1
|
Rodríguez-Mortera R, Torres P, Fernàndez-Bernal A, Berdún R, Ramírez-Núñez O, Martín-Garí M, Serrano JC, He JC, Prat J, Pamplona R, Uribarri J, Portero-Otin M. Non-enzymatic modification of aminophospholipids induces angiogenesis, inflammation, and insulin signaling dysregulation in human renal glomerular endothelial cells in vitro. Free Radic Biol Med 2025; 235:15-24. [PMID: 40268103 DOI: 10.1016/j.freeradbiomed.2025.04.030] [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] [Scholar Register] [Received: 02/25/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
AIMS/HYPOTHESIS Advanced glycation end-products (AGEs) formation in proteins are involved in healthy aging and a variety of diseases including Alzheimer's disease, atherosclerosis, and diabetic complications. However, the biological effects of the non-enzymatic modification of aminophospholipids (lipid-AGEs) at cellular level are poorly understood. This study aimed to investigate the effects of lipid-AGEs on angiogenesis, inflammation, insulin signaling, and mitochondrial function in human renal glomerular endothelial cells (HRGEC), exploring their potential role in the pathophysiology of diabetic nephropathy (DN). METHODS HRGEC cells were exposed to non-enzymatically modified phosphatidylethanolamine (PE) by AGEs (lipid-AGEs), non-modified PE (nmPE) (aminophospholipid without modification), employed as a negative control, and lipopolysaccharides (LPS) as a positive control. Angiogenesis was assessed through vascular network formation metrics, including capillary area, junction density, and endpoints, under different extracellular matrices. Gene expression of inflammatory and angiogenic markers was quantified by RT-qPCR. Insulin signaling components, including IRS1 and AKT phosphorylation, were evaluated by immunoblotting. Mitochondrial function was assessed using high-resolution respirometry to determine ATP production rates from glycolysis and oxidative phosphorylation. RESULTS Lipid-AGEs induced dose-, time-, and matrix-dependent angiogenesis, with effects comparable to LPS, particularly in Engelbreth-Holm-Swarm extracellular matrix (ECM) (capillary area increase: 25 %, p < 0.05). Lipid-AGEs significantly upregulated the expression of inflammatory genes IL8 and NFKB (p < 0.05), and the angiogenesis-related markers TGFB1 and ANGPT2 (p < 0.05). Insulin signaling was disrupted, as lipid-AGEs enhanced inhibitory phosphorylation of IRS1 (Ser-1101, 1.8-fold increase, p < 0.01) and modulated AKT (Ser-473) and p42/p44 ERK activation. At lower doses, lipid-AGEs reduced eNOS phosphorylation (p < 0.05) impairing insulin responsiveness. High-resolution respirometry revealed that lipid-AGEs reduced basal oxygen consumption rates (OCR) by 20 % (p < 0.05), with no significant changes in glycolytic ATP production. CONCLUSION Lipid-AGEs induce angiogenesis, inflammation, and insulin signaling disruption in HRGEC, contributing to endothelial dysfunction. These findings underscore the potential role of lipid-AGEs in age-related decline of renal function, as well as the pathogenic potential in DN highlighting their relevance as therapeutic targets for mitigating vascular and metabolic complications in diabetes.
Collapse
Affiliation(s)
- Reyna Rodríguez-Mortera
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Pascual Torres
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Anna Fernàndez-Bernal
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Rebeca Berdún
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Omar Ramírez-Núñez
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Meritxell Martín-Garí
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - José Ce Serrano
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - John C He
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joan Prat
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Jaime Uribarri
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Lleida Biomedical Research Institute (UdL-IRBLleida), Lleida, Spain.
| |
Collapse
|
2
|
Attachaipanich T, Sriwichaiin S, Apaijai N, Thanyaratsarun T, Thongmung N, Vathesatogkit P, Sritara P, Chattipakorn N, Kitiyakara C, Chattipakorn SC. Obesity classified by anthropometric parameters was associated with mitochondrial bioenergetics impairment of peripheral blood mononuclear cells in the elderly population. Exp Gerontol 2025; 202:112724. [PMID: 40037474 DOI: 10.1016/j.exger.2025.112724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025]
Abstract
Waist circumference (WC), waist-to-height ratio (WHtR), and waist-to-hip ratio (WHR), even in individuals who have a normal body mass index (BMI), are correlated with cardiovascular events. The aim of this study is to establish the association between obesity and mitochondrial bioenergetics in peripheral blood mononuclear cells (PBMCs). The study included 1584 subjects from the Electricity Generating Authority of Thailand (EGAT) cohort. The mean age of participants in this study was 68.4 years. There was 24.2 % diabetes mellitus (DM) with a mean HbA1c level of 6.8. WC, WHR, and WHtR were associated with decreased basal, maximal respiration, spare respiratory capacity (SRC), and ATP production, whereas BMI was only associated with reduced maximal respiration and SRC. We further stratified the participants into four groups based on obesity classified by WHR and DM status: Non-DM/Non-obese (n = 468), Non-DM/Obese (n = 733), DM/Non-obese (n = 84), and DM/Obese (n = 299). Both obesity and DM were associated with mitochondrial bioenergetic impairment and increased mitochondrial oxidative stress. Interestingly, there was no difference in mitochondrial bioenergetics impairment between non-DM/Obese and DM participants. Our study demonstrated that WC, WHR, and WHtR better reflected underlying mitochondrial dysfunction in PBMCs compared to BMI. Furthermore, obesity was associated with mitochondrial dysfunction to the same degree as DM.
Collapse
Affiliation(s)
- Tanawat Attachaipanich
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thanaphat Thanyaratsarun
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nisakron Thongmung
- Office of Research Academic and Innovation, Faculty of Medicine, Ramathibodi Hospital Mahidol University, Bangkok 10400, Thailand
| | - Prin Vathesatogkit
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Piyamitr Sritara
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand.
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
3
|
Hill Z, McCarty PJ, Boles RG, Frye RE. A Mitochondrial Supplement Improves Function and Mitochondrial Activity in Autism: A Double-Blind Placebo-Controlled Cross-Over Trial. Int J Mol Sci 2025; 26:2479. [PMID: 40141123 PMCID: PMC11941969 DOI: 10.3390/ijms26062479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Autism spectrum disorder (ASD) is associated with mitochondrial dysfunction, but studies demonstrating the efficacy of treatments are scarce. We sought to determine whether a mitochondrial-targeted dietary supplement designed for children with ASD improved mitochondrial function and ASD symptomatology using a double-blind placebo-controlled cross-over design. Sixteen children [mean age 9 years 4 months; 88% male] with non-syndromic ASD and mitochondrial enzyme abnormalities, as measured by MitoSwab (Religen, Plymouth Meeting, PA, USA), received weight-adjusted SpectrumNeeds® (NeuroNeeds, Old Lyme, CT, USA) and QNeeds® (NeuroNeeds, Old Lyme, CT, USA) and placebos matched on taste, texture and appearance during two separate 12-week blocks. Which product was received first was randomized. The treatment significantly normalized citrate synthase and complex IV activity as measured by the MitoSwab. Mitochondrial respiration of peripheral blood mononuclear cell respiration, as measured by the Seahorse XFe96 (Agilent, Santa Clara, CA, USA) with the mitochondrial oxidative stress test, became more resilient to oxidative stress after the treatment, particularly in children with poor neurodevelopment. The mitochondrial supplement demonstrated significant improvement in standardized parent-rated scales in neurodevelopment, social withdrawal, and hyperactivity with large effect sizes (Cohen's d' = 0.77-1.25), while changes measured by the clinical and psychometric instruments were not significantly different. Adverse effects were minimal. This small study on children with ASD and mitochondrial abnormalities demonstrates that a simple, well-tolerated mitochondrial-targeted dietary supplement can improve mitochondrial physiology and ASD symptoms. Further larger controlled studies need to verify and extend these findings. These findings are significant as children with ASD have few other effective treatments.
Collapse
Affiliation(s)
- Zoë Hill
- Autism Discovery and Treatment Foundation, Phoenix, AZ 85050, USA;
| | | | - Richard G. Boles
- Mitochondrial and Molecular Medicine, Pasadena, CA 91105, USA;
- NeuroNeeds, Old Lyme, CT 06371, USA
| | - Richard E. Frye
- Autism Discovery and Treatment Foundation, Phoenix, AZ 85050, USA;
- Rossignol Medical Center, Phoenix, AZ 85050, USA
| |
Collapse
|
4
|
Zhang K, Zhu YW, Tang AQ, Zhou ZT, Yang YL, Liu ZH, Li Y, Liang XY, Feng ZF, Wang J, Jiang T, Jiang QY, Wu DD. Role of 3-mercaptopyruvate sulfurtransferase in cancer: Molecular mechanisms and therapeutic perspectives. Transl Oncol 2025; 52:102272. [PMID: 39813769 PMCID: PMC11783123 DOI: 10.1016/j.tranon.2025.102272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/10/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
The occurrence and development of tumor is mediated by a wide range of complex mechanisms. Subsequent to nitric oxide and carbon monoxide, hydrogen sulfide (H2S) holds the distinction of being the third identified gasotransmitter. Alternation of H2S level has been widely demonstrated to induce an array of disturbances in important cancer cell signaling pathways. As a result, the effects of H2S-catalyzing enzymes in cancers also attract widspread attention. 3-mercaptopyruvate sulfurtransferase (3-MST) is privileged to be one of them. In fact, 3-MST is overexpressed in many tumors including human colon cancer, lung adenocarcinoma, and bladder urothelial carcinoma. But it is also lowly expressed in hepatocellular carcinoma. In this review, we focus on the generation of endogenous H2S and polysulfides, facilitated by 3-MST. Additionally, we delve deeply into the potential role of 3-MST in tumorigenesis and development. The impact of 3-MST inhibition on the development of tumors and its potential for tumor therapy are also highlighted.
