aggregation and loss of its function. Compromised membrane repair further worsens sarcolemma fragility and amplifies oxidative stress in a vicious cycle. This article is to review existing literature supporting the concept that ALS is a disease of oxidative-stress induced disruption of muscle membrane repair that compromise the integrity of the NMJs and hence augmenting muscle membrane repair mechanisms could represent a viable therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Li Dong
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Lyle W. Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19122, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| | - Jianjie Ma
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| |
Collapse
|
36
|
SIRT3-Mediated CypD-K166 Deacetylation Alleviates Neuropathic Pain by Improving Mitochondrial Dysfunction and Inhibiting Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4722647. [PMID: 36092157 PMCID: PMC9458368 DOI: 10.1155/2022/4722647] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/21/2022] [Accepted: 08/17/2022] [Indexed: 12/29/2022]
Abstract
Numerous studies have shown that mitochondrial dysfunction manifested by increased mitochondrial permeability transition pore (mPTP) opening and reactive oxygen species (ROS) level, and decreased mitochondrial membrane potential (MMP) plays an important role in the development of neuropathic pain. Sirtuin3 (SIRT3), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, has been shown to inhibit mitochondrial oxidative stress. However, the role of SIRT3 in neuropathic pain is unclear. In this study, we found that the protein and mRNA levels of SIRT3 were significantly downregulated in the spinal cords of spared nerve injury- (SNI-) induced neuropathic pain mice, while overexpression of spinal SIRT3 reversed SNI-induced pain hypersensitivity. Further study showed that SIRT3 overexpression reduced the acetylation level of lysine 166 (K166) on cyclophilin D (CypD), the regulatory component of the mPTP, inhibited the mPTP opening, decreased ROS and malondialdehyde (MDA) levels, and increased MMP and manganese superoxide dismutase (MnSOD) in SNI mice. Point mutation of K166 to arginine on CypD (CypD-K166R) abrogated SNI-induced mitochondrial dysfunction and neuropathic pain in mice. Moreover, inhibiting mPTP opening by cyclosporin A (CsA) improved mitochondrial function and neuropathic pain in SNI mice. Together, these data show that SIRT3 is necessary to prevent neuropathic pain by deacetylating CypD-K166 and further improving mitochondrial dysfunction. This study may shed light on a potential drug target for the treatment of neuropathic pain.
Collapse
|
37
|
Stamerra CA, Di Giosia P, Giorgini P, Ferri C, Sukhorukov VN, Sahebkar A. Mitochondrial Dysfunction and Cardiovascular Disease: Pathophysiology and Emerging Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9530007. [PMID: 35958017 PMCID: PMC9363184 DOI: 10.1155/2022/9530007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Mitochondria ensure the supply of cellular energy through the production of ATP via oxidative phosphorylation. The alteration of this process, called mitochondrial dysfunction, leads to a reduction in ATP and an increase in the production of reactive oxygen species (ROS). Mitochondrial dysfunction can be caused by mitochondrial/nuclear DNA mutations, or it can be secondary to pathological conditions such as cardiovascular disease, aging, and environmental stress. The use of therapies aimed at the prevention/correction of mitochondrial dysfunction, in the context of the specific treatment of cardiovascular diseases, is a topic of growing interest. In this context, the data are conflicting since preclinical studies are numerous, but there are no large randomized studies.
