1
|
Qu Y, Liu ZX, Zheng XX, Wu SN, An JQ, Zou MH, Zhang ZR. MFN2-mediated decrease in mitochondria-associated endoplasmic reticulum membranes contributes to sunitinib-induced endothelial dysfunction and hypertension. J Mol Cell Cardiol 2025; 200:45-60. [PMID: 39848488 DOI: 10.1016/j.yjmcc.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/30/2024] [Revised: 12/04/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Treatment of cancer patients with tyrosine kinase inhibitors (TKIs) often results in hypertension, but the underlying mechanism remains unclear. This study aimed to examine the role of mitochondrial morphology and function, particularly mitochondria-associated endoplasmic reticulum membranes (MAMs), in sunitinib-induced hypertension. METHODS Both in vitro and in vivo experiments performed to assesse reactive oxygen species (ROS), nitric oxide (NO), endothelium-dependent vasorelaxation, systemic blood pressure, and mitochondrial function in human umbilical vein endothelial cells (HUVECs) and C57BL/6 mouse aortic endothelial cells, under vehicle or sunitinib treatment condition. RESULTS Sunitinib increased mitochondrial ROS accumulation, decreased oxygen consumption rate, ATP production, and mitochondrial calcium ([Ca2+]M) levels, and impaired endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) signaling in HUVECs. In addition, sunitinib also decreased mitochondrial membrane potential, elongated mitochondria, and reduced MAMs. Remarkably, these effects were reversed by an adeno-virus linker (Ad-linker) that reinforces MAMs. Engineered augmentation of MAMs using AAV-FLT1-linker significantly mitigated sunitinib-induced hypertension, by restoring endothelium-dependent relaxation in mice, highlighting the crucial role of MAMs in this process. Further analyses revealed that sunitinib enhanced Akt-mediated expression of mitofusin 2 (MFN2), causing mitochondrial elongation, and induced dephosphorylation of inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) at residues Y1737/Y1738, reducing [Ca2+]M. Our study suggests that increased MFN2 expression and IP3R1 dephosphorylation are critical in sunitinib-induced MAMs reduction and [Ca2+]M homeostasis. CONCLUSION Sunitinib induces mitochondrial dysfunction, Akt/MFN2-mediated decrease in MAMs and mitochondrial elongation, and IP3R1 dephosphorylation in endothelial cells, leading to endothelial dysfunction and hypertension. Our results provide the potential therapeutic targets for combating TKI-induced hypertension.
Collapse
Affiliation(s)
- Yao Qu
- Department of Cardiology, Harbin Medical University Cancer Hospital, NHC Key Laboratory of Cell Transplantation, Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Institute of Metabolic Disease, Heilongjiang Academy of Medical Sciences, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Zhi-Xue Liu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Xiao-Xu Zheng
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Sheng-Nan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Jun-Qing An
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA
| | - Ming-Hui Zou
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China; Center for Molecular and Translational Medicine, Georgia State University, Atlanta, USA.
| | - Zhi-Ren Zhang
- Department of Cardiology, Harbin Medical University Cancer Hospital, NHC Key Laboratory of Cell Transplantation, Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Institute of Metabolic Disease, Heilongjiang Academy of Medical Sciences, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin, China.
| |
Collapse
|
2
|
Ronderos V, Bollag WB. Therapeutic benefits of glycerol in dry eye disease. Front Med (Lausanne) 2025; 11:1531670. [PMID: 39882517 PMCID: PMC11774691 DOI: 10.3389/fmed.2024.1531670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Dry eye disease (DED) is one of the most commonly diagnosed eye disorders, with a prevalence ranging from 5 to 50%, depending on the geographic location. DED is a multifactorial disorder of the tears and ocular surface, which results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is also accompanied by increased osmolarity of the tear film and inflammation of the surface of the eye. Multiple therapeutic agents have been used to treat DED, with glycerol emerging as a promising component of ophthalmic preparations, due to its humectant, lubricant, demulcent, and osmoprotective properties. This review aims to evaluate the current evidence concerning the therapeutic benefits of glycerol in managing DED, focusing on its possible mechanisms of action, clinical efficacy, and potential advantages over other treatments.
Collapse
Affiliation(s)
- Victoria Ronderos
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Charlie Norwood VA Medical Center, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
3
|
Singh A, Bocher O, Zeggini E. Insights into the molecular underpinning of type 2 diabetes complications. Hum Mol Genet 2025:ddae203. [PMID: 39807636 DOI: 10.1093/hmg/ddae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Type 2 diabetes (T2D) complications pose a significant global health challenge. Omics technologies have been employed to investigate these complications and identify the biological pathways involved. In this review, we focus on four major T2D complications: diabetic kidney disease, diabetic retinopathy, diabetic neuropathy, and cardiovascular complications. We discuss advancements in omics research, summarizing findings from genetic, epigenomic, transcriptomic, proteomic, and metabolomic studies across different ancestries and disease-relevant tissues. We stress the importance of integrating multi-omics techniques to elucidate the biological mechanisms underlying T2D complications and advocate for ancestrally diverse studies. Ultimately, these insights will improve risk prediction for T2D complications and inform translation strategies.
Collapse
Affiliation(s)
- Archit Singh
- Technical University of Munich (TUM), TUM School of Medicine and Health, Graduate School of Experimental Medicine and Health Sciences, Ismaninger Straße 22, Munich 81675, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
- Munich School for Data Science (MUDS), Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München- German Research Center for Environmental Health, Ingolstädter Landstraße 1, Neuherberg 85764, Germany
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Ismaninger Straße 22, Munich 81675, Germany
| |
Collapse
|
4
|
Vassallo N. Poration of mitochondrial membranes by amyloidogenic peptides and other biological toxins. J Neurochem 2025; 169:e16213. [PMID: 39213385 DOI: 10.1111/jnc.16213] [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] [Academic Contribution Register] [Received: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria are essential organelles known to serve broad functions, including in cellular metabolism, calcium buffering, signaling pathways and the regulation of apoptotic cell death. Maintaining the integrity of the outer (OMM) and inner mitochondrial membranes (IMM) is vital for mitochondrial health. Cardiolipin (CL), a unique dimeric glycerophospholipid, is the signature lipid of energy-converting membranes. It plays a significant role in maintaining mitochondrial architecture and function, stabilizing protein complexes and facilitating efficient oxidative phosphorylation (OXPHOS) whilst regulating cytochrome c release from mitochondria. CL is especially enriched in the IMM and at sites of contact between the OMM and IMM. Disorders of protein misfolding, such as Alzheimer's and Parkinson's diseases, involve amyloidogenic peptides like amyloid-β, tau and α-synuclein, which form metastable toxic oligomeric species that interact with biological membranes. Electrophysiological studies have shown that these oligomers form ion-conducting nanopores in membranes mimicking the IMM's phospholipid composition. Poration of mitochondrial membranes disrupts the ionic balance, causing osmotic swelling, loss of the voltage potential across the IMM, release of pro-apoptogenic factors, and leads to cell death. The interaction between CL and amyloid oligomers appears to favour their membrane insertion and pore formation, directly implicating CL in amyloid toxicity. Additionally, pore formation in mitochondrial membranes is not limited to amyloid proteins and peptides; other biological peptides, as diverse as the pro-apoptotic Bcl-2 family members, gasdermin proteins, cobra venom cardiotoxins and bacterial pathogenic toxins, have all been described to punch holes in mitochondria, contributing to cell death processes. Collectively, these findings underscore the vulnerability of mitochondria and the involvement of CL in various pathogenic mechanisms, emphasizing the need for further research on targeting CL-amyloid interactions to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Tal-Qroqq, Malta
| |
Collapse
|
5
|
Senko D, Efimova O, Osetrova M, Anikanov N, Boyko M, Sharaev M, Morozova A, Zorkina Y, Kislov M, Kostyuk G, Stekolshchikova E, Khaitovich P. White matter lipidome alterations in the schizophrenia brain. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:123. [PMID: 39725684 DOI: 10.1038/s41537-024-00542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Numerous brain imaging studies have reported white matter alterations in schizophrenia, but the lipidome analysis of the corresponding tissue remains incomplete. In this study, we investigated the lipidome composition of six subcortical white matter regions corresponding to major axonal tracks in both control subjects and schizophrenia patients. All six regions exhibited a consistent pattern of quantitative lipidome alterations in schizophrenia, involving myelin-forming and mitochondria associated lipid classes. While alteration levels of myelin-forming lipids, particularly sphingolipids, aligned with the extent of the myelin changes reported in structural brain imaging studies, a significant decrease of mitochondria in the white matter, indicated by the lipidome alterations, was not previously investigated. To verify this effect, we performed lipidome analysis in a larger set of individuals and in the mitochondria-enriched membrane fraction, as well as directly quantified mitochondrial content. Our results suggest a substantial reduction of the mitochondrial quotient accompanied by the imbalance in myelin lipids in schizophrenia white matter.
Collapse
Affiliation(s)
- Dmitry Senko
- Skolkovo Institute of Science and Technology, Moscow, Russia.
| | - Olga Efimova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Maria Osetrova
- Skolkovo Institute of Science and Technology, Moscow, Russia
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
| | | | - Maria Boyko
- Skolkovo Institute of Science and Technology, Moscow, Russia
- BIMAI-lab, Sharjah, UAE
| | - Maksim Sharaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- BIMAI-lab, Sharjah, UAE
| | - Anna Morozova
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Yana Zorkina
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Maksim Kislov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Georgiy Kostyuk
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | | |
Collapse
|
6
|
Blumstein DM, MacManes MD. Impacts of dietary fat on multi tissue gene expression in the desert-adapted cactus mouse. J Exp Biol 2024; 227:jeb247978. [PMID: 39676723 PMCID: PMC11698062 DOI: 10.1242/jeb.247978] [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] [Academic Contribution Register] [Received: 05/07/2024] [Accepted: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Understanding the relationship between dietary fat and physiological responses is crucial in species adapted to arid environments where water scarcity is common. In this study, we present a comprehensive exploration of gene expression across five tissues (kidney, liver, lung, gastrointestinal tract and hypothalamus) and 17 phenotypic measurements, investigating the effects of dietary fat in the desert-adapted cactus mouse (Peromyscus eremicus). We show impacts on immune function, circadian gene regulation and mitochondrial function for mice fed a lower-fat diet compared with mice fed a higher-fat diet. In arid environments with severe water scarcity, even subtle changes in organismal health and water balance can affect physical performance, potentially impacting survival and reproductive success. This study sheds light on the complex interplay between diet, physiological processes and environmental adaptation, providing valuable insights into the multifaceted impacts of dietary choices on organismal well-being and adaptation strategies in arid habitats.