Collapse
Affiliation(s)
- Ka Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Ao-Qi Tang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Ze-Tao Zhou
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Lun Yang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Zi-Hui Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yan Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Xiao-Yi Liang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Zhi-Fen Feng
- School of Nursing and Health, Henan University, Kaifeng, Henan 475004, China
| | - Jun Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Tong Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
5
|
Moiz B, Walls M, Alpizar Vargas V, Addepalli A, Weber C, Li A, Sriram G, Clyne AM. Instationary metabolic flux analysis reveals that NPC1 inhibition increases glycolysis and decreases mitochondrial metabolism in brain microvascular endothelial cells. Neurobiol Dis 2025; 204:106769. [PMID: 39706535 DOI: 10.1016/j.nbd.2024.106769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
Niemann Pick Disease Type C (NP-C), a rare neurogenetic disease with no known cure, is caused by mutations in the cholesterol trafficking protein NPC1. Brain microvascular endothelial cells (BMEC) are thought to play a critical role in the pathogenesis of several neurodegenerative diseases; however, little is known about how these cells are altered in NP-C. In this study, we investigated how NPC1 inhibition perturbs BMEC metabolism in human induced pluripotent stem cell-derived BMEC (hiBMEC). We incorporated extracellular metabolite and isotope labeling data into an instationary metabolic flux analysis (INST-MFA) model to estimate intracellular metabolic fluxes. We found that NPC1 inhibition significantly increased glycolysis and pentose phosphate pathway flux while decreasing mitochondrial metabolism. These changes may have been driven by gene expression changes due to increased cholesterol biosynthesis, in addition to mitochondrial cholesterol accumulation. We corroborated these findings in primary BMEC, an alternative in vitro human brain endothelial model. Finally, we found that co-treatment with hydroxypropyl-β cyclodextrin (HPβCD) partially restored metabolic phenotype in U18666A-treated BMECs, suggesting that this drug may have therapeutic effects on the brain endothelium in NP-C. Together, our data highlight the importance of NPC1 in BMEC metabolism and implicate brain endothelial dysfunction in NP-C pathogenesis.
Collapse
Affiliation(s)
- Bilal Moiz
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Matthew Walls
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Viviana Alpizar Vargas
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Anirudh Addepalli
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Callie Weber
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Andrew Li
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America
| | - Ganesh Sriram
- Department of Chemical and Biochemical Engineering, University of Maryland, College Park, MD 20742, United States of America
| | - Alisa Morss Clyne
- Department of Bioengineering, University of Maryland, College Park, MD 20742, United States of America.
| |
Collapse
|
6
|
Rowland EC, D’Antuono M, Jermakowicz A, Ayad NG. MAT2a and AHCY inhibition disrupts antioxidant metabolism and reduces glioblastoma cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624981. [PMID: 39605416 PMCID: PMC11601785 DOI: 10.1101/2024.11.23.624981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant adult brain tumor that inevitably recurs with a fatal prognosis. This is due in part to metabolic reprogramming that allows tumors to evade treatment. We therefore must uncover the pathways mediating these adaptations to develop novel and effective treatments. We searched for genes that are essential in GBM cells as measured by a whole-genome pan-cancer CRISPR screen available from DepMap and identified the methionine metabolism genes MAT2A and AHCY. We conducted genetic knockdown, evaluated mitochondrial respiration, and performed targeted metabolomics to study the function of these genes in GBM. We demonstrate that MAT2A or AHCY knockdown induces oxidative stress, hinders cellular respiration, and reduces the survival of GBM cells. Furthermore, selective MAT2a or AHCY inhibition reduces GBM cell viability, impairs oxidative metabolism, and changes the metabolic profile of these cells towards oxidative stress and cell death. Mechanistically, MAT2a or AHCY regulates spare respiratory capacity, the redox buffer cystathionine, lipid and amino acid metabolism, and prevents DNA damage in GBM cells. Our results point to the methionine metabolic pathway as a novel vulnerability point in GBM.
Collapse
Affiliation(s)
- Emma C. Rowland
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Matthew D’Antuono
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Anna Jermakowicz
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Nagi G. Ayad
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| |
Collapse
|
7
|
Torres S, Hardesty J, Barrios M, Garcia-Ruiz C, Fernandez-Checa JC, Singal AK. Mitochondria and Alcohol-Associated Liver Disease: Pathogenic Role and Target for Therapy. Semin Liver Dis 2024. [PMID: 39317216 DOI: 10.1055/a-2421-5658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Alcohol-associated liver disease (ALD) is one of the leading causes of chronic liver disease and a major cause of liver-related death. ALD is a multifactorial disease triggered by the oxidative metabolism of alcohol which leads to the activation of multiple factors that promote the progression from steatosis to more advanced stages like alcohol-associated steatohepatitis (AH) that culminate in alcohol-associated cirrhosis and hepatocellular carcinoma. Poor understanding of the complex heterogeneous pathology of ALD has limited drug development for this disease. Alterations in mitochondrial performance are considered a crucial event in paving the progression of ALD due to the crucial role of mitochondria in energy production, intermediate metabolism, calcium homeostasis, and cell fate decisions. Therefore, understanding the role of mitochondria in eliciting steatosis and progression toward AH may open the door to new opportunities for treatment. In this review, we will cover the physiological function of mitochondria, its contribution to ALD in experimental models and human disease, and explore whether targeting mitochondria may represent a game changer in the treatment of ALD.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Josiah Hardesty
- Division of Gastroenterology and Hepatology, University of Louisville, Louisville, Kentucky
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Monica Barrios
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ashwani K Singal
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Unidad Associada IMIM/IIBB-CSIC, Barcelona, Spain
- Division of Gastroenterology and Hepatology, University of Louisville, Louisville, Kentucky
- Transplant Hepatology, Trager Transplant Center and Jewish Hospital, University of Health, Louisville, Kentucky
- Department of Clinical Research, Robley Rex VA Medical Center, Louisville, Kentucky
| |
Collapse
|
8
|
Hough RF, Alvira CM, Bastarache JA, Erzurum SC, Kuebler WM, Schmidt EP, Shimoda LA, Abman SH, Alvarez DF, Belvitch P, Bhattacharya J, Birukov KG, Chan SY, Cornfield DN, Dudek SM, Garcia JGN, Harrington EO, Hsia CCW, Islam MN, Jonigk DD, Kalinichenko VV, Kolb TM, Lee JY, Mammoto A, Mehta D, Rounds S, Schupp JC, Shaver CM, Suresh K, Tambe DT, Ventetuolo CE, Yoder MC, Stevens T, Damarla M. Studying the Pulmonary Endothelium in Health and Disease: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 71:388-406. [PMID: 39189891 PMCID: PMC11450313 DOI: 10.1165/rcmb.2024-0330st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Lung endothelium resides at the interface between the circulation and the underlying tissue, where it senses biochemical and mechanical properties of both the blood as it flows through the vascular circuit and the vessel wall. The endothelium performs the bidirectional signaling between the blood and tissue compartments that is necessary to maintain homeostasis while physically separating both, facilitating a tightly regulated exchange of water, solutes, cells, and signals. Disruption in endothelial function contributes to vascular disease, which can manifest in discrete vascular locations along the artery-to-capillary-to-vein axis. Although our understanding of mechanisms that contribute to endothelial cell injury and repair in acute and chronic vascular disease have advanced, pathophysiological mechanisms that underlie site-specific vascular disease remain incompletely understood. In an effort to improve the translatability of mechanistic studies of the endothelium, the American Thoracic Society convened a workshop to optimize rigor, reproducibility, and translation of discovery to advance our understanding of endothelial cell function in health and disease.