Collapse
Affiliation(s)
- Cosimo Andrea Stamerra
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Di Giosia
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
- Department of Internal Medicine, Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paolo Giorgini
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Claudio Ferri
- University of L'Aquila, Department of Life, Health and Environmental Sciences, Building Delta 6, San Salvatore Hospital, Via Vetoio, Coppito 67100 L'Aquila, Italy
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Hou D, Hu F, Mao Y, Yan L, Zhang Y, Zheng Z, Wu A, Forouzanfar T, Pathak JL, Wu G. Cationic antimicrobial peptide NRC-03 induces oral squamous cell carcinoma cell apoptosis via CypD-mPTP axis-mediated mitochondrial oxidative stress. Redox Biol 2022; 54:102355. [PMID: 35660629 PMCID: PMC9511698 DOI: 10.1016/j.redox.2022.102355] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/14/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023] Open
Abstract
Pleurocidin-family cationic antimicrobial peptide NRC-03 exhibits potent and selective cytotoxicity towards cancer cells. However, the anticancer effect of NRC-03 in oral squamous cell carcinoma (OSCC) and the molecular mechanism of NRC-03 induced cancer cell death is still unclear. This study focused to investigate mitochondrial oxidative stress-mediated altered mitochondrial function involved in NRC-03-induced apoptosis of OSCC cells. NRC-03 entered the OSCC cells more easily than that of normal cells and bound to mitochondria as well as the nucleus, causing cell membrane blebbing, mitochondria swelling, and DNA fragmentation. NRC-03 induced high oxygen consumption, reactive oxygen species (ROS) release, mitochondrial dysfunction, and apoptosis in OSCC cells. Non-specific antioxidant N-acetyl-l-cysteine (NAC), or mitochondria-specific antioxidant mitoquinone (MitoQ) alleviated NRC-03-induced apoptosis and mitochondrial dysfunction indicated that NRC-03 exerts a cytotoxic effect in cancer cells via inducing cellular and mitochondrial oxidative stress. Moreover, the expression of cyclophilin D (CypD), the key component of mitochondrial permeability transition pore (mPTP), was upregulated in NRC-03-treated cancer cells. Blockade of CypD by siRNA-mediated depletion or pharmacological inhibitor cyclosporine A (CsA) significantly suppressed NRC-03-induced mitochondrial oxidative stress, mitochondrial dysfunction, and apoptosis. NRC-03 also activated MAPK/ERK and NF-κB pathways. Importantly, intratumoral administration of NRC-03 inhibited the growth of CAL-27 cells-derived tumors on xenografted animal models. Taken together, our study indicates that NRC-03 induces apoptosis in OSCC cells via the CypD-mPTP axis mediated mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Dan Hou
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China; Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC/VUmc and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, 1081 HZ, the Netherlands
| | - Fengjun Hu
- Institute of Information Technology, Zhejiang Shuren University, Hangzhou, Zhejiang, 310000, China
| | - Yixin Mao
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China; Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China; Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, 1081 HZ, Netherlands
| | - Liang Yan
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yuhui Zhang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Zhichao Zheng
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Antong Wu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China
| | - Tymour Forouzanfar
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC/VUmc and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, 1081 HZ, the Netherlands
| | - Janak L Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC/VUmc and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, 1081 HZ, the Netherlands; Department of Oral Cell Biology, Academic Centre of Dentistry Amsterdam (ACTA), University van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081LA, Netherlands.
| |
Collapse
|
39
|
Quarato G, Llambi F, Guy CS, Min J, Actis M, Sun H, Narina S, Pruett-Miller SM, Peng J, Rankovic Z, Green DR. Ca 2+-mediated mitochondrial inner membrane permeabilization induces cell death independently of Bax and Bak. Cell Death Differ 2022; 29:1318-1334. [PMID: 35726022 DOI: 10.1038/s41418-022-01025-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 12/15/2022] Open
Abstract
The ability of mitochondria to buffer a rapid rise in cytosolic Ca2+ is a hallmark of proper cell homeostasis. Here, we employed m-3M3FBS, a putative phospholipase C (PLC) agonist, to explore the relationships between intracellular Ca2+ imbalance, mitochondrial physiology, and cell death. m-3M3FBS induced a potent dose-dependent Ca2+ release from the endoplasmic reticulum (ER), followed by a rise in intra-mitochondrial Ca2+. When the latter exceeded the organelle buffering capacity, an abrupt mitochondrial inner membrane permeabilization (MIMP) occurred, releasing matrix contents into the cytosol. MIMP was followed by cell death that was independent of Bcl-2 family members and inhibitable by the intracellular Ca2+ chelator BAPTA-AM. Cyclosporin A (CsA), capable of blocking the mitochondrial permeability transition (MPT), completely prevented cell death induced by m-3M3FBS. However, CsA acted upstream of mitochondria by preventing Ca2+ release from ER stores. Therefore, loss of Ca2+ intracellular balance and mitochondrial Ca2+ overload followed by MIMP induced a cell death process that is distinct from Bcl-2 family-regulated mitochondrial outer membrane permeabilization (MOMP). Further, the inhibition of cell death by CsA or its analogues can be independent of effects on the MPT.