Collapse
Affiliation(s)
- Danielle M. Blumstein
- University of New Hampshire, Molecular, Cellular, and Biomedical Sciences Department, Durham, NH 03824, USA
| | - Matthew D. MacManes
- University of New Hampshire, Molecular, Cellular, and Biomedical Sciences Department, Durham, NH 03824, USA
| |
Collapse
|
7
|
Pascuali N, Tobias F, Valyi-Nagy K, Salih S, Veiga-Lopez A. Delineating lipidomic landscapes in human and mouse ovaries: Spatial signatures and chemically-induced alterations via MALDI mass spectrometry imaging: Spatial ovarian lipidomics. ENVIRONMENT INTERNATIONAL 2024; 194:109174. [PMID: 39644787 DOI: 10.1016/j.envint.2024.109174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/26/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
This study addresses the critical gap in understanding the ovarian lipidome's abundance, distribution, and vulnerability to environmental disruptors, a largely unexplored field. Leveraging the capabilities of matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI MSI), we embarked on a novel exploration of the ovarian lipidome in both mouse and human healthy tissues. Our findings revealed that the obesogenic chemical tributyltin (TBT), at environmentally relevant exposures, exerts a profound and region-specific impact on the mouse ovarian lipidome. TBT exposure predominantly affects lipid species in antral follicles and oocytes, suggesting a targeted disruption of lipid homeostasis in these biologically relevant regions. Our comprehensive approach, integrating advanced lipidomic techniques and bioinformatic analyses, documented the disruptive effects of TBT, an environmental chemical, on the ovarian lipid landscape. Similar to mice, our research also unveiled distinct spatial lipidomic signatures corresponding to specific ovarian compartments in a healthy human ovary that may also be vulnerable to disruption by chemical exposures. Findings from this study not only underscore the vulnerability of the ovarian lipidome to environmental factors but also lay the groundwork for unraveling the molecular pathways underlying ovarian toxicity mediated through lipid dysregulation.
Collapse
Affiliation(s)
- Natalia Pascuali
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Fernando Tobias
- Integrated Molecular Structure Education and Research Center, Department of Chemistry, Northwestern University, Evanston, IL, USA
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Sana Salih
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Almudena Veiga-Lopez
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA; Chicago Center for Health and Environment, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
8
|
Luo W, Xu Z, Li F, Ding L, Wang R, Lin Y, Mao X, Chen X, Li Y, Lu Z, Xie H, Wang H, Zhu Z, Lu Y, Guo L, Yu X, Xia L, He HH, Li G. m6Am Methyltransferase PCIF1 Promotes LPP3 Mediated Phosphatidic Acid Metabolism and Renal Cell Carcinoma Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404033. [PMID: 39422663 PMCID: PMC11633504 DOI: 10.1002/advs.202404033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/16/2024] [Revised: 09/29/2024] [Indexed: 10/19/2024]
Abstract
N6-methyl-2'-O-methyladenosine (m6Am), occurring adjacent to the 7-methylguanosine (m7G) cap structure and catalyzed by the newly identified writer PCIF1 (phosphorylated CTD interacting factor 1), has been implicated in the pathogenesis of various diseases. However, its involvement in renal cell carcinoma (RCC) remains unexplored. Here, significant upregulation of PCIF1 and m6Am levels in RCC tissues are identified, unveiling their oncogenic roles both in vitro and in vivo. Mechanically, employing m6Am-Exo-Seq, LPP3 (phospholipid phosphatase 3) mRNA is identified as a key downstream target whose translation is enhanced by m6Am modification. Furthermore, LPP3 is revealed as a key regulator of phosphatidic acid metabolism, critical for preventing its accumulation in mitochondria and facilitating mitochondrial fission. Consequently, Inhibition of the PCIF1/LPP3 axis significantly altered mitochondrial morphology and reduced RCC tumor progression. In addition, depletion of PCIF1 sensitizes RCC to sunitinib treatment. This study highlights the intricate interplay between m6Am modification, phosphatidic acid metabolism, and mitochondrial dynamics, offering a promising therapeutic avenue for RCC.
Collapse
Affiliation(s)
- Wenqin Luo
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Zhehao Xu
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Fan Li
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Lifeng Ding
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Ruyue Wang
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yudong Lin
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xudong Mao
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Xianjiong Chen
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yang Li
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Zeyi Lu
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Haiyun Xie
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Huan Wang
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Ziwei Zhu
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Yi Lu
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Luying Guo
- Kidney Disease Center of First Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
| | - Xiaojing Yu
- Department of RadiologySir Run Run Shaw hospitalZhejiang University School of MedicineHangzhou310016China
| | - Liqun Xia
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Housheng Hansen He
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Gonghui Li
- Department of UrologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| |
Collapse
|
9
|
Uddin J, Sharma A, Wu D, Tomar S, Ganesan V, Reichel PE, Thota LNR, Cabrera-Silva RI, Marella S, Idelman G, Tay HL, Raya-Sandino A, Reynolds MB, Elesela S, Haberman Y, Denson LA, Parkos CA, O’Riordan MX, Lukacs NW, O’Dwyer DN, Divanovic S, Nusrat A, Weaver TE, Hogan SP. STARD7 maintains intestinal epithelial mitochondria architecture, barrier integrity, and protection from colitis. JCI Insight 2024; 9:e172978. [PMID: 39576011 PMCID: PMC11601949 DOI: 10.1172/jci.insight.172978] [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] [Academic Contribution Register] [Received: 06/12/2023] [Accepted: 09/25/2024] [Indexed: 11/29/2024] Open
Abstract
Susceptibility to inflammatory bowel diseases (IBDs), Crohn's disease (CD), and ulcerative colitis (UC) is linked with loss of intestinal epithelial barrier integrity and mitochondria dysfunction. Steroidogenic acute regulatory (StAR) protein-related lipid transfer (START) domain-containing protein 7 (STARD7) is a phosphatidylcholine-specific (PC-specific) lipid transfer protein that transports PC from the ER to the mitochondria, facilitating mitochondria membrane stabilization and respiration function. The aim of this study was to define the contribution of STARD7 in the regulation of the intestinal epithelial mitochondrial function and susceptibility to colitis. In silico analyses identified significantly reduced expression of STARD7 in patients with UC, which was associated with downregulation of metabolic function and a more severe disease phenotype. STARD7 was expressed in intestinal epithelial cells, and STARD7 knockdown resulted in deformed mitochondria and diminished aerobic respiration. Loss of mitochondria function was associated with reduced expression of tight junction proteins and loss of intestinal epithelial barrier integrity that could be recovered by AMPK activation. Stard7+/- mice were more susceptible to the development of DSS-induced and Il10-/- spontaneous models of colitis. STARD7 is critical for intestinal epithelial mitochondrial function and barrier integrity, and loss of STARD7 function increases susceptibility to IBD.
Collapse
Affiliation(s)
- Jazib Uddin
- Division of Experimental Pathology, Department of Pathology, and
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ankit Sharma
- Division of Experimental Pathology, Department of Pathology, and
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sunil Tomar
- Division of Experimental Pathology, Department of Pathology, and
| | - Varsha Ganesan
- Division of Experimental Pathology, Department of Pathology, and
| | - Paula E. Reichel
- Division of Experimental Pathology, Department of Pathology, and
| | | | | | - Sahiti Marella
- Division of Experimental Pathology, Department of Pathology, and
| | - Gila Idelman
- Division of Experimental Pathology, Department of Pathology, and
| | - Hock L. Tay
- Division of Experimental Pathology, Department of Pathology, and
| | | | - Mack B. Reynolds
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Srikanth Elesela
- Division of Experimental Pathology, Department of Pathology, and
| | - Yael Haberman
- Sheba Medical Center, Tel-Hashomer, and
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lee A. Denson
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Mary X.D. O’Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicholas W. Lukacs
- Division of Experimental Pathology, Department of Pathology, and
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David N. O’Dwyer
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Senad Divanovic
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance and
| | - Asma Nusrat
- Division of Experimental Pathology, Department of Pathology, and
| | - Timothy E. Weaver
- Divisions of Neonatology, Perinatal Biology, and Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Simon P. Hogan
- Division of Experimental Pathology, Department of Pathology, and
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Couto-Lima CA, Machado MCR, Anhezini L, Oliveira MT, Molina RADS, da Silva RR, Lopes GS, Trinca V, Colón DF, Peixoto PM, Monesi N, Alberici LC, Ramos RGP, Espreafico EM. EMC1 Is Required for the Sarcoplasmic Reticulum and Mitochondrial Functions in the Drosophila Muscle. Biomolecules 2024; 14:1258. [PMID: 39456191 PMCID: PMC11506464 DOI: 10.3390/biom14101258] [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] [Academic Contribution Register] [Received: 08/24/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
EMC1 is part of the endoplasmic reticulum (ER) membrane protein complex, whose functions include the insertion of transmembrane proteins into the ER membrane, ER-mitochondria contact, and lipid exchange. Here, we show that the Drosophila melanogaster EMC1 gene is expressed in the somatic musculature and the protein localizes to the sarcoplasmic reticulum (SR) network. Muscle-specific EMC1 RNAi led to severe motility defects and partial late pupae/early adulthood lethality, phenotypes that are rescued by co-expression with an EMC1 transgene. Motility impairment in EMC1-depleted flies was associated with aberrations in muscle morphology in embryos, larvae, and adults, including tortuous and misaligned fibers with reduced size and weakness. They were also associated with an altered SR network, cytosolic calcium overload, and mitochondrial dysfunction and dysmorphology that impaired membrane potential and oxidative phosphorylation capacity. Genes coding for ER stress sensors, mitochondrial biogenesis/dynamics, and other EMC components showed altered expression and were mostly rescued by the EMC1 transgene expression. In conclusion, EMC1 is required for the SR network's mitochondrial integrity and influences underlying programs involved in the regulation of muscle mass and shape. We believe our data can contribute to the biology of human diseases caused by EMC1 mutations.
Collapse
Affiliation(s)
- Carlos Antonio Couto-Lima
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Department of Biotechnology, College of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal 14884-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Maiaro Cabral Rosa Machado
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Lucas Anhezini
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió 57072-900, AL, Brazil
| | - Marcos Túlio Oliveira
- Department of Biotechnology, College of Agricultural and Veterinary Sciences, Sao Paulo State University, Jaboticabal 14884-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Roberto Augusto da Silva Molina
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Rodrigo Ribeiro da Silva
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Gabriel Sarti Lopes
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Vitor Trinca
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - David Fernando Colón
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Pablo M. Peixoto
- Baruch College and Graduate Center, The City University of New York, New York, NY 10010, USA
| | - Nadia Monesi
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Ricardo Guelerman P. Ramos
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| | - Enilza Maria Espreafico
- Department of Cell and Molecular Biology, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
- Cellular and Molecular Biology Program, Faculty of Medicine of Ribeirão Preto, University of São Paulo (FMRP-USP), Ribeirão Preto 14049-900, SP, Brazil
| |
Collapse
|
11
|
Venkatraman K, Lee CT, Budin I. Setting the curve: the biophysical properties of lipids in mitochondrial form and function. J Lipid Res 2024; 65:100643. [PMID: 39303982 PMCID: PMC11513603 DOI: 10.1016/j.jlr.2024.100643] [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] [Academic Contribution Register] [Received: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024] Open
Abstract
Mitochondrial membranes are defined by their diverse functions, complex geometries, and unique lipidomes. In the inner mitochondrial membrane, highly curved membrane folds known as cristae house the electron transport chain and are the primary sites of cellular energy production. The outer mitochondrial membrane is flat by contrast, but is critical for the initiation and mediation of processes key to mitochondrial physiology: mitophagy, interorganelle contacts, fission and fusion dynamics, and metabolite transport. While the lipid composition of both the inner mitochondrial membrane and outer mitochondrial membrane have been characterized across a variety of cell types, a mechanistic understanding for how individual lipid classes contribute to mitochondrial structure and function remains nebulous. In this review, we address the biophysical properties of mitochondrial lipids and their related functional roles. We highlight the intrinsic curvature of the bulk mitochondrial phospholipid pool, with an emphasis on the nuances surrounding the mitochondrially-synthesized cardiolipin. We also outline emerging questions about other lipid classes - ether lipids, and sterols - with potential roles in mitochondrial physiology. We propose that further investigation is warranted to elucidate the specific properties of these lipids and their influence on mitochondrial architecture and function.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Christopher T Lee
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Lee RG, Rudler DL, Rackham O, Filipovska A. Interorganelle phospholipid communication, a house not so divided. Trends Endocrinol Metab 2024; 35:872-883. [PMID: 38972781 DOI: 10.1016/j.tem.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/13/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024]
Abstract
The presence of membrane-bound organelles with specific functions is one of the main hallmarks of eukaryotic cells. Organelle membranes are composed of specific lipids that govern their function and interorganelle communication. Discoveries in cell biology using imaging and omic technologies have revealed the mechanisms that drive membrane remodeling, organelle contact sites, and metabolite exchange. The interplay between multiple organelles and their interdependence is emerging as the next frontier for discovery using 3D reconstruction of volume electron microscopy (vEM) datasets. We discuss recent findings on the links between organelles that underlie common functions and cellular pathways. Specifically, we focus on the metabolism of ether glycerophospholipids that mediate organelle dynamics and their communication with each other, and the new imaging techniques that are powering these discoveries.