Collapse
|
9
|
Tamarindo GH, Ribeiro CF, Silva ADT, Castro A, Caruso ÍP, Souza FP, Taboga SR, Loda M, Góes RM. The polyunsaturated fatty acid docosahexaenoic affects mitochondrial function in prostate cancer cells. Cancer Metab 2024; 12:24. [PMID: 39113152 PMCID: PMC11308158 DOI: 10.1186/s40170-024-00348-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) shows a rewired metabolism featuring increased fatty acid uptake and synthesis via de novo lipogenesis, both sharply related to mitochondrial physiology. The docosahexaenoic acid (DHA) is an omega-3 polyunsaturated fatty acid (PUFA) that exerts its antitumoral properties via different mechanisms, but its specific action on mitochondria in PCa is not clear. Therefore, we investigated whether the DHA modulates mitochondrial function in PCa cell lines. METHODS Here, we evaluated mitochondrial function of non-malignant PNT1A and the castration-resistant (CRPC) prostate 22Rv1 and PC3 cell lines in response to DHA incubation. For this purpose, we used Seahorse extracellular flux assay to assess mitochondria function, [14C]-glucose to evaluate its oxidation as well as its contribution to fatty acid synthesis, 1H-NMR for metabolite profile determination, MitoSOX for superoxide anion production, JC-1 for mitochondrial membrane polarization, mass spectrometry for determination of phosphatidylglycerol levels and composition, staining with MitoTracker dye to assess mitochondrial morphology under super-resolution in addition to Transmission Electron Microscopy, In-Cell ELISA for COX-I and SDH-A protein expression and flow cytometry (Annexin V and 7-AAD) for cell death estimation. RESULTS In all cell lines DHA decreased basal respiratory activity, ATP production, and the spare capacity in mitochondria. Also, the omega-3 induced mitochondrial hyperpolarization, ROS overproduction and changes in membrane phosphatidylglycerol composition. In PNT1A, DHA led to mitochondrial fragmentation and it increased glycolysis while in cancer cells it stimulated glucose oxidation, but decreased de novo lipogenesis specifically in 22Rv1, indicating a metabolic shift. In all cell lines, DHA modulated several metabolites related to energy metabolism and it was incorporated in phosphatidylglycerol, a precursor of cardiolipin, increasing the unsaturation index in the mitochondrial membrane. Accordingly, DHA triggered cell death mainly in PNT1A and 22Rv1. CONCLUSION In conclusion, mitochondrial metabolism is significantly affected by the PUFA supplementation to the point that cells are not able to proliferate or survive under DHA-enriched condition. Moreover, combination of DHA supplementation with inhibition of metabolism-related pathways, such as de novo lipogenesis, may be synergistic in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Guilherme Henrique Tamarindo
- Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | | | - Alana Della Torre Silva
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil
| | - Alex Castro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Ícaro Putinhon Caruso
- Department of Biophysics, Institute of Biosciences, Humanities and Exact Science, São Paulo State University, São José Do Rio Preto, São Paulo, Brazil
- Institute of Medical Biochemistry and National Center for Structure Biology and Bioimaging (CENABIO), National Center for Nuclear Magnetic Resonance of Macromolecules, Federal University of Rio de Janeiro, Ilha Do Fundão, Rio de Janeiro, Brazil
| | - Fátima Pereira Souza
- Department of Biophysics, Institute of Biosciences, Humanities and Exact Science, São Paulo State University, São José Do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Rejane Maira Góes
- Department of Biological Sciences, IBILCE - UNESP. Rua Cristovão Colombo, 2265 Jardim Nazareth, São José Do Rio Preto, São Paulo, 15054-000, Brazil.
| |
Collapse
|
10
|
Attachaipanich T, Sriwichaiin S, Apaijai N, Kerdphoo S, Thongmung N, Vathesatogkit P, Sritara P, Chattipakorn N, Kitiyakara C, Chattipakorn SC. An Increase in Vascular Stiffness Is Positively Associated With Mitochondrial Bioenergetics Impairment of Peripheral Blood Mononuclear Cells in the Older Adults. J Gerontol A Biol Sci Med Sci 2024; 79:glae095. [PMID: 38567632 DOI: 10.1093/gerona/glae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Indexed: 04/04/2024] Open
Abstract
The cardio-ankle vascular index (CAVI) is a noninvasive parameter reflecting vascular stiffness. CAVI correlates with the burden of atherosclerosis and future cardiovascular events. Mitochondria of peripheral blood mononuclear cells (PBMCs) have been identified as a noninvasive source for assessing systemic mitochondrial bioenergetics. This study aimed to investigate the relationship between CAVI values and mitochondrial bioenergetics of PBMCs in the older adults.. This cross-sectional study enrolled participants from the Electricity Generating Authority of Thailand between 2017 and 2018. A total of 1 640 participants with an ankle-brachial index greater than 0.9 were included in this study. All participants were stratified into 3 groups based on their CAVI values as high (CAVI ≥ 9), moderate (9 > CAVI ≥ 8), and low (CAVI < 8), in which each group comprised 702, 507, and 431 participants, respectively. The extracellular flux analyzer was used to measure mitochondrial respiration of isolated PBMCs. The mean age of the participants was 67.9 years, and 69.6% of them were male. After adjusted with potential confounders including age, sex, smoking status, body mass index, diabetes, dyslipidemia, hypertension, and creatinine clearance, participants with high CAVI values were independently associated with impaired mitochondrial bioenergetics, including decreased basal respiration, maximal respiration, and spare respiratory capacity, as well as increased mitochondrial reactive oxygen species. This study demonstrated that CAVI measurement reflects the underlying impairment of cellular mitochondrial bioenergetics in PBMCs. Further longitudinal studies are necessary to establish both a causal relationship between CAVI measurement and underlying cellular dysfunction.
Collapse
Affiliation(s)
- Tanawat Attachaipanich
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sasiwan Kerdphoo
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nisakron Thongmung
- Office of Research Academic and Innovation, Faculty of Medicine Ramathibodi Hospital Mahidol University, Bangkok, Thailand
| | - Prin Vathesatogkit
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital Mahidol University, Bangkok, Thailand
| | - Piyamitr Sritara
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital Mahidol University, Bangkok, Thailand
| | - Nipon Chattipakorn
- Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok
| | - Chagriya Kitiyakara
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital Mahidol University, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Faculty of Medicine, Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
11
|
Nunes PR, Oliveira PF, Rebelo I, Sandrim VC, Alves MG. Relevance of real-time analyzers to determine mitochondrial quality in endothelial cells and oxidative stress in preeclampsia. Vascul Pharmacol 2024; 155:107372. [PMID: 38583694 DOI: 10.1016/j.vph.2024.107372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Oxidative stress and mitochondrial dysfunction are important elements for the pathophysiology of preeclampsia (PE), a multisystemic hypertensive syndrome of pregnancy, characterized by endothelial dysfunction and responsible for a large part of maternal and fetal morbidity and mortality worldwide. Researchers have dedicated their efforts to unraveling the intricate ways in which certain molecules influence both energy metabolism and oxidative stress. Exploring established methodologies from existing literature, shows that these investigations predominantly focus on the placenta, identified as a pivotal source that drives the changes observed in the disease. In this review, we discuss the role of oxidative stress in pathophysiology of PE, as well as metabolic/endothelial dysfunction. We further discuss the use of seahorse analyzers to study real-time bioenergetics of endothelial cells. Although the benefits are clear, few studies have presented results using this method to assess mitochondrial metabolism in these cells. We performed a search on MEDLINE/PubMed using the terms "Seahorse assay and endothelial dysfunction in HUVEC" as well as "Seahorse assay and preeclampsia". From our research, we selected 16 original peer-review papers for discussion. Notably, the first search retrieved studies involving Human Umbilical Vein Endothelial Cells (HUVECs) but none investigating bioenergetics in PE while the second search retrieved studies exploring the technique in PE but none of the studies used HUVECs. Additional studies are required to investigate real-time mitochondrial bioenergetics in PE. Clearly, there is a need for more complete studies to examine the nuances of mitochondrial bioenergetics, focusing on the contributions of HUVECs in the context of PE.
Collapse
Affiliation(s)
- Priscila R Nunes
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), 18618-689 Sao Paulo, Brazil
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Irene Rebelo
- UCIBIO-REQUIMTE, Laboratory of Biochemistry, Department of Biologic Sciences, Pharmaceutical Faculty, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB- Institute for Health and Bioeconomy, Laboratory of Biochemistry, Department of Biologic Sciences, Pharmaceutical Faculty, University of Porto, 4050-313 Porto, Portugal
| | - Valeria C Sandrim
- Department of Pharmacology and Biophysics, Institute of Biosciences, Sao Paulo State University (Unesp), 18618-689 Sao Paulo, Brazil
| | - Marco G Alves
- iBiMED - Institute of Biomedicine and Department of Medical Sciences University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
12
|
Ang BJ, Suardi N, Abduraman MA. Exploring differentiation-dependent responses to 532 nm green laser photobiomodulation in SHSY5Y neuroblastoma cells. Lasers Med Sci 2024; 39:147. [PMID: 38822930 DOI: 10.1007/s10103-024-04102-2] [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] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Photobiomodulation (PBM) holds promise as a therapy modality, but its applicability is hindered by the lack of a quantitative model to predict the optimal dose for all forms of PBM. This study investigated the optimal PBM parameters for 532 nm green laser irradiation on SHSY5Y neuroblastoma cells, a commonly used in vitro model for neurodegenerative disease studies. A two-tailed, two sample t-test with equal variance was used to obtain the p-values and statistical significance. There are 3 sets of parameters showing significant ( p < 0 . 01 ) positive percentage biostimulation. 160 m W , 15 m i n produce a percentage biostimulation of ( 9 ± 10 ) % ; 180 m W , 5 m i n produce a percentage biostimulation of ( 19 ± 7 ) % ; and ( 200 m W , 5 m i n ) produce a percentage biostimulation of ( 9 ± 2 ) % . The highest significant ( p < 0 . 01 ) percentage bioinhibition observed is for 220 m W , 15 m i n (dose: 1008 J / c m 2 ) producing a bioinhibition of ( 54 ± 1 ) % . After identifying several parameters that produce noticeable photobiological effects (biostimulation and bioinhibition), this study compared the reaction of undifferentiated and differentiated SHSY5Y cells to laser irradiation and found that undifferentiated SHSY5Y cells shows greater photobiological effect from 532 nm laser irradiation ( p < 0 . 01 ) . This study demonstrated the differentiation-dependant photobiological effect of SHSY5Y in 532 nm laser PBM. This shows that considerations on the differentiation state of cells is important in PBM studies. The hypothesis of difference in intracellular reactive oxygen species (ROS) accumulation from laser irradiation can serve as a versatile explanation of the observed difference in photobiological effect. Further investigation into the role of ROS as a mediator of various photobiological effects from laser of different wavelengths is warranted.