Collapse
Affiliation(s)
- Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Fabien Llambi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Relay Therapeutics, Cambridge, MA, 02139, USA
| | - Cliff S Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jaeki Min
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.,Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Marisa Actis
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Huan Sun
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology and The Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology & Therapeutic, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
40
|
NAFLD: Mechanisms, Treatments, and Biomarkers. Biomolecules 2022; 12:biom12060824. [PMID: 35740949 PMCID: PMC9221336 DOI: 10.3390/biom12060824] [Citation(s) in RCA: 200] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed metabolic-associated fatty liver disease (MAFLD), is one of the most common causes of liver diseases worldwide. NAFLD is growing in parallel with the obesity epidemic. No pharmacological treatment is available to treat NAFLD, specifically. The reason might be that NAFLD is a multi-factorial disease with an incomplete understanding of the mechanisms involved, an absence of accurate and inexpensive imaging tools, and lack of adequate non-invasive biomarkers. NAFLD consists of the accumulation of excess lipids in the liver, causing lipotoxicity that might progress to metabolic-associated steatohepatitis (NASH), liver fibrosis, and hepatocellular carcinoma. The mechanisms for the pathogenesis of NAFLD, current interventions in the management of the disease, and the role of sirtuins as potential targets for treatment are discussed here. In addition, the current diagnostic tools, and the role of non-coding RNAs as emerging diagnostic biomarkers are summarized. The availability of non-invasive biomarkers, and accurate and inexpensive non-invasive diagnosis tools are crucial in the detection of the early signs in the progression of NAFLD. This will expedite clinical trials and the validation of the emerging therapeutic treatments.
Collapse
|
41
|
ATG7-enhanced impaired autophagy exacerbates acute pancreatitis by promoting regulated necrosis via the miR-30b-5p/CAMKII pathway. Cell Death Dis 2022; 13:211. [PMID: 35256590 PMCID: PMC8901675 DOI: 10.1038/s41419-022-04657-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 01/18/2023]
Abstract
The present study was performed to explore whether and how impaired autophagy could modulate calcium/calmodulin-dependent protein kinase II (CAMKII)-regulated necrosis in the pathogenesis of acute pancreatitis (AP). Wistar rats and AR42J cells were used for AP modeling. When indicated, genetic regulation of CAMKII or ATG7 was performed prior to AP induction. AP-related necrotic injury was positively regulated by the incubation level of CAMKII. ATG7 positively modulated the level of CAMKII and necrosis following AP induction, indicating that there might be a connection between impaired autophagy and CAMKII-regulated necrosis in the pathogenesis of AP. microRNA (miR)-30b-5p was predicted and then verified as the upstream regulator of CAMKII mRNA in our setting of AP. Given that the level of miR-30b-5p was negatively correlated with the incubation levels of ATG7 after AP induction, a rescue experiment was performed and indicated that the miR-30b-5p mimic compromised ATG7 overexpression-induced upregulation of CAMKII-regulated necrosis after AP induction. In conclusion, our results indicate that ATG7-enhanced impaired autophagy exacerbates AP by promoting regulated necrosis via the miR-30b-5p/CAMKII pathway.