Collapse
Affiliation(s)
- Richard G Lee
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia
| | - Danielle L Rudler
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia
| | - Oliver Rackham
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia; Curtin Medical School, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Aleksandra Filipovska
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia; The University of Western Australia Centre for Child Health Research, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia.
| |
Collapse
|
13
|
Li S, Xu R, Yao Y, Rousseau D. ATAD3 is a limiting factor in mitochondrial biogenesis and adipogenesis of white adipocyte-like 3T3-L1 cells. Cell Biol Int 2024; 48:1473-1489. [PMID: 38923254 DOI: 10.1002/cbin.12206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
ATAD3 is a vital ATPase of the inner mitochondrial membrane of pluri-cellular eukaryotes, with largely unknown functions but early required for organism development as necessary for mitochondrial biogenesis. ATAD3 knock-down in C. elegans inhibits at first the development of adipocyte-like intestinal tissue so we used mouse adipocyte model 3T3-L1 cells to analyze ATAD3 functions during adipogenesis and lipogenesis in a mammalian model. ATAD3 function was studied by stable and transient modulation of ATAD3 expression in adipogenesis- induced 3T3-L1 cells using Knock-Down and overexpression strategies, exploring different steps of adipocyte differentiation and lipogenesis. We show that (i) an increase in ATAD3 is preceding differentiation-induced mitochondrial biogenesis; (ii) downregulation of ATAD3 inhibits adipogenesis, lipogenesis, and impedes overexpression of many mitochondrial proteins; (iii) ATAD3 re-expression rescues the phenotype of ATAD3 KD, and (iv) differentiation and lipogenesis are accelerated by ATAD3 overexpression, but inhibited by expression of a dominant-negative mutant. We further show that the ATAD3 KD phenotype is not due to altered insulin signal but involves a limitation of mitochondrial biogenesis linked to Drp1. These results demonstrate that ATAD3 is limiting for in vitro mitochondrial biogenesis and adipogenesis/lipogenesis and therefore that ATAD3 mutation/over- or under-expression could be involved in adipogenic and lipogenic pathologies.
Collapse
Affiliation(s)
- Shuijie Li
- Department of Biology, University Grenoble Alpes, Grenoble, France
| | - Rui Xu
- Institute of Biochemistry and Cell Biology of Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai, China
| | - Yao Yao
- Institute of Biochemistry and Cell Biology of Shanghai Institutes for Biological Sciences Chinese Academy of Sciences, Shanghai, China
| | - Denis Rousseau
- Department of Biology, University Grenoble Alpes, Grenoble, France
- Laboratoire des Matériaux et du Génie Physique-Interfaces entre Matériaux et Matière Biologique -Institut National Polytechnique-Centre National de la Recherche Scientifique - Unité Mixte de Recherche, Grenoble, France
| |
Collapse
|
14
|
Klemm RW, Carvalho P. Lipid Droplets Big and Small: Basic Mechanisms That Make Them All. Annu Rev Cell Dev Biol 2024; 40:143-168. [PMID: 39356808 DOI: 10.1146/annurev-cellbio-012624-031419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/04/2024]
Abstract
Lipid droplets (LDs) are dynamic storage organelles with central roles in lipid and energy metabolism. They consist of a core of neutral lipids, such as triacylglycerol, which is surrounded by a monolayer of phospholipids and specialized surface proteins. The surface composition determines many of the LD properties, such as size, subcellular distribution, and interaction with partner organelles. Considering the diverse energetic and metabolic demands of various cell types, it is not surprising that LDs are highly heterogeneous within and between cell types. Despite their diversity, all LDs share a common biogenesis mechanism. However, adipocytes have evolved specific adaptations of these basic mechanisms, enabling the regulation of lipid and energy metabolism at both the cellular and organismal levels. Here, we discuss recent advances in the understanding of both the general mechanisms of LD biogenesis and the adipocyte-specific adaptations controlling these fascinating organelles.
Collapse
Affiliation(s)
- Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom;
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
15
|
Rockenfeller P. Phospholipid Scramblase Activity of VDAC Dimers: New Implications for Cell Death, Autophagy and Ageing. Biomolecules 2024; 14:1218. [PMID: 39456151 PMCID: PMC11506367 DOI: 10.3390/biom14101218] [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] [Academic Contribution Register] [Received: 08/27/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Voltage-dependent anion channels (VDACs) are important proteins of the outer mitochondrial membrane (OMM). Their beta-barrel structure allows for efficient metabolite exchange between the cytosol and mitochondria. VDACs have further been implicated in the control of regulated cell death. Historically, VDACs have been pictured as part of the mitochondrial permeability transition pore (MPTP). New concepts of regulated cell death involving VDACs include its oligomerisation to form a large pore complex in the OMM; however, alternative VDAC localisation to the plasma membrane has been suggested in the literature and will be discussed regarding its potential role during cell death. Very recently, a phospholipid scramblase activity has been attributed to VDAC dimers, which explains the manifold lipidomic changes observed in VDAC-deficient yeast strains. In this review, I highlight the recent advances regarding VDAC's phospholipid scramblase function and discuss how this new insight sheds new light on VDAC's implication in regulated cell death, autophagy, and ageing.
Collapse
Affiliation(s)
- Patrick Rockenfeller
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| |
Collapse
|
16
|
Neto MV, De Rossi G, Berkowitz BA, Seabra MC, Luthert PJ, Futter CE, Burgoyne T. Daily Light Onset and Plasma Membrane Tethers Regulate Mitochondria Redistribution within the Retinal Pigment Epithelium. Cells 2024; 13:1100. [PMID: 38994953 PMCID: PMC11240580 DOI: 10.3390/cells13131100] [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] [Academic Contribution Register] [Received: 05/03/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
The retinal pigment epithelium (RPE) is an essential component of the retina that plays multiple roles required to support visual function. These include light onset- and circadian rhythm-dependent tasks, such as daily phagocytosis of photoreceptor outer segments. Mitochondria provide energy to the highly specialized and energy-dependent RPE. In this study, we examined the positioning of mitochondria and how this is influenced by the onset of light. We identified a population of mitochondria that are tethered to the basal plasma membrane pre- and post-light onset. Following light onset, mitochondria redistributed apically and interacted with melanosomes and phagosomes. In a choroideremia mouse model that has regions of the RPE with disrupted or lost infolding of the plasma membrane, the positionings of only the non-tethered mitochondria were affected. This provides evidence that the tethering of mitochondria to the plasma membrane plays an important role that is maintained under these disease conditions. Our work shows that there are subpopulations of RPE mitochondria based on their positioning after light onset. It is likely they play distinct roles in the RPE that are needed to fulfil the changing cellular demands throughout the day.
Collapse
Affiliation(s)
- Matilde V. Neto
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Giulia De Rossi
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Bruce A. Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Miguel C. Seabra
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Philip J. Luthert
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Clare E. Futter
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
17
|
Nakamura K, Aoyama-Ishiwatari S, Nagao T, Paaran M, Obara CJ, Sakurai-Saito Y, Johnston J, Du Y, Suga S, Tsuboi M, Nakakido M, Tsumoto K, Kishi Y, Gotoh Y, Kwak C, Rhee HW, Seo JK, Kosako H, Potter C, Carragher B, Lippincott-Schwartz J, Polleux F, Hirabayashi Y. PDZD8-FKBP8 tethering complex at ER-mitochondria contact sites regulates mitochondrial complexity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.22.554218. [PMID: 38895210 PMCID: PMC11185567 DOI: 10.1101/2023.08.22.554218] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/21/2024]
Abstract
Mitochondria-ER membrane contact sites (MERCS) represent a fundamental ultrastructural feature underlying unique biochemistry and physiology in eukaryotic cells. The ER protein PDZD8 is required for the formation of MERCS in many cell types, however, its tethering partner on the outer mitochondrial membrane (OMM) is currently unknown. Here we identified the OMM protein FKBP8 as the tethering partner of PDZD8 using a combination of unbiased proximity proteomics, CRISPR-Cas9 endogenous protein tagging, Cryo-Electron Microscopy (Cryo-EM) tomography, and correlative light-EM (CLEM). Single molecule tracking revealed highly dynamic diffusion properties of PDZD8 along the ER membrane with significant pauses and capture at MERCS. Overexpression of FKBP8 was sufficient to narrow the ER-OMM distance, whereas independent versus combined deletions of these two proteins demonstrated their interdependence for MERCS formation. Furthermore, PDZD8 enhances mitochondrial complexity in a FKBP8-dependent manner. Our results identify a novel ER-mitochondria tethering complex that regulates mitochondrial morphology in mammalian cells.
Collapse
|
18
|
Teixeira P, Galland R, Chevrollier A. Super-resolution microscopies, technological breakthrough to decipher mitochondrial structure and dynamic. Semin Cell Dev Biol 2024; 159-160:38-51. [PMID: 38310707 DOI: 10.1016/j.semcdb.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/11/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
Mitochondria are complex organelles with an outer membrane enveloping a second inner membrane that creates a vast matrix space partitioned by pockets or cristae that join the peripheral inner membrane with several thin junctions. Several micrometres long, mitochondria are generally close to 300 nm in diameter, with membrane layers separated by a few tens of nanometres. Ultrastructural data from electron microscopy revealed the structure of these mitochondria, while conventional optical microscopy revealed their extraordinary dynamics through fusion, fission, and migration processes but its limited resolution power restricted the possibility to go further. By overcoming the limits of light diffraction, Super-Resolution Microscopy (SRM) now offers the potential to establish the links between the ultrastructure and remodelling of mitochondrial membranes, leading to major advances in our understanding of mitochondria's structure-function. Here we review the contributions of SRM imaging to our understanding of the relationship between mitochondrial structure and function. What are the hopes for these new imaging approaches which are particularly important for mitochondrial pathologies?