Collapse
Affiliation(s)
- Beng Jiong Ang
- School of Physics, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Nursakinah Suardi
- School of Physics, Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| | - Muhammad Asyraf Abduraman
- Eman Biodiscoveries Sdn Bhd, A1-4, Lot 5, Persiaran 2/1, Kedah Halal Park, Sungai Petani Industrial Area, 08000, Sungai Petani, Kedah, Malaysia
| |
Collapse
|
13
|
Chen X, Xu Y, Ju Y, Gu P. Metabolic Regulation of Endothelial Cells: A New Era for Treating Wet Age-Related Macular Degeneration. Int J Mol Sci 2024; 25:5926. [PMID: 38892113 PMCID: PMC11172501 DOI: 10.3390/ijms25115926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Wet age-related macular degeneration (wet AMD) is a primary contributor to visual impairment and severe vision loss globally, but the prevailing treatments are often unsatisfactory. The development of conventional treatment strategies has largely been based on the understanding that the angiogenic switch of endothelial cells (ECs) is mainly dictated by angiogenic growth factors. Even though treatments targeting vascular endothelial growth factor (VEGF), like ranibizumab, are widely administered, more than half of patients still exhibit inadequate or null responses, suggesting the involvement of other pathogenic mechanisms. With advances in research in recent years, it has become well recognized that EC metabolic regulation plays an active rather than merely passive responsive role in angiogenesis. Disturbances of these metabolic pathways may lead to excessive neovascularization in angiogenic diseases such as wet AMD, therefore targeted modulation of EC metabolism represents a promising therapeutic strategy for wet AMD. In this review, we comprehensively discuss the potential applications of EC metabolic regulation in wet AMD treatment from multiple perspectives, including the involvement of ECs in wet AMD pathogenesis, the major endothelial metabolic pathways, and novel therapeutic approaches targeting metabolism for wet AMD.
Collapse
Affiliation(s)
- Xirui Chen
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yang Xu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Yahan Ju
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (X.C.)
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
14
|
Boullard NG, Paris JJ, Shariat-Madar Z, Mahdi F. Increased Prolylcarboxypeptidase Expression Can Serve as a Biomarker of Senescence in Culture. Molecules 2024; 29:2219. [PMID: 38792081 PMCID: PMC11123917 DOI: 10.3390/molecules29102219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Prolylcarboxypeptidase (PRCP, PCP, Lysosomal Pro-X-carboxypeptidase, Angiotensinase C) controls angiotensin- and kinin-induced cell signaling. Elevation of PRCP appears to be activated in chronic inflammatory diseases [cardiovascular disease (CVD), diabetes] in proportion to severity. Vascular endothelial cell senescence and mitochondrial dysfunction have consistently been shown in models of CVD in aging. Cellular senescence, a driver of age-related dysfunction, can differentially alter the expression of lysosomal enzymes due to lysosomal membrane permeability. There is a lack of data demonstrating the effect of age-related dysfunction on the expression and function of PRCP. To explore the changes in PRCP, the PRCP-dependent prekallikrein (PK) pathway was characterized in early- and late-passage human pulmonary artery endothelial cells (HPAECs). Detailed kinetic analysis of cells treated with high molecular weight kininogen (HK), a precursor of bradykinin (BK), and PK revealed a mechanism by which senescent HPAECs activate the generation of kallikrein upon the assembly of the HK-PK complex on HPAECs in parallel with an upregulation of PRCP and endothelial nitric oxide (NO) synthase (eNOS) and NO formation. The NO production and expression of both PRCP and eNOS increased in early-passage HPAECs and decreased in late-passage HPAECs. Low activity of PRCP in late-passage HPAECs was associated with rapid decreased telomerase reverse transcriptase mRNA levels. We also found that, with an increase in the passage number of HPAECs, reduced PRCP altered the respiration rate. These results indicated that aging dysregulates PRCP protein expression, and further studies will shed light into the complexity of the PRCP-dependent signaling pathway in aging.
Collapse
Affiliation(s)
| | - Jason J. Paris
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.J.P.); (Z.S.-M.)
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.J.P.); (Z.S.-M.)
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.J.P.); (Z.S.-M.)
| |
Collapse
|
15
|
Matrella ML, Valletti A, Gigante I, De Rasmo D, Signorile A, Russo S, Lobasso S, Lobraico D, Dibattista M, Pacelli C, Cocco T. High OXPHOS efficiency in RA-FUdr-differentiated SH-SY5Y cells: involvement of cAMP signalling and respiratory supercomplexes. Sci Rep 2024; 14:7411. [PMID: 38548913 PMCID: PMC10978939 DOI: 10.1038/s41598-024-57613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/20/2024] [Indexed: 04/01/2024] Open
Abstract
Neurons are highly dependent on mitochondria to meet their bioenergetic needs and understanding the metabolic changes during the differentiation process is crucial in the neurodegeneration context. Several in vitro approaches have been developed to study neuronal differentiation and bioenergetic changes. The human SH-SY5Y cell line is a widely used cellular model and several differentiation protocols have been developed to induce a neuron-like phenotype including retinoic acid (RA) treatment. In this work we obtained a homogeneous functional population of neuron-like cells by a two-step differentiation protocol in which SH-SY5Y cells were treated with RA plus the mitotic inhibitor 2-deoxy-5-fluorouridine (FUdr). RA-FUdr treatment induced a neuronal phenotype characterized by increased expression of neuronal markers and electrical properties specific to excitable cells. In addition, the RA-FUdr differentiated cells showed an enrichment of long chain and unsaturated fatty acids (FA) in the acyl chain composition of cardiolipin (CL) and the bioenergetic analysis evidences a high coupled and maximal respiration associated with high mitochondrial ATP levels. Our results suggest that the observed high oxidative phosphorylation (OXPHOS) capacity may be related to the activation of the cyclic adenosine monophosphate (cAMP) pathway and the assembly of respiratory supercomplexes (SCs), highlighting the change in mitochondrial phenotype during neuronal differentiation.
Collapse
Affiliation(s)
- Maria Laura Matrella
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Alessio Valletti
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
- MASMEC Biomed S.p.A, 70026, Modugno, Italy
| | - Isabella Gigante
- National Institute of Gastroenterology- IRCCS "Saverio De Bellis", Via Turi 27, Castellana Grotte, 70013, Bari, Italy
| | - Domenico De Rasmo
- Bioenergetics and Molecular Biotechnologies, CNR-Institute of Biomembranes, 70124, Bari, Italy
| | - Anna Signorile
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Silvia Russo
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Simona Lobasso
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Donatella Lobraico
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Michele Dibattista
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy.
| | - Tiziana Cocco
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124, Bari, Italy.
| |
Collapse
|
16
|
Ay M, Charli A, Langley M, Jang A, Padhi P, Jin H, Anantharam V, Kalyanaraman B, Kanthasamy A, Kanthasamy AG. Mito-metformin protects against mitochondrial dysfunction and dopaminergic neuronal degeneration by activating upstream PKD1 signaling in cell culture and MitoPark animal models of Parkinson's disease. Front Neurosci 2024; 18:1356703. [PMID: 38449738 PMCID: PMC10915001 DOI: 10.3389/fnins.2024.1356703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Impaired mitochondrial function and biogenesis have strongly been implicated in the pathogenesis of Parkinson's disease (PD). Thus, identifying the key signaling mechanisms regulating mitochondrial biogenesis is crucial to developing new treatment strategies for PD. We previously reported that protein kinase D1 (PKD1) activation protects against neuronal cell death in PD models by regulating mitochondrial biogenesis. To further harness the translational drug discovery potential of targeting PKD1-mediated neuroprotective signaling, we synthesized mito-metformin (Mito-Met), a mitochondria-targeted analog derived from conjugating the anti-diabetic drug metformin with a triphenylphosphonium functional group, and then evaluated the preclinical efficacy of Mito-Met in cell culture and MitoPark animal models of PD. Mito-Met (100-300 nM) significantly activated PKD1 phosphorylation, as well as downstream Akt and AMPKα phosphorylation, more potently than metformin, in N27 dopaminergic neuronal cells. Furthermore, treatment with Mito-Met upregulated the mRNA and protein expression of mitochondrial transcription factor A (TFAM) implying that Mito-Met can promote mitochondrial biogenesis. Interestingly, Mito-Met significantly increased mitochondrial bioenergetics capacity in N27 dopaminergic cells. Mito-Met also reduced mitochondrial fragmentation induced by the Parkinsonian neurotoxicant MPP+ in N27 cells and protected against MPP+-induced TH-positive neurite loss in primary neurons. More importantly, Mito-Met treatment (10 mg/kg, oral gavage for 8 week) significantly improved motor deficits and reduced striatal dopamine depletion in MitoPark mice. Taken together, our results demonstrate that Mito-Met possesses profound neuroprotective effects in both in vitro and in vivo models of PD, suggesting that pharmacological activation of PKD1 signaling could be a novel neuroprotective translational strategy in PD and other related neurocognitive diseases.