Collapse
|
42
|
Boyenle ID, Oyedele AK, Ogunlana AT, Adeyemo AF, Oyelere FS, Akinola OB, Adelusi TI, Ehigie LO, Ehigie AF. Targeting the mitochondrial permeability transition pore for drug discovery: Challenges and opportunities. Mitochondrion 2022; 63:57-71. [PMID: 35077882 DOI: 10.1016/j.mito.2022.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/29/2022]
Abstract
Several drug targets have been amenable to drug discovery pursuit not until the characterization of the mitochondrial permeability transition pore (MPTP), a pore with an undefined molecular identity that forms on the inner mitochondrial membrane upon mitochondrial permeability transition (MPT) under the influence of calcium overload and oxidative stress. The opening of the pore which is presumed to cause cell death in certain human diseases also has implications under physiological parlance. Different models for this pore have been postulated following its first identification in the last six decades. The mitochondrial community has witnessed many protein candidates such as; voltage-dependent anion channel (VDAC), adenine nucleotide translocase (ANT), Mitochondrial phosphate carrier (PiC), Spastic Paralegin (SPG7), disordered proteins, and F1Fo ATPase. However, genetic studies have cast out most of these candidates with only F1Fo ATPase currently under intense argument. Cyclophilin D (CyPD) remains the widely accepted positive regulator of the MPTP known to date, but no drug candidate has emerged as its inhibitor, raising concern issues for therapeutics. Thus, in this review, we discuss various models of MPTP reported with the hope of stimulating further research in this field. We went beyond the classical description of the MPTP to ascribe a 'two-edged sword property' to the pore for therapeutic function in human disease because its inhibition and activation have pharmacological relevance. We suggested putative proteins upstream to CyPD that can regulate its activity and prevent cell deaths in neurodegenerative disease and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ibrahim Damilare Boyenle
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdulquddus Kehinde Oyedele
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdeen Tunde Ogunlana
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Aishat Folashade Adeyemo
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Olateju Balikis Akinola
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Temitope Isaac Adelusi
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Leonard Ona Ehigie
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Adeola Folasade Ehigie
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
43
|
Zhang S, Rao S, Yang M, Ma C, Hong F, Yang S. Role of Mitochondrial Pathways in Cell Apoptosis during He-Patic Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23042357. [PMID: 35216473 PMCID: PMC8877300 DOI: 10.3390/ijms23042357] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sen Zhang
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Sijing Rao
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Meiwen Yang
- Department of Surgery, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China;
| | - Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Fengfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
44
|
Cyclophilin D regulation of the mitochondrial permeability transition pore. CURRENT OPINION IN PHYSIOLOGY 2022; 25:100486. [PMID: 35296110 PMCID: PMC8920311 DOI: 10.1016/j.cophys.2022.100486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The opening of the mitochondrial permeability transition pore (PTP) has been proposed to play a critical role in activating cell death in many settings, including cardiac ischemia and reperfusion. Although the identity of pore forming unit of the PTP is still debated, it is generally agreed that cyclophilin D (CyPD) is a regulator of the PTP. This manuscript will focus on understanding how CyPD might regulate the PTP and how understanding CyPD might give insight about the identify and regulation of the PTP.
Collapse
|
45
|
Abdelmoaty AAA, Zhang P, Lin W, Fan YJ, Ye SN, Xu JH. C0818, a novel curcumin derivative, induces ROS-dependent cytotoxicity in human hepatocellular carcinoma cells in vitro via disruption of Hsp90 function. Acta Pharmacol Sin 2022; 43:446-456. [PMID: 33824458 PMCID: PMC8792041 DOI: 10.1038/s41401-021-00642-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/08/2021] [Indexed: 02/03/2023]
Abstract
Heat shock protein 90 (Hsp90) is the most common molecular chaperone that controls the maturation of many oncoproteins critical in tumor development. Hsp90 has been considered as a promising target for cancer treatment, but the clinical significance of Hsp90 and the mechanisms of Hsp90 regulating the tumor-promoting effects in hepatocellular carcinoma (HCC) remain obscure. Previous studies have shown that curcumin, a polyphenol derived from the plant turmeric (Curcuma longa), inhibits tumor growth, which may provide an effective alternative therapy for HCC. Compared to curcumin, a novel derivative of curcumin, 3,5-(E)-Bis(3-methoxy-4-hydroxybenzal)-4-piperidinone hydrochloride (C0818) that is more potent in Hsp90 inhibition and antitumor activity. In this study, we investigated the effect of C0818 on HCC cells in vitro and its relation to Hsp90 inhibition. We showed that C0818 concentration-dependently inhibited the proliferation, the colony formation and induced apoptosis in HepG2 and Sk-Hep-1 cells. C0818 concentration-dependently inhibited DNA synthesis and induced G2/M phase arrest in HepG2 and Sk-Hep-1 cells. We further demonstrated that C0818 induced ROS- and caspase-dependent apoptosis in HCC cells through the mitochondrial-mediated pathway. C0818 induced the degradation of Hsp90 client proteins as RAS, C-Raf, P-C-Raf, Erk, P-ERK, MEK, P-MEK, Akt and P-Akt, which led to subsequent inhibition of the RAS/RAF/MEK/ERK and PI3K/AKT pathways. We revealed that C0818 could inhibit the binding of Hsp90 with its clients without affecting their transcription, which subsequently induced the degradation of Hsp90 clients by the proteasome rather than the lysosome. These results are of potential importance for elucidating a novel Hsp90 inhibitor targeting HCC.