Collapse
Affiliation(s)
- Pauline Teixeira
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MITOLAB, SFR ICAT, F-49000 Angers, France
| | - Rémi Galland
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Arnaud Chevrollier
- Univ. Angers, INSERM, CNRS, MITOVASC, Equipe MITOLAB, SFR ICAT, F-49000 Angers, France.
| |
Collapse
|
19
|
Tong L, Chen Z, Li Y, Wang X, Yang C, Li Y, Zhu Y, Lu Y, Liu Q, Xu N, Shao S, Wu L, Zhang P, Wu G, Wu X, Chen X, Fang J, Jia R, Xu T, Li B, Zheng L, Liu J, Tong X. Transketolase promotes MAFLD by limiting inosine-induced mitochondrial activity. Cell Metab 2024; 36:1013-1029.e5. [PMID: 38547864 DOI: 10.1016/j.cmet.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/07/2023] [Revised: 01/10/2024] [Accepted: 03/06/2024] [Indexed: 05/12/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has a global prevalence of about 25% and no approved therapy. Using metabolomic and proteomic analyses, we identified high expression of hepatic transketolase (TKT), a metabolic enzyme of the pentose phosphate pathway, in human and mouse MAFLD. Hyperinsulinemia promoted TKT expression through the insulin receptor-CCAAT/enhancer-binding protein alpha axis. Utilizing liver-specific TKT overexpression and knockout mouse models, we demonstrated that TKT was sufficient and required for MAFLD progression. Further metabolic flux analysis revealed that Tkt deletion increased hepatic inosine levels to activate the protein kinase A-cAMP response element binding protein cascade, promote phosphatidylcholine synthesis, and improve mitochondrial function. Moreover, insulin induced hepatic TKT to limit inosine-dependent mitochondrial activity. Importantly, N-acetylgalactosamine (GalNAc)-siRNA conjugates targeting hepatic TKT showed promising therapeutic effects on mouse MAFLD. Our study uncovers how hyperinsulinemia regulates TKT-orchestrated inosine metabolism and mitochondrial function and provides a novel therapeutic strategy for MAFLD prevention and treatment.
Collapse
Affiliation(s)
- Lingfeng Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhangbing Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yangyang Li
- Unit of Immune and Metabolic Regulation, School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Xinxia Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Changjie Yang
- Department of Liver Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yakui Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Qi Liu
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Nannan Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sijia Shao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guangyu Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoyu Wu
- Key Laboratory of Pediatric Hematology and Oncology, Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiaosong Chen
- Department of Liver Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Junwei Fang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai 200032, China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Tianle Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liang Zheng
- Key Laboratory of Pediatric Hematology and Oncology, Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Synvida Biotechnology Co., Ltd, Shanghai, China.
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
20
|
Xiao T, English AM, Wilson ZN, Maschek J, Cox JE, Hughes AL. The phospholipids cardiolipin and phosphatidylethanolamine differentially regulate MDC biogenesis. J Cell Biol 2024; 223:e202302069. [PMID: 38497895 PMCID: PMC10949074 DOI: 10.1083/jcb.202302069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
Cells utilize multiple mechanisms to maintain mitochondrial homeostasis. We recently characterized a pathway that remodels mitochondria in response to metabolic alterations and protein overload stress. This remodeling occurs via the formation of large membranous structures from the mitochondrial outer membrane called mitochondrial-derived compartments (MDCs), which are eventually released from mitochondria and degraded. Here, we conducted a microscopy-based screen in budding yeast to identify factors that regulate MDC formation. We found that two phospholipids, cardiolipin (CL) and phosphatidylethanolamine (PE), differentially regulate MDC biogenesis. CL depletion impairs MDC biogenesis, whereas blocking mitochondrial PE production leads to constitutive MDC formation. Additionally, in response to metabolic MDC activators, cellular and mitochondrial PE declines, and overexpressing mitochondrial PE synthesis enzymes suppress MDC biogenesis. Altogether, our data indicate a requirement for CL in MDC biogenesis and suggest that PE depletion may stimulate MDC formation downstream of MDC-inducing metabolic stress.
Collapse
Affiliation(s)
- Tianyao Xiao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Alyssa M. English
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Zachary N. Wilson
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - J.Alan. Maschek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integration. Physiology, University of Utah College of Health, Salt Lake City, UT, USA
| | - James E. Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
| | - Adam L. Hughes
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
21
|
Blumstein DM, MacManes MD. Impacts of dietary fat on multi tissue gene expression in the desert-adapted cactus mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592397. [PMID: 38746252 PMCID: PMC11092757 DOI: 10.1101/2024.05.03.592397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/16/2024]
Abstract
Understanding the relationship between dietary fat and physiological responses is crucial in species adapted to arid environments where water scarcity is common. In this study, we present a comprehensive exploration of gene expression across five tissues (kidney, liver, lung, gastrointestinal tract, and hypothalamus) and 19 phenotypic measurements, investigating the effects of dietary fat in the desert-adapted cactus mouse ( Peromyscus eremicus ). We show impacts on immune function, circadian gene regulation, and mitochondrial function for mice fed a lower-fat diet compared to mice fed a higher-fat diet. In arid environments with severe water scarcity, even subtle changes in organismal health and water balance can affect physical performance, potentially impacting survival and reproductive success. The study sheds light on the complex interplay between diet, physiological processes, and environmental adaptation, providing valuable insights into the multifaceted impacts of dietary choices on organismal well-being and adaptation strategies in arid habitats.
Collapse
|
22
|
Saukko-Paavola AJ, Klemm RW. Remodelling of mitochondrial function by import of specific lipids at multiple membrane-contact sites. FEBS Lett 2024; 598:1274-1291. [PMID: 38311340 DOI: 10.1002/1873-3468.14813] [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] [Academic Contribution Register] [Received: 11/29/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Organelles form physical and functional contact between each other to exchange information, metabolic intermediates, and signaling molecules. Tethering factors and contact site complexes bring partnering organelles into close spatial proximity to establish membrane contact sites (MCSs), which specialize in unique functions like lipid transport or Ca2+ signaling. Here, we discuss how MCSs form dynamic platforms that are important for lipid metabolism. We provide a perspective on how import of specific lipids from the ER and other organelles may contribute to remodeling of mitochondria during nutrient starvation. We speculate that mitochondrial adaptation is achieved by connecting several compartments into a highly dynamic organelle network. The lipid droplet appears to be a central hub in coordinating the function of these organelle neighborhoods.
Collapse
Affiliation(s)
| | - Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
23
|
Mavuduru VA, Vadupu L, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial phospholipid transport: Role of contact sites and lipid transport proteins. Prog Lipid Res 2024; 94:101268. [PMID: 38195013 DOI: 10.1016/j.plipres.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/11/2024]
Abstract
One of the major constituents of mitochondrial membranes is the phospholipids, which play a key role in maintaining the structure and the functions of the mitochondria. However, mitochondria do not synthesize most of the phospholipids in situ, necessitating the presence of phospholipid import pathways. Even for the phospholipids, which are synthesized within the inner mitochondrial membrane (IMM), the phospholipid precursors must be imported from outside the mitochondria. Therefore, the mitochondria heavily rely on the phospholipid transport pathways for its proper functioning. Since, mitochondria are not part of a vesicular trafficking network, the molecular mechanisms of how mitochondria receive its phospholipids remain a relevant question. One of the major ways that hydrophobic phospholipids can cross the aqueous barrier of inter or intraorganellar spaces is by apposing membranes, thereby decreasing the distance of transport, or by being sequestered by lipid transport proteins (LTPs). Therefore, with the discovery of LTPs and membrane contact sites (MCSs), we are beginning to understand the molecular mechanisms of phospholipid transport pathways in the mitochondria. In this review, we will present a brief overview of the recent findings on the molecular architecture and the importance of the MCSs, both the intraorganellar and interorganellar contact sites, in facilitating the mitochondrial phospholipid transport. In addition, we will also discuss the role of LTPs for trafficking phospholipids through the intermembrane space (IMS) of the mitochondria. Mechanistic insights into different phospholipid transport pathways of mitochondria could be exploited to vary the composition of membrane phospholipids and gain a better understanding of their precise role in membrane homeostasis and mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Vijay Aditya Mavuduru
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Lavanya Vadupu
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522502, India
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore; Department of Clinical Neuroscience, Karolinska Institute, Stockholm 17176, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore; Cognitive Neuroimaging Centre, Nanyang Technological University, Singapore, 59 Nanyang Drive, 636921, Singapore.
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur, Andhra Pradesh 522240, India.
| |
Collapse
|
24
|
Li M, Zhang Y, Yu G, Gu L, Zhu H, Feng S, Xiong X, Jian Z. Mitochondria-associated endoplasmic reticulum membranes tethering protein VAPB-PTPIP51 protects against ischemic stroke through inhibiting the activation of autophagy. CNS Neurosci Ther 2024; 30:e14707. [PMID: 38584329 PMCID: PMC10999572 DOI: 10.1111/cns.14707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2024] [Revised: 03/09/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024] Open
Abstract
AIMS Mitochondria-associated endoplasmic reticulum membranes (MAMs) serve as a crucial bridge connecting the endoplasmic reticulum (ER) and mitochondria within cells. Vesicle-associated membrane protein-associated protein B (VAPB) and protein tyrosine phosphatase interacting protein 51 (PTPIP51) are responsible for the formation and stability of MAMs, which have been implicated in the pathogenesis of various diseases. However, the role of MAMs in ischemic stroke (IS) remains unclear. We aimed to investigate the role of MAMs tethering protein VAPB-PTPIP51 in experimental cerebral ischemia. METHODS We simulated cerebral ischemia-reperfusion injury (CIRI) by using a mouse middle cerebral artery occlusion (MCAO) model. RESULTS We observed a decrease in VAPB-PTPIP51 expression in the brain tissue. Our findings suggested compromised MAMs after MCAO, as a decreased mitochondria-ER contact (MERC) coverage and an increased distance were observed through the transmission electron microscope (TEM). Upon VAPB or PTPIP51 knockdown, the damage to MAMs was exacerbated, accompanied by excessive autophagy activation and increased reactive oxygen species (ROS) production, resulting in an enlarged infarct area and exacerbated neurological deficits. Notably, we observed that this damage was concomitant with the inhibition of the PI3K/AKT/mTOR pathway and was successfully mitigated by the treatment with the PI3K activator. CONCLUSIONS Our findings suggest that the downregulation of VAPB-PTPIP51 expression after IS mediates structural damage to MAMs. This may exacerbate CIRI by inhibiting the PI3K pathway and activating autophagy, thus providing new therapeutic targets for IS.
Collapse
Affiliation(s)
- Mingyang Li
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yonggang Zhang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Guixiang Yu
- Department of OphthalmologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Lijuan Gu
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hua Zhu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Shi Feng
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhihong Jian
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
25
|
Erazo-Oliveras A, Muñoz-Vega M, Salinas ML, Wang X, Chapkin RS. Dysregulation of cellular membrane homeostasis as a crucial modulator of cancer risk. FEBS J 2024; 291:1299-1352. [PMID: 36282100 PMCID: PMC10126207 DOI: 10.1111/febs.16665] [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] [Academic Contribution Register] [Received: 06/18/2022] [Revised: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Cellular membranes serve as an epicentre combining extracellular and cytosolic components with membranous effectors, which together support numerous fundamental cellular signalling pathways that mediate biological responses. To execute their functions, membrane proteins, lipids and carbohydrates arrange, in a highly coordinated manner, into well-defined assemblies displaying diverse biological and biophysical characteristics that modulate several signalling events. The loss of membrane homeostasis can trigger oncogenic signalling. More recently, it has been documented that select membrane active dietaries (MADs) can reshape biological membranes and subsequently decrease cancer risk. In this review, we emphasize the significance of membrane domain structure, organization and their signalling functionalities as well as how loss of membrane homeostasis can steer aberrant signalling. Moreover, we describe in detail the complexities associated with the examination of these membrane domains and their association with cancer. Finally, we summarize the current literature on MADs and their effects on cellular membranes, including various mechanisms of dietary chemoprevention/interception and the functional links between nutritional bioactives, membrane homeostasis and cancer biology.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Mónica Muñoz-Vega
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Xiaoli Wang
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases; Texas A&M University; College Station, Texas, 77843; USA
- Department of Nutrition; Texas A&M University; College Station, Texas, 77843; USA
- Center for Environmental Health Research; Texas A&M University; College Station, Texas, 77843; USA
| |
Collapse
|
26
|
Chen Z, Pan Z, Huang C, Zhu X, Li N, Huynh H, Xu J, Huang L, Vaz FM, Liu J, Han Z, Ouyang K. Cardiac lipidomic profiles in mice undergo changes from fetus to adult. Life Sci 2024; 341:122484. [PMID: 38311219 DOI: 10.1016/j.lfs.2024.122484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
AIMS Lipids are essential cellular components with many important biological functions. Disturbed lipid biosynthesis and metabolism has been shown to cause cardiac developmental abnormality and cardiovascular diseases. In this study, we aimed to investigate the composition and the molecular profiles of lipids in mammalian hearts between embryonic and adult stages and uncover the underlying links between lipid and cardiac development and maturation. MATERIALS AND METHODS We collected mouse hearts at the embryonic day 11.5 (E11.5), E15.5, and the age of 2 months, 4 months and 10 months, and performed lipidomic analysis to determine the changes of the composition, molecular species, and relative abundance of cardiac lipids between embryonic and adult stages. Additionally, we also performed the electronic microscopy and RNA sequencing in both embryonic and adult mouse hearts. KEY FINDINGS The relative abundances of certain phospholipids and sphingolipids including cardiolipin, phosphatidylglycerol, phosphatidylethanolamine, and ceramide, are different between embryonic and adult hearts. Such lipidomic changes are accompanied with increased densities of mitochondrial membranes and elevated expression of genes related to mitochondrial formation in adult mouse hearts. We also analyzed individual molecular species of phospholipids and sphingolipids, and revealed that the composition and distribution of lipid molecular species in hearts also change with development. SIGNIFICANCE Our study provides not only a lipidomic view of mammalian hearts when developing from the embryonic to the adult stage, but also a potential pool of lipid indicators for cardiac cell development and maturation.