Collapse
Affiliation(s)
- Muhammet Ay
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Adhithiya Charli
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Monica Langley
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Ahyoung Jang
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Piyush Padhi
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Huajun Jin
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Vellareddy Anantharam
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | | | - Arthi Kanthasamy
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| |
Collapse
|
17
|
Shimada K, Lu Y, Ikawa M. Disruption of testis-enriched cytochrome c oxidase subunit COX6B2 but not COX8C leads to subfertility. Exp Anim 2024; 73:1-10. [PMID: 37423748 PMCID: PMC10877148 DOI: 10.1538/expanim.23-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023] Open
Abstract
Mammalian sperm flagellum contains the midpiece characterized by a mitochondrial sheath that packs tightly around the axoneme and outer dense fibers. Mitochondria are known as the "powerhouse" of the cell, and produce ATP through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS). However, the contribution of the TCA cycle and OXPHOS to sperm motility and male fertility is less clear. Cytochrome c oxidase (COX) is an oligomeric complex localized within the mitochondrial inner membrane, and the terminal enzyme of the mitochondrial electron transport chain in eukaryotes. Both COX6B2 and COX8C are testis-enriched COX subunits whose functions in vivo are poorly studied. Here, we generated Cox6b2 and Cox8c knockout (KO) mice using the CRISPR/Cas9 system. We examined their fertility and sperm mitochondrial function to determine the significance of testis-enriched COX subunits in male fertility. The mating test revealed that disrupting COX6B2 induces male subfertility, while disrupting COX8C does not affect male fertility. Cox6b2 KO spermatozoa showed low sperm motility, but mitochondrial function was normal according to oxygen consumption rates. Therefore, low sperm motility seems to cause subfertility in Cox6b2 KO male mice. These results also indicate that testis-enriched COX, COX6B2 and COX8C, are not essential for OXPHOS in mouse spermatozoa.
Collapse
Affiliation(s)
- Keisuke Shimada
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yonggang Lu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
18
|
Motawe ZY, Abdelmaboud SS, Breslin JW. Evaluation of Glycolysis and Mitochondrial Function in Endothelial Cells Using the Seahorse Analyzer. Methods Mol Biol 2024; 2711:241-256. [PMID: 37776463 PMCID: PMC11368073 DOI: 10.1007/978-1-0716-3429-5_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
Endothelial bioenergetics have emerged as a key regulator of endothelial barrier function. Glycolytic parameters have been linked to barrier enhancement, and interruption with mitochondrial complexes was shown to disrupt endothelial barrier. Therefore, a new technology that has been introduced to assess bioenergetics and metabolism has also made it possible to determine roles of specific energy production pathways in endothelial health. The Seahorse extracellular flux analysis by Agilent technologies is a state of the art tool that has been more frequently used to evaluate bioenergetics of endothelial cells. This chapter includes details about different assays that can be used to study endothelial cells using the Seahorse analyzer and how interpretation of the results can provide novel insight about endothelial metabolism.
Collapse
Affiliation(s)
- Zeinab Y Motawe
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Salma S Abdelmaboud
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
19
|
Gao T, Cheng S, Lu H, Li X, Weng X, Ge J. Histidine Triad Nucleotide-Binding Protein 1 Improves Critical Limb Ischemia by Regulating Mitochondrial Homeostasis. Nutrients 2023; 15:4859. [PMID: 38068718 PMCID: PMC10708213 DOI: 10.3390/nu15234859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/11/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Critical limb ischemia (CLI) is a common complication of diabetes mellitus that typically occurs in the later stages of the disease. Vascularization is indeed an important physiological process involving the formation of new blood vessels from existing ones. It occurs in response to various normal and pathophysiological conditions, and one of its critical roles is to compensate for inadequate oxygen supply, which is often seen in situations like chronic limb ischemia (CLI). Histidine triad nucleotide-binding protein 1 (Hint1) is a member of the Hint family that has been shown to attenuate cardiac hypertrophy, but its role in vascularization still needs to be clarified. In this study, we investigated the role of Hint1 in CLI. We found that Hint1 is significantly reduced in the muscle tissue of STZ-induced diabetic mice and high-glucose (HG)-treated endothelial cells (ECs). Hint1 deletion impaired blood flow recovery and vascularization, whereas Hint1 overexpression promoted these processes. In addition, our in vitro study showed that Hint1 deficiency aggravated mitochondrial dysfunction in ECs, as evidenced by impaired mitochondrial respiration, decreased mitochondrial membrane potential, and increased reactive oxygen species. Our findings suggest that Hint1 deficiency impairs blood perfusion by damaging mitochondrial function and that Hint1 may represent a potential therapeutic target for treating CLI.
Collapse
Affiliation(s)
- Tingwen Gao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiology, Rizhao International Heart Hospital, Rizhao 276825, China
| | - Shuo Cheng
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiao Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China; (T.G.); (H.L.); (X.L.)
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
20
|
Alsharabasy AM, Aljaabary A, Bohara R, Farràs P, Glynn SA, Pandit A. Nitric Oxide-Scavenging, Anti-Migration Effects, and Glycosylation Changes after Hemin Treatment of Human Triple-Negative Breast Cancer Cells: A Mechanistic Study. ACS Pharmacol Transl Sci 2023; 6:1416-1432. [PMID: 37854626 PMCID: PMC10580390 DOI: 10.1021/acsptsci.3c00115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Indexed: 10/20/2023]
Abstract
The enhanced expression of nitric oxide (•NO) synthase predicts triple-negative breast cancer outcome and its resistance to different therapeutics. Our earlier work demonstrated the efficiency of hemin to scavenge the intra- and extracellular •NO, proposing its potency as a therapeutic agent for inhibiting cancer cell migration. In continuation, the present work evaluates the effects of •NO on the migration of MDA-MB-231 cells and how hemin modulates the accompanied cellular behavior, focusing on the corresponding expression of cellular glycoproteins, migration-associated markers, and mitochondrial functions. We demonstrated for the first time that while •NO induced cell migration, hemin contradicted that by •NO-scavenging. This was in combination with modulation of the •NO-enhanced glycosylation patterns of cellular proteins with inhibition of the expression of specific proteins involved in the epithelial-mesenchymal transition. These effects were in conjunction with changes in the mitochondrial functions related to both •NO, hemin, and its nitrosylated product. Together, these results suggest that hemin can be employed as a potential anti-migrating agent targeting •NO-scavenging and regulating the expression of migration-associated proteins.
Collapse
Affiliation(s)
- Amir M. Alsharabasy
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Amal Aljaabary
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Raghvendra Bohara
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Pau Farràs
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- School
of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway H91 TK33, Ireland
| | - Sharon A. Glynn
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- Discipline
of Pathology, Lambe Institute for Translational Research, School of
Medicine, University of Galway, Galway H91 YR71, Ireland
| | - Abhay Pandit
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
21
|
Bottermann K, Spychala A, Eliacik A, Amin E, Moussavi-Torshizi SE, Klöcker N, Gödecke A, Heinen A. Extracellular flux analysis in intact cardiac tissue slices-A novel tool to investigate cardiac substrate metabolism in mouse myocardium. Acta Physiol (Oxf) 2023; 239:e14004. [PMID: 37227741 DOI: 10.1111/apha.14004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/17/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
AIM Cardiac pathologies are accompanied by alterations in substrate metabolism, and extracellular flux analysis is a standard tool to investigate metabolic disturbances, especially in immortalized cell lines. However, preparations of primary cells, such as adult cardiomyocytes require enzymatic dissociation and cultivation affecting metabolism. Therefore, we developed a flux analyzer-based method for the assessment of substrate metabolism in intact vibratome-sliced mouse heart tissue. METHODS Oxygen consumption rates were determined using a Seahorse XFe24-analyzer and "islet capture plates." We demonstrate that tissue slices are suitable for extracellular flux analysis and metabolize both free fatty acids (FFA) and glucose/glutamine. Functional integrity of tissue slices was proven by optical mapping-based assessment of action potentials. In a proof-of-principle approach, the sensitivity of the method was tested by analyzing substrate metabolism in the remote myocardium after myocardial infarction (I/R). RESULTS Here, I/R increased uncoupled OCR compared with sham animals indicating a stimulated metabolic capacity. This increase was caused by a higher glucose/glutamine metabolism, whereas FFA oxidation was unchanged. CONCLUSION In conclusion, we describe a novel method to analyze cardiac substrate metabolism in intact cardiac tissue slices by extracellular flux analysis. The proof-of-principle experiment demonstrated that this approach has a sensitivity allowing the investigation of pathophysiologically relevant disturbances in cardiac substrate metabolism.