Collapse
Affiliation(s)
- Ahmed Attia Ahmed Abdelmoaty
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, China
| | - Ping Zhang
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, China
| | - Wen Lin
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Ying-Juan Fan
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, China
| | - Sheng-Nan Ye
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
| | - Jian-Hua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
46
|
Kobayashi T, Uchino H, Elmér E, Ogihara Y, Fujita H, Sekine S, Ishida Y, Saiki I, Shibata S, Kawachi A. Disease Outcome and Brain Metabolomics of Cyclophilin-D Knockout Mice in Sepsis. Int J Mol Sci 2022; 23:961. [PMID: 35055146 PMCID: PMC8779771 DOI: 10.3390/ijms23020961] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction resulting from a systemic inflammatory response to infection, but the mechanism remains unclear. The mitochondrial permeability transition pore (MPTP) could play a central role in the neuronal dysfunction, induction of apoptosis, and cell death in SAE. The mitochondrial isomerase cyclophilin D (CypD) is known to control the sensitivity of MPTP induction. We, therefore, established a cecal ligation and puncture (CLP) model, which is the gold standard in sepsis research, using CypD knockout (CypD KO) mice, and analyzed the disease phenotype and the possible molecular mechanism of SAE through metabolomic analyses of brain tissue. A comparison of adult, male wild-type, and CypD KO mice demonstrated statistically significant differences in body temperature, mortality, and histological changes. In the metabolomic analysis, the main finding was the maintenance of reduced glutathione (GSH) levels and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio in the KO animals following CLP. In conclusion, we demonstrate that CypD is implicated in the pathogenesis of SAE, possibly related to the inhibition of MPTP induction and, as a consequence, the decreased production of ROS and other free radicals, thereby protecting mitochondrial and cellular function.
Collapse
Affiliation(s)
- Takayuki Kobayashi
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Hiroyuki Uchino
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Eskil Elmér
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden;
| | - Yukihiko Ogihara
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Hidetoshi Fujita
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan;
| | - Shusuke Sekine
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Yusuke Ishida
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Iwao Saiki
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Shoichiro Shibata
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| | - Aya Kawachi
- Department of Anesthesiology, Tokyo Medical University, Tokyo 160-0023, Japan; (H.U.); (Y.O.); (S.S.); (Y.I.); (I.S.); (S.S.); (A.K.)
| |
Collapse
|
47
|
Gierhardt M, Pak O, Sydykov A, Kraut S, Schäffer J, Garcia C, Veith C, Zeidan EM, Brosien M, Quanz K, Esfandiary A, Saraji A, Hadzic S, Kojonazarov B, Wilhelm J, Ghofrani HA, Schermuly RT, Seeger W, Grimminger F, Herden C, Schulz R, Weissmann N, Heger J, Sommer N. Genetic deletion of p66shc and/or cyclophilin D results in decreased pulmonary vascular tone. Cardiovasc Res 2022; 118:305-315. [PMID: 33119054 PMCID: PMC8752355 DOI: 10.1093/cvr/cvaa310] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 10/15/2020] [Indexed: 11/13/2022] Open
Abstract
AIMS The pulmonary vascular tone and hypoxia-induced alterations of the pulmonary vasculature may be regulated by the mitochondrial membrane permeability transition pore (mPTP) that controls mitochondrial calcium load and apoptosis. We thus investigated, if the mitochondrial proteins p66shc and cyclophilin D (CypD) that regulate mPTP opening affect the pulmonary vascular tone. METHODS AND RESULTS Mice deficient for p66shc (p66shc-/-), CypD (CypD-/-), or both proteins (p66shc/CypD-/-) exhibited decreased pulmonary vascular resistance (PVR) compared to wild-type mice determined in isolated lungs and in vivo. In contrast, systemic arterial pressure was only lower in CypD-/- mice. As cardiac function and pulmonary vascular remodelling did not differ between genotypes, we determined alterations of vascular contractility in isolated lungs and calcium handling in pulmonary arterial smooth muscle cells (PASMC) as underlying reason for decreased PVR. Potassium chloride (KCl)-induced pulmonary vasoconstriction and KCl-induced cytosolic calcium increase determined by Fura-2 were attenuated in all gene-deficient mice. In contrast, KCl-induced mitochondrial calcium increase determined by the genetically encoded Mito-Car-GECO and calcium retention capacity were increased only in CypD-/- and p66shc/CypD-/- mitochondria indicating that decreased mPTP opening affected KCl-induced intracellular calcium peaks in these cells. All mouse strains showed a similar pulmonary vascular response to chronic hypoxia, while acute hypoxic pulmonary vasoconstriction was decreased in gene-deficient mice indicating that CypD and p66shc regulate vascular contractility but not remodelling. CONCLUSIONS We conclude that p66shc specifically regulates the pulmonary vascular tone, while CypD also affects systemic pressure. However, only CypD acts via regulation of mPTP opening and mitochondrial calcium regulation.