Collapse
Affiliation(s)
- Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Zhixiang Pan
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Helen Huynh
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, the Netherlands
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| |
Collapse
|
27
|
Ahola S, Langer T. Ferroptosis in mitochondrial cardiomyopathy. Trends Cell Biol 2024; 34:150-160. [PMID: 37419738 DOI: 10.1016/j.tcb.2023.06.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 07/09/2023]
Abstract
Ferroptosis is a form of necrotic cell death characterized by iron-dependent lipid peroxidation culminating in membrane rupture. Accumulating evidence links ferroptosis to multiple cardiac diseases and identifies mitochondria as important regulators of ferroptosis. Mitochondria are not only a major source of reactive oxygen species (ROS) but also counteract ferroptosis by preserving cellular redox balance and oxidative defense. Recent evidence has revealed that the mitochondrial integrated stress response limits oxidative stress and ferroptosis in oxidative phosphorylation (OXPHOS)-deficient cardiomyocytes and protects against mitochondrial cardiomyopathy. We summarize the multiple ways in which mitochondria modulate the susceptibility of cells to ferroptosis, and discuss the implications of ferroptosis for cardiomyopathies in mitochondrial disease.
Collapse
Affiliation(s)
- Sofia Ahola
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
28
|
Han X, Huang S, Zhuang Z, Zhang X, Xie M, Lou N, Hua M, Zhuang X, Yu S, Chen S. Phosphatidate phosphatase Lipin1 involves in diabetic encephalopathy pathogenesis via regulating synaptic mitochondrial dynamics. Redox Biol 2024; 69:102996. [PMID: 38103341 PMCID: PMC10770635 DOI: 10.1016/j.redox.2023.102996] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Diabetic encephalopathy (DE) is a common central nervous system complication of diabetes mellitus without effective therapy currently. Recent studies have highlighted synaptic mitochondrial damages as a possible pathological basis for DE, but the underlying mechanisms remain unclear. Our previous work has revealed that phosphatidate phosphatase Lipin1, a critical enzyme involved with phospholipid synthesis, is closely related to the pathogenesis of DE. Here, we demonstrate that Lipin1 is significantly down-regulated in rat hippocampus of DE. Knock-down of Lipin1 within hippocampus of normal rats induces dysregulation of homeostasis in synaptic mitochondrial dynamics with an increase of mitochondrial fission and a decrease of fusion, then causes synaptic mitochondrial dysfunction, synaptic plasticity deficits as well as cognitive impairments, similar to that observed in response to chronic hyperglycemia exposure. In contrast, an up-regulation of Lipin1 within hippocampus in the DE model ameliorates this cascade of dysfunction. We also find that the effect of Lipin1 that regulating mitochondrial dynamics results from maintaining appropriate phospholipid components in the mitochondrial membrane. In conclusion, alterations in hippocampal Lipin1 contribute to hippocampal synaptic mitochondrial dysfunction and cognitive deficits observed in DE. Targeting Lipin1 might be a potential therapeutic strategy for the clinical treatment of DE.
Collapse
Affiliation(s)
- Xiaolin Han
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Shan Huang
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Ziyun Zhuang
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China; Department of Endocrinology and Metabolism, First People's Hospital of Jinan, Jinan, 250011, China
| | - Xiaochen Zhang
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China; Department of Clinical Medicine, Heze Medical College, Heze, 274009, China
| | - Min Xie
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Nengjun Lou
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan, 250033, China
| | - Mengyu Hua
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Xianghua Zhuang
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan, 250033, China.
| | - Shuyan Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Shihong Chen
- Depratment of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan, 250033, China.
| |
Collapse
|
29
|
Headley CA, Gautam S, Olmo‐Fontanez A, Garcia‐Vilanova A, Dwivedi V, Akhter A, Schami A, Chiem K, Ault R, Zhang H, Cai H, Whigham A, Delgado J, Hicks A, Tsao PS, Gelfond J, Martinez‐Sobrido L, Wang Y, Torrelles JB, Turner J. Extracellular Delivery of Functional Mitochondria Rescues the Dysfunction of CD4 + T Cells in Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303664. [PMID: 37990641 PMCID: PMC10837346 DOI: 10.1002/advs.202303664] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/05/2023] [Revised: 09/17/2023] [Indexed: 11/23/2023]
Abstract
Mitochondrial dysfunction alters cellular metabolism, increases tissue oxidative stress, and may be principal to the dysregulated signaling and function of CD4+ T lymphocytes in the elderly. In this proof of principle study, it is investigated whether the transfer of functional mitochondria into CD4+ T cells that are isolated from old mice (aged CD4+ T cells), can abrogate aging-associated mitochondrial dysfunction, and improve the aged CD4+ T cell functionality. The results show that the delivery of exogenous mitochondria to aged non-activated CD4+ T cells led to significant mitochondrial proteome alterations highlighted by improved aerobic metabolism and decreased cellular mitoROS. Additionally, mito-transferred aged CD4+ T cells showed improvements in activation-induced TCR-signaling kinetics displaying markers of activation (CD25), increased IL-2 production, enhanced proliferation ex vivo. Importantly, immune deficient mouse models (RAG-KO) showed that adoptive transfer of mito-transferred naive aged CD4+ T cells, protected recipient mice from influenza A and Mycobacterium tuberculosis infections. These findings support mitochondria as targets of therapeutic intervention in aging.
Collapse
Affiliation(s)
- Colwyn A. Headley
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
- Biomedical Sciences Graduate ProgramThe Ohio State UniversityColumbusOhio43201USA
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCA94305USA
| | - Shalini Gautam
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | | | | | - Varun Dwivedi
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Anwari Akhter
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Alyssa Schami
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Kevin Chiem
- Disease Intervention & Prevention ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Russell Ault
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
- Biomedical Sciences Graduate ProgramThe Ohio State UniversityColumbusOhio43201USA
| | - Hao Zhang
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Hong Cai
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Alison Whigham
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Jennifer Delgado
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Amberlee Hicks
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Philip S. Tsao
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCA94305USA
| | - Jonathan Gelfond
- UT‐Health San AntonioDepartment of Epidemiology & BiostatisticsSan AntonioTexas78229USA
| | - Luis Martinez‐Sobrido
- Disease Intervention & Prevention ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Yufeng Wang
- Department of Molecular Microbiology and ImmunologySouth Texas Center for Emerging Infectious DiseasesThe University of Texas at San AntonioSan AntonioTX78249USA
| | - Jordi B. Torrelles
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| | - Joanne Turner
- Host‐Pathogen Interactions ProgramTexas Biomedical Research InstituteSan AntonioTexas78227USA
| |
Collapse
|
30
|
Dasgupta SK, Gollamudi J, Rivera S, Poche RA, Rumbaut RE, Thiagarajan P. β2-glycoprotein I promotes the clearance of circulating mitochondria. PLoS One 2024; 19:e0293304. [PMID: 38271349 PMCID: PMC10810532 DOI: 10.1371/journal.pone.0293304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2023] [Accepted: 10/10/2023] [Indexed: 01/27/2024] Open
Abstract
β2-glycoprotein I (β2-Gp1) is a cardiolipin-binding plasma glycoprotein. It is evolutionarily conserved from invertebrates, and cardiolipin-bound β2-Gp1 is a major target of antiphospholipid antibodies seen in autoimmune disorders. Cardiolipin is almost exclusively present in mitochondria, and mitochondria are present in circulating blood. We show that β2-Gp1 binds to cell-free mitochondria (CFM) in the circulation and promotes its phagocytosis by macrophages at physiological plasma concentrations. Exogenous CFM had a short circulation time of less than 10 minutes in mice. Following infusion of CFM, β2-Gp1-deficient mice had significantly higher levels of transfused mitochondria at 5 minutes (9.9 ± 6.4 pg/ml versus 4.0 ± 2.3 pg/ml in wildtype, p = 0.01) and at 10 minutes (3.0 ± 3.6 pg/ml versus 1.0 ± 0.06 pg/ml in wild-type, p = 0.033, n = 10). In addition, the splenic macrophages had less phagocytosed CFM in β2-Gp1-deficient mice (24.4 ± 2.72% versus 35.6 ± 3.5 in wild-type, p = 0.001, n = 5). A patient with abnormal β2-Gp1, unable to bind cardiolipin, has increased CFM in blood (5.09 pg/ml versus 1.26 ± 1.35 in normal) and his plasma induced less phagocytosis of CFM by macrophages (47.3 ± 1.6% versus 54.3 ± 1.3, p = 0.01) compared to normal plasma. These results show the evolutionarily conserved β2-Gp1 is one of the mediators of the clearance of CFM in circulation.
Collapse
Affiliation(s)
- Swapan Kumar Dasgupta
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center and Departments of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jahnavi Gollamudi
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Stefanie Rivera
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center and Departments of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ross A. Poche
- Department of Medicine Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rolando E. Rumbaut
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Perumal Thiagarajan
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center and Departments of Pathology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
31
|
Sengupta S, Levy DL. Organelle Communication with the Nucleus. Results Probl Cell Differ 2024; 73:3-23. [PMID: 39242372 PMCID: PMC11409190 DOI: 10.1007/978-3-031-62036-2_1] [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] [Academic Contribution Register] [Indexed: 09/09/2024]
Abstract
Compartmentalization of cellular components is critical to the spatiotemporal and environmental regulation of biochemical activities inside a cell, ensures the proper division of cellular labor and resources, and increases the efficiency of metabolic processes. However, compartmentalization also poses a challenge as organelles often need to communicate across these compartments to complete reaction pathways. These communication signals are often critical aspects of the cellular response to changing environmental conditions. A central signaling hub in the cell, the nucleus communicates with mitochondria, lysosomes, the endoplasmic reticulum, and the Golgi body to ensure optimal organellar and cellular performance. Here we review different mechanisms by which these organelles communicate with the nucleus, focusing on anterograde and retrograde signaling of mitochondria, localization-based signaling of lysosomes, the unfolded protein response of the endoplasmic reticulum, and evidence for nucleus-Golgi signaling. We also include a brief overview of some less well-characterized mechanisms of communication between non-nuclear organelles.