Collapse
Affiliation(s)
- Katharina Bottermann
- Institute for Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andre Spychala
- Institute for Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Asena Eliacik
- Institute for Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Ehsan Amin
- Institute of Neural und Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - S Erfan Moussavi-Torshizi
- Institute of Neural und Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neural und Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Axel Gödecke
- Institute for Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Andre Heinen
- Institute for Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Minzaghi D, Pavel P, Kremslehner C, Gruber F, Oberreiter S, Hagenbuchner J, Del Frari B, Blunder S, Gruber R, Dubrac S. Excessive Production of Hydrogen Peroxide in Mitochondria Contributes to Atopic Dermatitis. J Invest Dermatol 2023; 143:1906-1918.e8. [PMID: 37085042 DOI: 10.1016/j.jid.2023.03.1680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/07/2023] [Accepted: 03/30/2023] [Indexed: 04/23/2023]
Abstract
Atopic dermatitis (AD) is a complex disease characterized by chronic recurring eczema and pruritus. In addition, patients with AD display increased cutaneous and systemic levels of oxidative damage markers, whose source remains elusive. In this study, we investigated oxidative and mitochondrial stress in AD epidermis. The levels of superoxide dismutase 2 and hydrogen peroxide are augmented in the mitochondria of flaky tail (ft/ft) mouse keratinocytes, which is associated with the inhibition of the glutathione system and catalase. Furthermore, reduced levels of glutathione peroxidase 4 are associated with accumulation of malondialdehyde, 4-hydroxy-2-nonenal, and oxidized phosphatidylcholines in ft/ft epidermis. Cytochrome c is markedly increased in ft/ft epidermis, hence showing mitochondrial stress. Topical application of MitoQ, which is a mitochondrial-targeting antioxidant, to ft/ft mouse skin reduced damage to macromolecules and inflammation and restored epidermal homeostasis. Absence of alteration in the expression of superoxide dismutase 2, catalase, and glutathione peroxidase 4 and limited lipid peroxidation as well as oxidized phosphatidylcholines in the epidermis of Flg-/- mice suggest that FLG deficiency marginally contributes to oxidative stress in ft/ft epidermis. Increased superoxide dismutase 2, lipid peroxidation, and cytochrome c in the epidermis of patients with AD, associated with reduced antioxidant response in primary AD keratinocytes, corroborate mitochondrial dysfunction and lack of cellular adjustment to oxidative stress in AD epidermis.
Collapse
Affiliation(s)
- Deborah Minzaghi
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Petra Pavel
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sophie Oberreiter
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Blunder
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
23
|
Zoccarato A. Extending the cardiac metabolism toolkit: Bringing extracellular flux analyses to the living cardiac tissue slice. Acta Physiol (Oxf) 2023; 239:e14032. [PMID: 37548383 DOI: 10.1111/apha.14032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
24
|
Dragoni S, Turowski P. Vascular Signalling. Cells 2023; 12:2038. [PMID: 37626847 PMCID: PMC10453014 DOI: 10.3390/cells12162038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
In all vertebrates, closed blood and open lymph circulatory systems are essential for the delivery of nutrients and oxygen to tissues, waste clearance, and immune function [...].
Collapse
Affiliation(s)
- Silvia Dragoni
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Patric Turowski
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
25
|
Yao ZK, Jean YH, Lin SC, Lai YC, Chen NF, Tseng CC, Chen WF, Wen ZH, Kuo HM. Manoalide Induces Intrinsic Apoptosis by Oxidative Stress and Mitochondrial Dysfunction in Human Osteosarcoma Cells. Antioxidants (Basel) 2023; 12:1422. [PMID: 37507960 PMCID: PMC10376204 DOI: 10.3390/antiox12071422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor that produces immature osteoid. Metastatic OS has a poor prognosis with a death rate of >70%. Manoalide is a natural sesterterpenoid isolated from marine sponges. It is a phospholipase A2 inhibitor with anti-inflammatory, analgesic, and anti-cancer properties. This study aimed to investigate the mechanism and effect of manoalide on OS cells. Our experiments showed that manoalide induced cytotoxicity in 143B and MG63 cells (human osteosarcoma). Treatment with manoalide at concentrations of 10, 20, and 40 µM for 24 and 48 h reduced MG63 cell viability to 45.13-4.40% (p < 0.01). Meanwhile, manoalide caused reactive oxygen species (ROS) overproduction and disrupted antioxidant proteins, activating the apoptotic proteins caspase-9/-3 and PARP (Poly (ADP-ribose) polymerase). Excessive levels of ROS in the mitochondria affected oxidative phosphorylation, ATP generation, and membrane potential (ΔΨm). Additionally, manoalide down-regulated mitochondrial fusion protein and up-regulated mitochondrial fission protein, resulting in mitochondrial fragmentation and impaired function. On the contrary, a pre-treatment with n-acetyl-l-cysteine ameliorated manoalide-induced apoptosis, ROS, and antioxidant proteins in OS cells. Overall, our findings show that manoalide induces oxidative stress, mitochondrial dysfunction, and apoptosis, causing the cell death of OS cells, showing potential as an innovative alternative treatment in human OS.
Collapse
Affiliation(s)
- Zhi-Kang Yao
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung 90059, Taiwan
| | - Sung-Chun Lin
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung 90059, Taiwan
| | - Yu-Cheng Lai
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Orthopedics, Asia University Hospital, Taichung 41354, Taiwan
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Center for General Education, Cheng Shiu University, Kaohsiung 833301, Taiwan
| | - Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833301, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Hsiao-Mei Kuo
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833301, Taiwan
| |
Collapse
|
26
|
Lee WE, Genetzakis E, Figtree GA. Novel Strategies in the Early Detection and Treatment of Endothelial Cell-Specific Mitochondrial Dysfunction in Coronary Artery Disease. Antioxidants (Basel) 2023; 12:1359. [PMID: 37507899 PMCID: PMC10376062 DOI: 10.3390/antiox12071359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Although elevated cholesterol and other recognised cardiovascular risk factors are important in the development of coronary artery disease (CAD) and heart attack, the susceptibility of humans to this fatal process is distinct from other animals. Mitochondrial dysfunction of cells in the arterial wall, particularly the endothelium, has been strongly implicated in the pathogenesis of CAD. In this manuscript, we review the established evidence and mechanisms in detail and explore the potential opportunities arising from analysing mitochondrial function in patient-derived cells such as endothelial colony-forming cells easily cultured from venous blood. We discuss how emerging technology and knowledge may allow us to measure mitochondrial dysfunction as a potential biomarker for diagnosis and risk management. We also discuss the "pros and cons" of animal models of atherosclerosis, and how patient-derived cell models may provide opportunities to develop novel therapies relevant for humans. Finally, we review several targets that potentially alleviate mitochondrial dysfunction working both via direct and indirect mechanisms and evaluate the effect of several classes of compounds in the cardiovascular context.
Collapse
Affiliation(s)
- Weiqian E. Lee
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Elijah Genetzakis
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (W.E.L.); (E.G.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
27
|
Ma ZR, Li HP, Cai SZ, Du SY, Chen X, Yao J, Cao X, Zhen YF, Wang Q. The mitochondrial protein TIMM44 is required for angiogenesis in vitro and in vivo. Cell Death Dis 2023; 14:307. [PMID: 37147302 PMCID: PMC10163060 DOI: 10.1038/s41419-023-05826-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
The mitochondrial integrity and function in endothelial cells are essential for angiogenesis. TIMM44 (translocase of inner mitochondrial membrane 44) is essential for integrity and function of mitochondria. Here we explored the potential function and the possible mechanisms of TIMM44 in angiogenesis. In HUVECs, human retinal microvascular endothelial cells and hCMEC/D3 brain endothelial cells, silence of TIMM44 by targeted shRNA largely inhibited cell proliferation, migration and in vitro capillary tube formation. TIMM44 silencing disrupted mitochondrial functions in endothelial cells, causing mitochondrial protein input arrest, ATP reduction, ROS production, and mitochondrial depolarization, and leading to apoptosis activation. TIMM44 knockout, by Cas9-sgRNA strategy, also disrupted mitochondrial functions and inhibited endothelial cell proliferation, migration and in vitro capillary tube formation. Moreover, treatment with MB-10 ("MitoBloCK-10"), a TIMM44 blocker, similarly induced mitochondrial dysfunction and suppressed angiogenic activity in endothelial cells. Contrarily, ectopic overexpression of TIMM44 increased ATP contents and augmented endothelial cell proliferation, migration and in vitro capillary tube formation. In adult mouse retinas, endothelial knockdown of TIMM44, by intravitreous injection of endothelial specific TIMM44 shRNA adenovirus, inhibited retinal angiogenesis, causing vascular leakage, acellular capillary growth, and retinal ganglion cells degeneration. Significant oxidative stress was detected in TIMM44-silenced retinal tissues. Moreover, intravitreous injection of MB-10 similarly induced oxidative injury and inhibited retinal angiogenesis in vivo. Together, the mitochondrial protein TIMM44 is important for angiogenesis in vitro and in vivo, representing as a novel and promising therapeutic target of diseases with abnormal angiogenesis.