Collapse
MESH Headings
- Animals
- Arterial Pressure
- Calcium/metabolism
- Calcium Signaling
- Cell Proliferation
- Cells, Cultured
- Peptidyl-Prolyl Isomerase F/deficiency
- Peptidyl-Prolyl Isomerase F/genetics
- Disease Models, Animal
- Gene Deletion
- Hypertension, Pulmonary/enzymology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Hypoxia/complications
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/enzymology
- Mitochondria/genetics
- Mitochondrial Permeability Transition Pore/metabolism
- Pulmonary Artery/enzymology
- Pulmonary Artery/physiopathology
- Src Homology 2 Domain-Containing, Transforming Protein 1/deficiency
- Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
- Vascular Remodeling
- Vascular Resistance
- Vasoconstriction
- Mice
Collapse
Affiliation(s)
- Mareike Gierhardt
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Oleg Pak
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Simone Kraut
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Julia Schäffer
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Claudia Garcia
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Christine Veith
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Esraa M Zeidan
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Monika Brosien
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Karin Quanz
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Azadeh Esfandiary
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Alireza Saraji
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Stefan Hadzic
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Baktybek Kojonazarov
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Jochen Wilhelm
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Department of Medicine, Imperial College London, Du Cane Road, Hammersmith Campus, London, W12 0NN, UK
| | - Ralph T Schermuly
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Werner Seeger
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Friedrich Grimminger
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus-Liebig University, Giessen, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Jacqueline Heger
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| |
Collapse
|
48
|
Sartori MR, Navarro CDC, Castilho RF, Vercesi AE. Enhanced resistance to Ca2+-induced mitochondrial permeability transition in the long-lived red-footed tortoise Chelonoidis carbonaria. J Exp Biol 2022; 225:jeb243532. [PMID: 34904632 DOI: 10.1242/jeb.243532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022]
Abstract
The interaction between supraphysiological cytosolic Ca2+ levels and mitochondrial redox imbalance mediates the mitochondrial permeability transition (MPT). The MPT is involved in cell death, diseases and aging. This study compared the liver mitochondrial Ca2+ retention capacity and oxygen consumption in the long-lived red-footed tortoise (Chelonoidis carbonaria) with those in the rat as a reference standard. Mitochondrial Ca2+ retention capacity, a quantitative measure of MPT sensitivity, was remarkably higher in tortoises than in rats. This difference was minimized in the presence of the MPT inhibitors ADP and cyclosporine A. However, the Ca2+ retention capacities of tortoise and rat liver mitochondria were similar when both MPT inhibitors were present simultaneously. NADH-linked phosphorylating respiration rates of tortoise liver mitochondria represented only 30% of the maximal electron transport system capacity, indicating a limitation imposed by the phosphorylation system. These results suggested underlying differences in putative MPT structural components [e.g. ATP synthase, adenine nucleotide translocase (ANT) and cyclophilin D] between tortoises and rats. Indeed, in tortoise mitochondria, titrations of inhibitors of the oxidative phosphorylation components revealed a higher limitation of ANT. Furthermore, cyclophilin D activity was approximately 70% lower in tortoises than in rats. Investigation of critical properties of mitochondrial redox control that affect MPT demonstrated that tortoise and rat liver mitochondria exhibited similar rates of H2O2 release and glutathione redox status. Overall, our findings suggest that constraints imposed by ANT and cyclophilin D, putative components or regulators of the MPT pore, are associated with the enhanced resistance to Ca2+-induced MPT in tortoises.