Collapse
Affiliation(s)
- Sourabh Sengupta
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Daniel L. Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
32
|
Sun H, Zhang J, Ye Q, Jiang T, Liu X, Zhang X, Zeng F, Li J, Zheng Y, Han X, Su C, Shi Y. LPGAT1 controls MEGDEL syndrome by coupling phosphatidylglycerol remodeling with mitochondrial transport. Cell Rep 2023; 42:113214. [PMID: 37917582 PMCID: PMC10729602 DOI: 10.1016/j.celrep.2023.113214] [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] [Academic Contribution Register] [Received: 05/08/2023] [Revised: 08/21/2023] [Accepted: 09/19/2023] [Indexed: 11/04/2023] Open
Abstract
Phosphatidylglycerol (PG) is a mitochondrial phospholipid required for mitochondrial cristae structure and cardiolipin synthesis. PG must be remodeled to its mature form at the endoplasmic reticulum (ER) after mitochondrial biosynthesis to achieve its biological functions. Defective PG remodeling causes MEGDEL (non-alcohol fatty liver disease and 3-methylglutaconic aciduria with deafness, encephalopathy, and Leigh-like) syndrome through poorly defined mechanisms. Here, we identify LPGAT1, an acyltransferase that catalyzes PG remodeling, as a candidate gene for MEGDEL syndrome. We show that PG remodeling by LPGAT1 at the ER is closely coordinated with mitochondrial transport through interaction with the prohibitin/TIMM14 mitochondrial import motor. Accordingly, ablation of LPGAT1 or TIMM14 not only causes aberrant fatty acyl compositions but also ER retention of newly remodeled PG, leading to profound loss in mitochondrial crista structure and respiration. Consequently, genetic deletion of the LPGAT1 in mice leads to cardinal features of MEGDEL syndrome, including 3-methylglutaconic aciduria, deafness, dilated cardiomyopathy, and premature death, which are highly reminiscent of those caused by TIMM14 mutations in humans.
Collapse
Affiliation(s)
- Haoran Sun
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Jun Zhang
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, 4939 Charles Katz Drive, San Antonio, TX 78229, USA
| | - Qianqian Ye
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China; Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, 4939 Charles Katz Drive, San Antonio, TX 78229, USA
| | - Ting Jiang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Xueling Liu
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Xiaoyang Zhang
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Fanyu Zeng
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China; Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, 4939 Charles Katz Drive, San Antonio, TX 78229, USA
| | - Jie Li
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Yue Zheng
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, 4939 Charles Katz Drive, San Antonio, TX 78229, USA
| | - Chuan Su
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 101 Longmian Avenue, Nanjing, Jiangsu Province 211166, China
| | - Yuguang Shi
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Pharmacology, University of Texas Health Science Center at San Antonio, 4939 Charles Katz Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
33
|
Semenov AY, Tikhonov AN. Electrometric and Electron Paramagnetic Resonance Measurements of a Difference in the Transmembrane Electrochemical Potential: Photosynthetic Subcellular Structures and Isolated Pigment-Protein Complexes. MEMBRANES 2023; 13:866. [PMID: 37999352 PMCID: PMC10673362 DOI: 10.3390/membranes13110866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/08/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
A transmembrane difference in the electrochemical potentials of protons (ΔμH+) serves as a free energy intermediate in energy-transducing organelles of the living cell. The contributions of two components of the ΔμH+ (electrical, Δψ, and concentrational, ΔpH) to the overall ΔμH+ value depend on the nature and lipid composition of the energy-coupling membrane. In this review, we briefly consider several of the most common instrumental (electrometric and EPR) methods for numerical estimations of Δψ and ΔpH. In particular, the kinetics of the flash-induced electrometrical measurements of Δψ in bacterial chromatophores, isolated bacterial reaction centers, and Photosystems I and II of the oxygenic photosynthesis, as well as the use of pH-sensitive molecular indicators and kinetic data regarding pH-dependent electron transport in chloroplasts, have been reviewed. Further perspectives on the application of these methods to solve some fundamental and practical problems of membrane bioenergetics are discussed.
Collapse
Affiliation(s)
- Alexey Yu. Semenov
- A.N. Belozersky Institute of Physical-Chemical Biology, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia;
| | | |
Collapse
|
34
|
Ali O, Szabó A. Review of Eukaryote Cellular Membrane Lipid Composition, with Special Attention to the Fatty Acids. Int J Mol Sci 2023; 24:15693. [PMID: 37958678 PMCID: PMC10649022 DOI: 10.3390/ijms242115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Biological membranes, primarily composed of lipids, envelop each living cell. The intricate composition and organization of membrane lipids, including the variety of fatty acids they encompass, serve a dynamic role in sustaining cellular structural integrity and functionality. Typically, modifications in lipid composition coincide with consequential alterations in universally significant signaling pathways. Exploring the various fatty acids, which serve as the foundational building blocks of membrane lipids, provides crucial insights into the underlying mechanisms governing a myriad of cellular processes, such as membrane fluidity, protein trafficking, signal transduction, intercellular communication, and the etiology of certain metabolic disorders. Furthermore, comprehending how alterations in the lipid composition, especially concerning the fatty acid profile, either contribute to or prevent the onset of pathological conditions stands as a compelling area of research. Hence, this review aims to meticulously introduce the intricacies of membrane lipids and their constituent fatty acids in a healthy organism, thereby illuminating their remarkable diversity and profound influence on cellular function. Furthermore, this review aspires to highlight some potential therapeutic targets for various pathological conditions that may be ameliorated through dietary fatty acid supplements. The initial section of this review expounds on the eukaryotic biomembranes and their complex lipids. Subsequent sections provide insights into the synthesis, membrane incorporation, and distribution of fatty acids across various fractions of membrane lipids. The last section highlights the functional significance of membrane-associated fatty acids and their innate capacity to shape the various cellular physiological responses.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
| | - András Szabó
- Agrobiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary;
- HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, 7400 Kaposvár, Hungary
| |
Collapse
|
35
|
Su H, Guo H, Qiu X, Lin TY, Qin C, Celio G, Yong P, Senders M, Han X, Bernlohr DA, Chen X. Lipocalin 2 regulates mitochondrial phospholipidome remodeling, dynamics, and function in brown adipose tissue in male mice. Nat Commun 2023; 14:6729. [PMID: 37872178 PMCID: PMC10593768 DOI: 10.1038/s41467-023-42473-2] [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] [Academic Contribution Register] [Received: 11/17/2022] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
Mitochondrial function is vital for energy metabolism in thermogenic adipocytes. Impaired mitochondrial bioenergetics in brown adipocytes are linked to disrupted thermogenesis and energy balance in obesity and aging. Phospholipid cardiolipin (CL) and phosphatidic acid (PA) jointly regulate mitochondrial membrane architecture and dynamics, with mitochondria-associated endoplasmic reticulum membranes (MAMs) serving as the platform for phospholipid biosynthesis and metabolism. However, little is known about the regulators of MAM phospholipid metabolism and their connection to mitochondrial function. We discover that LCN2 is a PA binding protein recruited to the MAM during inflammation and metabolic stimulation. Lcn2 deficiency disrupts mitochondrial fusion-fission balance and alters the acyl-chain composition of mitochondrial phospholipids in brown adipose tissue (BAT) of male mice. Lcn2 KO male mice exhibit an increase in the levels of CLs containing long-chain polyunsaturated fatty acids (LC-PUFA), a decrease in CLs containing monounsaturated fatty acids, resulting in mitochondrial dysfunction. This dysfunction triggers compensatory activation of peroxisomal function and the biosynthesis of LC-PUFA-containing plasmalogens in BAT. Additionally, Lcn2 deficiency alters PA production, correlating with changes in PA-regulated phospholipid-metabolizing enzymes and the mTOR signaling pathway. In conclusion, LCN2 plays a critical role in the acyl-chain remodeling of phospholipids and mitochondrial bioenergetics by regulating PA production and its function in activating signaling pathways.
Collapse
Affiliation(s)
- Hongming Su
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Hong Guo
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Xiaoxue Qiu
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Te-Yueh Lin
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Gail Celio
- University Imaging Centers, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Peter Yong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Mark Senders
- University Imaging Centers, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Xiaoli Chen
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA.
| |
Collapse
|
36
|
Choudhary A, Yu J, Kouznetsova VL, Kesari S, Tsigelny IF. Two-Stage Deep-Learning Classifier for Diagnostics of Lung Cancer Using Metabolites. Metabolites 2023; 13:1055. [PMID: 37887380 PMCID: PMC10609149 DOI: 10.3390/metabo13101055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
We developed a machine-learning system for the selective diagnostics of adenocarcinoma (AD), squamous cell carcinoma (SQ), and small-cell carcinoma lung (SC) cancers based on their metabolomic profiles. The system is organized as two-stage binary classifiers. The best accuracy for classification is 92%. We used the biomarkers sets that contain mostly metabolites related to cancer development. Compared to traditional methods, which exclude hierarchical classification, our method splits a challenging multiclass task into smaller tasks. This allows a two-stage classifier, which is more accurate in the scenario of lung cancer classification. Compared to traditional methods, such a "divide and conquer strategy" gives much more accurate and explainable results. Such methods, including our algorithm, allow for the systematic tracking of each computational step.
Collapse
Affiliation(s)
- Ashvin Choudhary
- School of Life Science, University of California, Los Angeles, CA 90095, USA;
| | - Jianpeng Yu
- School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Valentina L. Kouznetsova
- San Diego Supercomputer Center, University of California, San Diego, CA 92093, USA;
- IUL, La Jolla, CA 92038, USA
- CureScience Institute, San Diego, CA 92121, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute, Santa Monica, CA 90404, USA;
| | - Igor F. Tsigelny
- San Diego Supercomputer Center, University of California, San Diego, CA 92093, USA;
- IUL, La Jolla, CA 92038, USA
- CureScience Institute, San Diego, CA 92121, USA
- Department of Neurosciences, University of California, San Diego, CA 92093, USA
| |
Collapse
|
37
|
Su YA, Chiu HY, Chang YC, Sung CJ, Chen CW, Tei R, Huang XR, Hsu SC, Lin SS, Wang HC, Lin YC, Hsu JC, Bauer H, Feng Y, Baskin JM, Chang ZF, Liu YW. NME3 binds to phosphatidic acid and mediates PLD6-induced mitochondrial tethering. J Cell Biol 2023; 222:e202301091. [PMID: 37584589 PMCID: PMC10432850 DOI: 10.1083/jcb.202301091] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2023] [Revised: 06/10/2023] [Accepted: 07/26/2023] [Indexed: 08/17/2023] Open
Abstract
Mitochondria are dynamic organelles regulated by fission and fusion processes. The fusion of membranes requires elaborative coordination of proteins and lipids and is particularly crucial for the function and quality control of mitochondria. Phosphatidic acid (PA) on the mitochondrial outer membrane generated by PLD6 facilitates the fusion of mitochondria. However, how PA promotes mitochondrial fusion remains unclear. Here, we show that a mitochondrial outer membrane protein, NME3, is required for PLD6-induced mitochondrial tethering or clustering. NME3 is enriched at the contact interface of two closely positioned mitochondria depending on PLD6, and NME3 binds directly to PA-exposed lipid packing defects via its N-terminal amphipathic helix. The PA binding function and hexamerization confer NME3 mitochondrial tethering activity. Importantly, nutrient starvation enhances the enrichment efficiency of NME3 at the mitochondrial contact interface, and the tethering ability of NME3 contributes to fusion efficiency. Together, our findings demonstrate NME3 as a tethering protein promoting selective fusion between PLD6-remodeled mitochondria for quality control.