Collapse
Affiliation(s)
- Zhou-Rui Ma
- Department of Burns and Plastic Surgery, Children's hospital of Soochow University, Suzhou, China
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
| | - Hong-Peng Li
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Shi-Zhong Cai
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China
- Department of Child and Adolescent Healthcare, Children's Hospital of Soochow University, Suzhou, China
| | - Sheng-Yang Du
- Department of Orthopedics, Xuzhou First People's Hospital, Xuzhou, China
| | - Xia Chen
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.
| | - Xu Cao
- Suzhou Key Laboratory of Children's Structural Deformities, Suzhou, China.
- Department of Urology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yun-Fang Zhen
- Department of Orthopedics, Children's hospital of Soochow University, Suzhou, China.
| | - Qian Wang
- Department of Anesthesiology, Children's hospital of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Mas-Bargues C, Sanz-Ros J, Romero-García N, Huete-Acevedo J, Dromant M, Borrás C. Small extracellular vesicles from senescent stem cells trigger adaptive mechanisms in young stem cells by increasing antioxidant enzyme expression. Redox Biol 2023; 62:102668. [PMID: 36965438 PMCID: PMC10060362 DOI: 10.1016/j.redox.2023.102668] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/27/2023] Open
Abstract
Extracellular vesicles' biogenesis, shedding, and uptake are redox-sensitive. Indeed, oxidative stress conditions influence extracellular vesicles' release and content, which can modulate the redox status of the receiving cells. In this study, we aimed to assess the effect of extracellular vesicles from human dental pulp stem cells cultured under 21% O2 (senescent stem cells) on human dental pulp stem cells cultured under 3% O2 (young stem cells). Extracellular vesicles were isolated by ultracentrifugation from senescent stem cells and prepared for the treatment of young stem cells at a final concentration of 10 μg/mL. Cells were analyzed for antioxidant gene expression, mitochondrial bioenergetic parameters, ROS production, culture kinetics, and apoptosis. The results show that extracellular vesicles from senescent stem cells induce overexpression of antioxidant genes (MnSOD, CAT, and GPx) in young stem cells, which show an increased non-mitochondrial oxygen consumption, accompanied by reduced maximal respiration and spare respiratory capacity without altering mitochondrial membrane potential. This is accompanied by improved cell proliferation, viability, and migration rates and a reduction of apoptosis. In conclusion, extracellular vesicles from senescent stem cells trigger an adaptive response in young stem cells which improves their antioxidant defenses and their proliferation, migration, and survival rates. This suggests that extracellular vesicles can modulate the cells' microenvironment and the balance between proliferation and senescence.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain; Department of Cardiology, Hospital Universitari I Politècnic La Fe, 46026, Valencia, Spain.
| | - Nekane Romero-García
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain; Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari de Valencia, 46010, Valencia, Spain.
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| |
Collapse
|
29
|
Song Y, Leem J, Dhanani M, McKirnan MD, Ichikawa Y, Braza J, Harrington EO, Hammond HK, Roth DM, Patel HH. Impact of blood factors on endothelial cell metabolism and function in two diverse heart failure models. PLoS One 2023; 18:e0281550. [PMID: 36780477 PMCID: PMC9924994 DOI: 10.1371/journal.pone.0281550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Role of blood-based factors in development and progression of heart failure (HF) is poorly characterized. Blood contains factors released during pathophysiological states that may impact cellular function and provide mechanistic insights to HF management. We tested effects of blood from two distinct HF models on cardiac metabolism and identified possible cellular targets of the effects. Blood plasma was obtained from daunorubicin- and myocardial infarction-induced HF rabbits (Dauno-HF and MI-HF) and their controls (Dauno-Control and MI-Control). Effects of plasma on bioenergetics of myocardial tissue from healthy mice and cellular cardiac components were assessed using high-resolution respirometry and Seahorse flux analyzer. Since endothelial cell respiration was profoundly affected by HF plasma, effects of plasma on endothelial cell barrier function and death were further evaluated. Western-blotting and electron microscopy were performed to evaluate mitochondrial proteins and morphology. Brief exposure to HF plasma decreased cardiac tissue respiration. Endothelial cell respiration was most impacted by exposure to HF plasma. Endothelial cell monolayer integrity was decreased by incubation with Dauno-HF plasma. Apoptosis and necrosis were increased in cells incubated with Dauno-HF plasma for 24 h. Down-regulation of voltage-dependent anion-selective channel (VDAC)-1, translocase of outer membrane 20 (Tom20), and mitochondrial fission factor (MFF) in cells exposed to Dauno-HF plasma and mitochondrial signal transducer and activator of transcription 3 (Stat3) and MFF in cells exposed to MI-HF plasma were observed. Mitochondrial structure was disrupted in cells exposed to HF plasma. These findings indicate that endothelial cells and mitochondrial structure and function may be primary target where HF pathology manifests and accelerates. High-throughput blood-based screening of HF may provide innovative ways to advance disease diagnosis and management.
Collapse
Affiliation(s)
- Young Song
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joseph Leem
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - Mehul Dhanani
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - M. Dan McKirnan
- Department of Medicine, UCSD School of Medicine, San Diego, CA, United States of America
| | - Yasuhiro Ichikawa
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - Julie Braza
- Department of Medicine, Brown University and the Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, United States of America
| | - Elizabeth O. Harrington
- Department of Medicine, Brown University and the Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, United States of America
| | - H. Kirk Hammond
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Medicine, UCSD School of Medicine, San Diego, CA, United States of America
| | - David M. Roth
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Anesthesiology, UCSD School of Medicine, San Diego, CA, United States of America
| | - Hemal H. Patel
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Anesthesiology, UCSD School of Medicine, San Diego, CA, United States of America
| |
Collapse
|
30
|
Sun H, Zheng M, Liu J, Fan W, He H, Huang F. Melatonin promoted osteogenesis of human periodontal ligament cells by regulating mitochondrial functions through the translocase of the outer mitochondrial membrane 20. J Periodontal Res 2023; 58:53-69. [PMID: 36373245 DOI: 10.1111/jre.13068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Melatonin plays an important role in various beneficial functions, including promoting differentiation. However, effects on osteogenic differentiation, especially in human periodontal cells (hPDLCs), still remain inconclusive. Mitochondria are highly dynamic organelles that play an important role in various biological processes in cells, including energy metabolism and oxidative stress reaction. Furthermore, the translocase of the outer mitochondrial membrane 20 (TOM20) is responsible for recognizing and transporting precursor proteins. Thus, the objective of this study was to evaluate the functionality of melatonin on osteogenesis in human periodontal cells and to explore the involved mechanism of mitochondria. METHODS The hPDLCs were extracted and identified by flow cytometry and multilineage differentiation. We divided hPDLCs into control group, osteogenic induction group, and osteogenesis with melatonin treatment group (100, 10, and 1 μM). Then we used a specific siRNA to achieve interference of TOM20. Alizarin red and Alkaline phosphatase staining and activity assays were performed to evaluate osteogenic differentiation. Osteogenesis-related genes and proteins were measured by qPCR and western blot. Mitochondrial functions were tested using ATP, NAD+/NADH, JC-1, and Seahorse Mito Stress Test kits. Finally, TOM20 and mitochondrial dynamics-related molecules expression were also assessed by qPCR and western blot. RESULTS Our results showed that melatonin-treated hPDLCs had higher calcification and ALP activity as well as upregulated OCN and Runx2 expression at mRNA and protein levels, which was the most obvious in 1 μM melatonin-treated group. Meanwhile, melatonin supplement elevated intracellular ATP production and mitochondrial membrane potential by increasing mitochondrial oxidative metabolism, hence causing a lower NAD+ /NADH ratio. In addition, we also found that melatonin treatment raised TOM20 level and osteogenesis and mitochondrial functions were both suppressed after knocking down TOM20. CONCLUSION We found that melatonin promoted osteogenesis of hPDLCs and 1 μM melatonin had the most remarkable effect. Melatonin treatment can reinforce mitochondrial functions by upregulating TOM20.