Collapse
Affiliation(s)
- Marina R Sartori
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | - Claudia D C Navarro
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | - Roger F Castilho
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | - Anibal E Vercesi
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| |
Collapse
|
49
|
Hinrichsen F, Hamm J, Westermann M, Schröder L, Shima K, Mishra N, Walker A, Sommer N, Klischies K, Prasse D, Zimmermann J, Kaiser S, Bordoni D, Fazio A, Marinos G, Laue G, Imm S, Tremaroli V, Basic M, Häsler R, Schmitz RA, Krautwald S, Wolf A, Stecher B, Schmitt-Kopplin P, Kaleta C, Rupp J, Bäckhed F, Rosenstiel P, Sommer F. Microbial regulation of hexokinase 2 links mitochondrial metabolism and cell death in colitis. Cell Metab 2021; 33:2355-2366.e8. [PMID: 34847376 DOI: 10.1016/j.cmet.2021.11.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/07/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022]
Abstract
Hexokinases (HK) catalyze the first step of glycolysis limiting its pace. HK2 is highly expressed in gut epithelium, contributes to immune responses, and is upregulated during inflammation. We examined the microbial regulation of HK2 and its impact on inflammation using mice lacking HK2 in intestinal epithelial cells (Hk2ΔIEC). Hk2ΔIEC mice were less susceptible to acute colitis. Analyzing the epithelial transcriptome from Hk2ΔIEC mice during colitis and using HK2-deficient intestinal organoids and Caco-2 cells revealed reduced mitochondrial respiration and epithelial cell death in the absence of HK2. The microbiota strongly regulated HK2 expression and activity. The microbially derived short-chain fatty acid (SCFA) butyrate repressed HK2 expression via histone deacetylase 8 (HDAC8) and reduced mitochondrial respiration in wild-type but not in HK2-deficient Caco-2 cells. Butyrate supplementation protected wild-type but not Hk2ΔIEC mice from colitis. Our findings define a mechanism how butyrate promotes intestinal homeostasis and suggest targeted HK2-inhibition as therapeutic avenue for inflammation.
Collapse
Affiliation(s)
- Finn Hinrichsen
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Jacob Hamm
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Magdalena Westermann
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Lena Schröder
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - Neha Mishra
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Centre for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Nina Sommer
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Kenneth Klischies
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Daniela Prasse
- Institute of General Microbiology, University of Kiel, 24118 Kiel, Germany
| | | | - Sina Kaiser
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Dora Bordoni
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Antonella Fazio
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Georgios Marinos
- Institute of Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Georg Laue
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Simon Imm
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany; Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Ruth A Schmitz
- Institute of General Microbiology, University of Kiel, 24118 Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Andrea Wolf
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany; German Center for Infection Research (DZIF), partner site LMU Munich, Munich Germany
| | - Philippe Schmitt-Kopplin
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München, German Research Centre for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Christoph Kaleta
- Institute of Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany
| | - Felix Sommer
- Institute of Clinical Molecular Biology, University of Kiel, 24105 Kiel, Germany.
| |
Collapse
|
50
|
Martens MD, Karch J, Gordon JW. The molecular mosaic of regulated cell death in the cardiovascular system. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166297. [PMID: 34718119 DOI: 10.1016/j.bbadis.2021.166297] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/07/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022]
Abstract
Cell death is now understood to be a highly regulated process that contributes to normal development and tissue homeostasis, alongside its role in the etiology of various pathological conditions. Through detailed molecular analysis, we have come to know that all cells do not always die in the same way, and that there are at least 7 processes involved, including: apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, and autophagy-mediated cell death. These processes act as pieces in the mosaic of cardiomyocyte cell death, which come together depending on context and stimulus. This review details each individual process, as well as highlights how they come together to produce various cardiac pathologies. By knowing how the pieces go together we can aim towards the development of efficacious therapeutics, which will enable us to prevent cardiomyocyte loss in the face of stress, both reducing mortality and improving quality of life.
Collapse
Affiliation(s)
- Matthew D Martens
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada; The Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Canada
| | - Jason Karch
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Joseph W Gordon
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada; College of Nursing, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Manitoba, Canada; The Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Canada.
| |
Collapse
|