Collapse
Affiliation(s)
- You-An Su
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Chiu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chieh-Ju Sung
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Wei Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Reika Tei
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Xuang-Rong Huang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shao-Chun Hsu
- Imaging Core, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shan-Shan Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsien-Chu Wang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Jui-Cheng Hsu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Hermann Bauer
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Yuxi Feng
- Department of Experimental Pharmacology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jeremy M. Baskin
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Zee-Fen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
38
|
Lu K, Fang B, Liu Y, Xu F, Zhou C, Wang L, Chen L, Huang L. Metabolomics Analysis of DRG and Serum in the CCI Model of Mice. Brain Sci 2023; 13:1224. [PMID: 37626580 PMCID: PMC10452726 DOI: 10.3390/brainsci13081224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2023] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Neuropathic pain (NP) is a chronic and intractable disease that is widely present in the general population. It causes painful behavior and even mood changes such as anxiety and depression by altering the metabolism of substances. However, there have been limited metabolomics studies conducted in relation to neuropathic pain. Therefore, in this study, the effects of NP on metabolites in serum and the dorsal root ganglion (DRG) were investigated using a non-targeted metabolomics approach detected by gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS) to uncover differential metabolites and affected metabolic pathways associated with NP. Sixty mice were divided into the following two groups: a chronic constriction injury (CCI) of the sciatic nerve group and a sham group (n = 30, each). After 7 days of CCI modeling, the metabolite profiles of serum and the DRG were analyzed using GC/LC-MS for both the CCI and sham groups of mice. Multivariate analysis revealed differential metabolites and altered metabolic pathways between the CCI and sham groups. In the CCI group, our findings provided insights into the complex phospholipid, amino acid and acylcarnitine metabolic perturbations of DRG metabolism. In addition, phospholipid metabolic disorders and impaired glucose metabolism were observed in the serum. Moreover, the metabolic differences in the DRG and serum were correlated with each other. The results from this untargeted metabolomics study provide a perspective on the metabolic impact of NP on serum and the DRG.
Collapse
Affiliation(s)
- Kaimei Lu
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| | - Bin Fang
- Department of Anesthesiology, Shanghai General Hospital, Nanjing Medical University, Shanghai 200080, China;
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| | - Fangxia Xu
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| | - Chengcheng Zhou
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| | - Lijuan Wang
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| | - Lianhua Chen
- Department of Anesthesiology, Shanghai General Hospital, Nanjing Medical University, Shanghai 200080, China;
| | - Lina Huang
- Department of Anesthesiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China; (K.L.); (Y.L.); (F.X.); (C.Z.); (L.W.)
| |
Collapse
|
39
|
Nyenhuis SB, Wu X, Strub MP, Yim YI, Stanton AE, Baena V, Syed ZA, Canagarajah B, Hammer JA, Hinshaw JE. OPA1 helical structures give perspective to mitochondrial dysfunction. Nature 2023; 620:1109-1116. [PMID: 37612506 DOI: 10.1038/s41586-023-06462-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/06/2022] [Accepted: 07/19/2023] [Indexed: 08/25/2023]
Abstract
Dominant optic atrophy is one of the leading causes of childhood blindness. Around 60-80% of cases1 are caused by mutations of the gene that encodes optic atrophy protein 1 (OPA1), a protein that has a key role in inner mitochondrial membrane fusion and remodelling of cristae and is crucial for the dynamic organization and regulation of mitochondria2. Mutations in OPA1 result in the dysregulation of the GTPase-mediated fusion process of the mitochondrial inner and outer membranes3. Here we used cryo-electron microscopy methods to solve helical structures of OPA1 assembled on lipid membrane tubes, in the presence and absence of nucleotide. These helical assemblies organize into densely packed protein rungs with minimal inter-rung connectivity, and exhibit nucleotide-dependent dimerization of the GTPase domains-a hallmark of the dynamin superfamily of proteins4. OPA1 also contains several unique secondary structures in the paddle domain that strengthen its membrane association, including membrane-inserting helices. The structural features identified in this study shed light on the effects of pathogenic point mutations on protein folding, inter-protein assembly and membrane interactions. Furthermore, mutations that disrupt the assembly interfaces and membrane binding of OPA1 cause mitochondrial fragmentation in cell-based assays, providing evidence of the biological relevance of these interactions.
Collapse
Affiliation(s)
- Sarah B Nyenhuis
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Xufeng Wu
- Light Microscopy Facility, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Marie-Paule Strub
- Protein Expression Facility, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Yang-In Yim
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Abigail E Stanton
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
- Molecular Biology Department, Princeton University, Princeton, NJ, USA
| | - Valentina Baena
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Zulfeqhar A Syed
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Bertram Canagarajah
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - John A Hammer
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Jenny E Hinshaw
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
40
|
Kowaleski SJ, Hurmis CS, Coleman CN, Philips KD, Najor NA. SHE9 deletion mutants display fitness defects during diauxic shift in Saccharomyces cerevisiae . MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000899. [PMID: 37577108 PMCID: PMC10422129 DOI: 10.17912/micropub.biology.000899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Academic Contribution Register] [Received: 06/19/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023]
Abstract
Saccharomyces cerevisiae protein She9 is localized to the inner mitochondrial membrane and is required for normal mitochondrial morphology. While deletion mutants of SHE9 ( she9Δ ) are viable and display large ring-like mitochondrial structures, the molecular function of SHE9 is still unknown. We report a decreased growth of she9Δ cells during a diauxic shift, where mitochondria are primarily employing oxidative phosphorylation to generate ATP versus the alternative mechanism of glycolysis in high glucose conditions. Further bioinformatics analysis reveal putative functional protein associations, and proposes a model to aid in the understanding of the molecular function of She9.
Collapse
|
41
|
Al Amir Dache Z, Thierry AR. Mitochondria-derived cell-to-cell communication. Cell Rep 2023; 42:112728. [PMID: 37440408 DOI: 10.1016/j.celrep.2023.112728] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/08/2022] [Revised: 02/21/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
In addition to their intracellular mobility, mitochondria and their components can exist outside the cells from which they originate. As a result, they are capable of acting on non-parental distant cells and mediate intercellular communication in physiological conditions and in a variety of pathologies. It has recently been demonstrated that this horizontal transfer governs a wide range of biological processes, such as tissue homeostasis, the rescue of injured recipient cells, and tumorigenesis. In addition, due to mitochondria's bacterial ancestry, they and their components can be recognized as damage-associated molecular patterns (DAMPs) by the immune cells, leading to inflammation. Here, we provide an overview of the most current and significant findings concerning the different structures of extracellular mitochondria and their by-products and their functions in the physiological and pathological context. This account illustrates the ongoing expansion of our understanding of mitochondria's biological role and functions in mammalian organisms.
Collapse
Affiliation(s)
- Zahra Al Amir Dache
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France; INSERM U1316, CNRS UMR7057, Université Paris Cité, Paris, France
| | - Alain R Thierry
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France; ICM, Institut Régional du Cancer de Montpellier, 34298 Montpellier, France.
| |
Collapse
|
42
|
Wang J, Yu X, Wang T, Cai W, Hua T, Duan J, Zhang X, Zhu Y, Yao L. Metabolic changes of glycerophospholipids during the reparative phase after myocardial infarction injury. Front Cardiovasc Med 2023; 10:1122571. [PMID: 37383698 PMCID: PMC10294426 DOI: 10.3389/fcvm.2023.1122571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2022] [Accepted: 05/30/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Myocardial infarction (MI) is a fatal manifestation of coronary heart disease, and its underlying mechanism is still largely unknown. Lipid levels and composition alterations predict the risk of MI complications. Glycerophospholipids (GPLs) are important bioactive lipids and play a crucial role in the development of cardiovascular diseases. However, the metabolic changes in the GPLs profile during post-MI injury remain unknown. Methods In the current study, we constructed a classic MI model by ligating the left anterior descending branch and assessed the alterations in both plasma and myocardial GPLs profiles during the reparative phase post-MI by liquid chromatography-tandem mass spectrometry analysis. Results We found that myocardial GPLs, but not plasma GPLs, were markedly changed after MI injury. Importantly, MI injury is associated with decreased phosphatidylserine (PS) levels. Consistently, the expression of phosphatidylserine synthase 1 (PSS1), which catalyzes the formation of PS from its substrate phosphatidylcholine, was significantly reduced in heart tissues after MI injury. Furthermore, oxygen-glucose deprivation (OGD) inhibited PSS1 expression and reduced PS levels in primary neonatal rat cardiomyocytes, while overexpression of PSS1 restored the inhibition of PSS1 and the reduction in PS levels caused by OGD. Moreover, overexpression of PSS1 abrogated, whereas knockdown of PSS1 aggravated, OGD-induced cardiomyocyte apoptosis. Conclusions Our findings revealed that GPLs metabolism was involved in the reparative phase post-MI, and cardiac decreased PS levels, resulting from inhibition of PSS1, are important contributor to the reparative phase post-MI. PSS1 overexpression represents a promising therapeutic strategy to attenuate MI injury.
Collapse
|
43
|
Bassal MA. The Interplay between Dysregulated Metabolism and Epigenetics in Cancer. Biomolecules 2023; 13:944. [PMID: 37371524 DOI: 10.3390/biom13060944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/24/2023] [Revised: 05/21/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular metabolism (or energetics) and epigenetics are tightly coupled cellular processes. It is arguable that of all the described cancer hallmarks, dysregulated cellular energetics and epigenetics are the most tightly coregulated. Cellular metabolic states regulate and drive epigenetic changes while also being capable of influencing, if not driving, epigenetic reprogramming. Conversely, epigenetic changes can drive altered and compensatory metabolic states. Cancer cells meticulously modify and control each of these two linked cellular processes in order to maintain their tumorigenic potential and capacity. This review aims to explore the interplay between these two processes and discuss how each affects the other, driving and enhancing tumorigenic states in certain contexts.
Collapse
Affiliation(s)
- Mahmoud Adel Bassal
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
44
|
Sequeira V, Waddingham MT, Tsuchimochi H, Maack C, Pearson JT. Mechano-energetic uncoupling in hypertrophic cardiomyopathy: Pathophysiological mechanisms and therapeutic opportunities. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100036. [PMID: 39801694 PMCID: PMC11708264 DOI: 10.1016/j.jmccpl.2023.100036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a frequent inherited form of heart failure. The underlying cause of HCM is generally attributed to mutations in genes that encode for sarcomeric proteins, but the pathogenesis of the disease is also influenced by non-genetic factors, which can contribute to diastolic dysfunction and hypertrophic remodeling. Central to the pathogenesis of HCM is hypercontractility, a state that is an antecedent to several key derangements, including increased mitochondrial workload and oxidative stress. As a result, energy depletion and mechano-energetic uncoupling drive cardiac growth through signaling pathways such as ERK and/or potentially AMPK downregulation. Metabolic remodeling also occurs in HCM, characterized by decreased fatty acid oxidation and increased glucose uptake. In some instances, ketones may also feed the heart with energy and act as signaling molecules to reduce oxidative stress and hypertrophic signaling. In addition, arrhythmias are frequently triggered in HCM, resulting from the high Ca2+-buffering of the myofilaments and changes in the ATP/ADP ratio. Understanding the mechanisms driving the progression of HCM is critical to the development of effective therapeutic strategies. This paper presents evidence from both experimental and clinical studies that support the role of hypercontractility and cellular energy alterations in the progression of HCM towards heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - Mark T. Waddingham
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Christoph Maack
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
- Department of Physiology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
45
|
He Q, Qu M, Shen T, Su J, Xu Y, Xu C, Barkat MQ, Cai J, Zhu H, Zeng LH, Wu X. Control of mitochondria-associated endoplasmic reticulum membranes by protein S-palmitoylation: Novel therapeutic targets for neurodegenerative diseases. Ageing Res Rev 2023; 87:101920. [PMID: 37004843 DOI: 10.1016/j.arr.2023.101920] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/07/2022] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Mitochondria-associated endoplasmic reticulum membranes (MAMs) are dynamic coupling structures between mitochondria and the endoplasmic reticulum (ER). As a new subcellular structure, MAMs combine the two critical organelle functions. Mitochondria and the ER could regulate each other via MAMs. MAMs are involved in calcium (Ca2+) homeostasis, autophagy, ER stress, lipid metabolism, etc. Researchers have found that MAMs are closely related to metabolic syndrome and neurodegenerative diseases (NDs). The formation of MAMs and their functions depend on specific proteins. Numerous protein enrichments, such as the IP3R-Grp75-VDAC complex, constitute MAMs. The changes in these proteins govern the interaction between mitochondria and the ER; they also affect the biological functions of MAMs. S-palmitoylation is a reversible protein post-translational modification (PTM) that mainly occurs on protein cysteine residues. More and more studies have shown that the S-palmitoylation of proteins is closely related to their membrane localization. Here, we first briefly describe the composition and function of MAMs, reviewing the component and biological roles of MAMs mediated by S-palmitoylation, elaborating on S-palmitoylated proteins in Ca2+ flux, lipid rafts, and so on. We try to provide new insight into the molecular basis of MAMs-related diseases, mainly NDs. Finally, we propose potential drug compounds targeting S-palmitoylation.
Collapse
Affiliation(s)
- Qiangqiang He
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Pharmacology, Hangzhou City University, Hangzhou 310015, China
| | - Meiyu Qu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tingyu Shen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiakun Su
- Technology Center, China Tobacco Jiangxi Industrial Co. Ltd., Nanchang 330096, China
| | - Yana Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Muhammad Qasim Barkat
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jibao Cai
- Technology Center, China Tobacco Jiangxi Industrial Co. Ltd., Nanchang 330096, China
| | - Haibin Zhu
- Department of Gynecology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Hangzhou City University, Hangzhou 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
46
|
Mukherjee S, Das S, Bedi M, Vadupu L, Ball WB, Ghosh A. Methylglyoxal-mediated Gpd1 activation restores the mitochondrial defects in a yeast model of mitochondrial DNA depletion syndrome. Biochim Biophys Acta Gen Subj 2023; 1867:130328. [PMID: 36791826 DOI: 10.1016/j.bbagen.2023.130328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/10/2022] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Human MPV17, an evolutionarily conserved mitochondrial inner-membrane channel protein, accounts for the tissue-specific mitochondrial DNA depletion syndrome. However, the precise molecular function of the MPV17 protein is still elusive. Previous studies showed that the mitochondrial morphology and cristae organization are severely disrupted in the MPV17 knockout cells from yeast, zebrafish, and mammalian tissues. As mitochondrial cristae morphology is strictly regulated by the membrane phospholipids composition, we measured mitochondrial membrane phospholipids (PLs) levels in yeast Saccharomyces cerevisiae MPV17 ortholog, SYM1 (Stress-inducible Yeast MPV17) deleted cells. We found that Sym1 knockout decreases the mitochondrial membrane PL, phosphatidyl ethanolamine (PE), and inhibits respiratory growth at 37 ̊C on rich media. Both the oxygen consumption rate and the steady state expressions of mitochondrial complex II and super-complexes are compromised. Apart from mitochondrial PE defect a significant depletion of mitochondrial phosphatidyl-choline (PC) was noticed in the sym1∆ cells grown on synthetic media at both 30 ̊C and 37 ̊C temperatures. Surprisingly, exogenous supplementation of methylglyoxal (MG), an intrinsic side product of glycolysis, rescues the respiratory growth of Sym1 deficient yeast cells. Using a combination of molecular biology and lipid biochemistry, we uncovered that MG simultaneously restores both the mitochondrial PE/PC levels and the respiration by enhancing cytosolic NAD-dependent glycerol-3-phosphate dehydrogenase 1 (Gpd1) enzymatic activity. Further, MG is incapable to restore respiratory growth of the sym1∆gpd1∆ double knockout cells. Thus, our work provides Gpd1 activation as a novel strategy for combating Sym1 deficiency and PC/PE defects.
Collapse
Affiliation(s)
- Soumyajit Mukherjee
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata Pin-700019, India
| | - Shubhojit Das
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata Pin-700019, India
| | - Minakshi Bedi
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata Pin-700019, India
| | - Lavanya Vadupu
- Department of the Biological Sciences, SRM University- AP, Andhra Pradesh Pin- 522240, India
| | - Writoban Basu Ball
- Department of the Biological Sciences, SRM University- AP, Andhra Pradesh Pin- 522240, India
| | - Alok Ghosh
- Department of Biochemistry, University of Calcutta, 35 Ballygunge Circular Road, Kolkata Pin-700019, India.
| |
Collapse
|
47
|
Wu G, Cheng H, Guo H, Li Z, Li D, Xie Z. Tea polyphenol EGCG ameliorates obesity-related complications by regulating lipidomic pathway in leptin receptor knockout rats. J Nutr Biochem 2023; 118:109349. [PMID: 37085056 DOI: 10.1016/j.jnutbio.2023.109349] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/23/2022] [Revised: 03/20/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023]
Abstract
Tea polyphenol EGCG has been widely recognized for antiobesity effects. However, the molecular mechanism of lipidomic pathway related to lipid-lowering effect of EGCG is still not well understood. The aim of this study was to investigate the effects and mechanism of EGCG activated hepatic lipidomic pathways on ameliorating obesity-related complications by using newly developed leptin receptor knockout (Lepr KO) rats. Results showed that EGCG supplementation (100 mg/kg body weight) significantly decreased total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and triglyceride (TG) levels both in the serum and liver, and significantly improved glucose intolerance. In addition, EGCG alleviated fatty liver development and restored the normal liver function in Lepr KO rats. Liver lipidomic analysis revealed that EGCG dramatically changes overall composition of lipid classes. Notably, EGCG significantly decreased an array of triglycerides (TGs) and diglycerides (DGs) levels. While EGCG increased 31 glycerophospholipid species and 1 sphingolipid species levels, such as phosphatidylcholines (PCs), phosphatidylethanolamines (PEs), phosphatidylserines (PSs) and phosphatidylinositols (PIs) levels in the liver of Lepr KO rats. Moreover, 14 diversely regulated lipid species were identified as potential lipid biomarkers. Mechanistic analysis revealed that EGCG significantly activated the SIRT6/AMPK/SREBP1/FAS pathway to decrease DGs and TGs levels and upregulated glycerophospholipids synthesis pathways to increase glycerophospholipid level in the liver of Lepr KO rats. These findings suggested that the regulation of glycerolipids and glycerophospholipid homeostasis might be the key pathways for EGCG in ameliorating obesity-related complications in Lepr KO rats.
Collapse
Affiliation(s)
- Guohuo Wu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Huijun Cheng
- College of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, PR China
| | - Huimin Guo
- Center for Biotechnology, Anhui Agricultural University, Anhui 230036, PR China
| | - Zhuang Li
- Center for Biotechnology, Anhui Agricultural University, Anhui 230036, PR China
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China.
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences & Technology, Anhui Agricultural University, Hefei, Anhui 230036, PR China; College of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, PR China.
| |
Collapse
|
48
|
Tsepaeva OV, Nemtarev AV, Pashirova TN, Khokhlachev MV, Lyubina AP, Amerkhanova SK, Voloshina AD, Mironov VF. Novel triphenylphosphonium amphiphilic conjugates of glycerolipid type: synthesis, cytotoxic and antibacterial activity, and targeted cancer cell delivery. RSC Med Chem 2023; 14:454-469. [PMID: 36970146 PMCID: PMC10034156 DOI: 10.1039/d2md00363e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/03/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
This work deals with the creation of new cationic triphenylphosphonium amphiphilic conjugates of glycerolipid type (TPP-conjugates), bearing a pharmacophore terpenoid fragment (abietic acid and betulin) and a fatty acid residue in one hybrid molecule as a new generation of antitumor agents with high activity and selectivity. The TPP-conjugates showed high mitochondriotropy leading to the development of mitochondriotropic delivery systems such as TPP-pharmacosomes and TPP-solid lipid particles. Introducing the betulin fragment into the structure of a TPP-conjugate (compound 10) increases the cytotoxicity 3 times towards tumor cells of prostate adenocarcinoma DU-145 and 4 times towards breast carcinoma MCF-7 compared to TPP-conjugate 4a in the absence of betulin. TPP-hybrid conjugate 10 with two pharmacophore fragments, betulin and oleic acid, has significant cytotoxicity toward a wide range of tumor cells. The lowest IC50 of 10 is 0.3 μM toward HuTu-80. This is at the level of the reference drug doxorubicin. TPP-pharmacosomes (10/PC) increased the cytotoxic effect approximately 3 times toward HuTu-80 cells, providing high selectivity (SI = 480) compared to the normal liver cell line Chang liver.
Collapse
Affiliation(s)
- Olga V Tsepaeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Andrey V Nemtarev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Tatiana N Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Michail V Khokhlachev
- Kazan (Volga Region) Federal University Kremlevskaya Str. 18 420008 Kazan Russian Federation
| | - Anna P Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Syumbelya K Amerkhanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Alexandra D Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| | - Vladimir F Mironov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov Str. 8 420088 Kazan Russian Federation
| |
Collapse
|
49
|
Jung J, Gokhale S, Xie P. TRAF3: A novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes. Front Oncol 2023; 13:1081253. [PMID: 36776285 PMCID: PMC9911533 DOI: 10.3389/fonc.2023.1081253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/27/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondria, the organelle critical for cell survival and metabolism, are exploited by cancer cells and provide an important therapeutic target in cancers. Mitochondria dynamically undergo fission and fusion to maintain their diverse functions. Proteins controlling mitochondrial fission and fusion have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control, and cell survival. In a recent proteomic study, we identified the key mitochondrial fission factor, MFF, as a new interacting protein of TRAF3, a known tumor suppressor of multiple myeloma and other B cell malignancies. This interaction recruits the majority of cytoplasmic TRAF3 to mitochondria, allowing TRAF3 to regulate mitochondrial morphology, mitochondrial functions, and mitochondria-dependent apoptosis in resting B lymphocytes. Interestingly, recent transcriptomic, metabolic and lipidomic studies have revealed that TRAF3 also vitally regulates multiple metabolic pathways in B cells, including phospholipid metabolism, glucose metabolism, and ribonucleotide metabolism. Thus, TRAF3 emerges as a novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes and B cell malignancies. Here we review current knowledge in this area and discuss relevant clinical implications.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
50
|
Zou B, Shao L, Yu Q, Zhao Y, Li X, Dai R. Changes of mitochondrial lipid molecules, structure, cytochrome c and ROS of beef Longissimus lumborum and Psoas major during postmortem storage and their potential associations with beef quality. Meat Sci 2023; 195:109013. [DOI: 10.1016/j.meatsci.2022.109013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022]
|