Collapse
Affiliation(s)
- Haoyun Sun
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Miaomiao Zheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jiawei Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
31
|
Petrovskaya AV, Tverskoi AM, Barykin EP, Varshavskaya KB, Dalina AA, Mitkevich VA, Makarov AA, Petrushanko IY. Distinct Effects of Beta-Amyloid, Its Isomerized and Phosphorylated Forms on the Redox Status and Mitochondrial Functioning of the Blood-Brain Barrier Endothelium. Int J Mol Sci 2022; 24:ijms24010183. [PMID: 36613623 PMCID: PMC9820675 DOI: 10.3390/ijms24010183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The Alzheimer's disease (AD)-associated breakdown of the blood-brain barrier (BBB) promotes the accumulation of beta-amyloid peptide (Aβ) in the brain as the BBB cells provide Aβ transport from the brain parenchyma to the blood, and vice versa. The breakdown of the BBB during AD may be caused by the emergence of blood-borne Aβ pathogenic forms, such as structurally and chemically modified Aβ species; their effect on the BBB cells has not yet been studied. Here, we report that the effects of Aβ42, Aβ42, containing isomerized Asp7 residue (iso-Aβ42) or phosphorylated Ser8 residue (p-Aβ42) on the mitochondrial potential and respiration are closely related to the redox status changes in the mouse brain endothelial cells bEnd.3. Aβ42 and iso-Aβ42 cause a significant increase in nitric oxide, reactive oxygen species, glutathione, cytosolic calcium and the mitochondrial potential after 4 h of incubation. P-Aβ42 either does not affect or its effect develops after 24 h of incubation. Aβ42 and iso-Aβ42 activate mitochondrial respiration compared to p-Aβ42. The isomerized form promotes a greater cytotoxicity and mitochondrial dysfunction, causing maximum oxidative stress. Thus, Aβ42, p-Aβ42 and iso-Aβ42 isoforms differently affect the BBBs' cell redox parameters, significantly modulating the functioning of the mitochondria. The changes in the level of modified Aβ forms can contribute to the BBBs' breakdown during AD.
Collapse
|
32
|
Wilson C, Lee MD, Buckley C, Zhang X, McCarron JG. Mitochondrial ATP Production is Required for Endothelial Cell Control of Vascular Tone. FUNCTION 2022; 4:zqac063. [PMID: 36778749 PMCID: PMC9909368 DOI: 10.1093/function/zqac063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Arteries and veins are lined by nonproliferating endothelial cells that play a critical role in regulating blood flow. Endothelial cells also regulate tissue perfusion, metabolite exchange, and thrombosis. It is thought that endothelial cells rely on ATP generated via glycolysis, rather than mitochondrial oxidative phosphorylation, to fuel each of these energy-demanding processes. However, endothelial metabolism has mainly been studied in the context of proliferative cells, and little is known about energy production in endothelial cells within the fully formed vascular wall. Using intact arteries isolated from rats and mice, we show that inhibiting mitochondrial respiration disrupts endothelial control of vascular tone. Basal, mechanically activated, and agonist-evoked calcium activity in intact artery endothelial cells are each prevented by inhibiting mitochondrial ATP synthesis. Agonist-evoked calcium activity was also inhibited by blocking the transport of pyruvate, the master fuel for mitochondrial energy production, through the mitochondrial pyruvate carrier. The role for mitochondria in endothelial cell energy production is independent of species, sex, or vascular bed. These data show that a mitochondrial ATP supply is necessary for calcium-dependent, nitric oxide-mediated endothelial control of vascular tone, and identifies the critical role of endothelial mitochondrial energy production in fueling perfused blood vessel function.
Collapse
Affiliation(s)
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | | |
Collapse
|
33
|
Atallah R, Olschewski A, Heinemann A. Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1. Biomedicines 2022; 10:3089. [PMID: 36551845 PMCID: PMC9775124 DOI: 10.3390/biomedicines10123089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an essential process by which new blood vessels develop from existing ones. While adequate angiogenesis is a physiological process during, for example, tissue repair, insufficient and excessive angiogenesis stands on the pathological side. Fine balance between pro- and anti-angiogenic factors in the tissue environment regulates angiogenesis. Identification of these factors and how they function is a pressing topic to develop angiogenesis-targeted therapeutics. During the last decade, exciting data highlighted non-metabolic functions of intermediates of the mitochondrial Krebs cycle including succinate. Among these functions is the contribution of succinate to angiogenesis in various contexts and through different mechanisms. As the concept of targeting metabolism to treat a wide range of diseases is rising, in this review we summarize the mechanisms by which succinate regulates angiogenesis in normal and pathological settings. Gaining a comprehensive insight into how this metabolite functions as an angiogenic signal will provide a useful approach to understand diseases with aberrant or excessive angiogenic background, and may provide strategies to tackle them.
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
34
|
Piao S, Lin H, Tao X, Chen W. Mitochondrial toxicants in Xian-Ling-Gu-Bao induce liver injury by regulating the PI3K/mTOR signaling pathway: an in vitro study. BMC Complement Med Ther 2022; 22:317. [PMID: 36457007 PMCID: PMC9716976 DOI: 10.1186/s12906-022-03798-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Drug-induced mitochondrial toxicity is thought to be a common mechanism of drug hepatotoxicity. Xian-Ling-Gu-Bao (XLGB) oral preparation is a commonly used drug for osteoporosis in China. Classical safety evaluation studies have shown that the entire preparation and six Chinese herbal medicines have high safety, but the incidence of drug-induced liver damage due to XLGB remains high, the mechanism and toxic substances causing liver injury are still unclear. The purpose of this study is to identify compounds with potential mitochondrial liabilities in XLGB, and to clarify their underlying mechanisms and related pathways. METHODS The mitochondrial function analysis was performed using an extracellular flux assay, which simultaneously monitored both oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Through network pharmacology and in vitro experimental verification, the potential protein targets, signaling pathways and molecular mechanism of mitochondrial toxicity have been studied. RESULTS We observed a significant decrease in mitochondrial respiration of Psoraleae Fructus and its five compounds in fundamental bioenergetics parameters such as basal respiration, ATP-linked production and maximal respiration, indicating mitochondrial dysfunction. The network pharmacology results showed that the influence of XLGB on mitochondrial dysfunction was closely related to PI3K-Akt signaling pathway, mTOR signaling pathway and Apoptosis. Western blot showed that the levels of mTOR, p-mTOR (Ser2448), Raptor, PI3K (p110α), Beclin 1, ATG5 and Caspase-9 were up-regulated after treatment with psoralidin, psoralen and bavachin, and the expression of Bcl-2 was down-regulated after bavachinin treatment. CONCLUSIONS The hepatotoxicity of XLGB is associated with mitochondrial dysfunction. Five compounds in Psoraleae Fructus showed mitochondrial damage, they are psoralidin, isobavachalcone, bavachinin, bavachin and psoralen, especially psoralidin showed significant reduction in reserve capacity and respiratory control ratios. The molecular mechanism is related to the activation of PI3K/mTOR signaling pathway to inhibit autophagy and induce mitochondrial apoptosis.
Collapse
Affiliation(s)
- Shujuan Piao
- grid.73113.370000 0004 0369 1660Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003 China
| | - Hongwei Lin
- grid.73113.370000 0004 0369 1660Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003 China
| | - Xia Tao
- grid.73113.370000 0004 0369 1660Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003 China
| | - Wansheng Chen
- grid.73113.370000 0004 0369 1660Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003 China ,grid.412540.60000 0001 2372 7462Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
35
|
Mao L, Yuan X, Su J, Ma Y, Li C, Chen H, Zhang F. Human Umbilical Vein Endothelial Cells Survive on the Ischemic TCA Cycle under Lethal Ischemic Conditions. J Proteome Res 2022; 21:2385-2396. [PMID: 36074008 PMCID: PMC9552233 DOI: 10.1021/acs.jproteome.2c00255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
It is generally believed that vascular endothelial cells
(VECs)
rely on glycolysis instead of the tricarboxylic acid (TCA) cycle under
both normoxic and hypoxic conditions. However, the metabolic pattern
of human umbilical vein endothelial cells (HUVECs) under extreme ischemia
(hypoxia and nutrient deprivation) needs to be elucidated. We initiated
a lethal ischemic model of HUVECs, performed proteomics and bioinformatics,
and verified the metabolic pattern shift of HUVECs. Ischemic HUVECs
displayed extensive aerobic respiration, including upregulation of
the TCA cycle and mitochondrial respiratory chain in mitochondria
and downregulation of glycolysis in cytoplasm. The TCA cycle was enhanced
while the cell viability was decreased through the citrate synthase
pathway when substrates of the TCA cycle (acetate and/or pyruvate)
were added and vice versa when inhibitors of the TCA cycle (palmitoyl-CoA
and/or avidin) were applied. The inconsistency of the TCA cycle level
and cell viability suggested that the extensive TCA cycle can keep
cells alive yet generate toxic substances that reduce cell viability.
The data revealed that HUVECs depend on “ischemic TCA cycle”
instead of glycolysis to keep cells alive under lethal ischemic conditions,
but consideration must be given to relieve cell injury.
Collapse
Affiliation(s)
- Lisha Mao
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Xiaoqi Yuan
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Junlei Su
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Yaping Ma
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Chaofan Li
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Hongying Chen
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Fugui Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
36
|
Vallabh NA, Armstrong J, Czanner G, McDonagh B, Choudhary A, Criddle DN, Willoughby CE. Evidence of impaired mitochondrial cellular bioenergetics in ocular fibroblasts derived from glaucoma patients. Free Radic Biol Med 2022; 189:102-110. [PMID: 35872337 DOI: 10.1016/j.freeradbiomed.2022.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 11/20/2022]
Abstract
Glaucoma is a progressive optic neuropathy characterized by the neurodegeneration of the retinal ganglion cells (RGCs) resulting in irreversible visual impairment and eventual blindness. RGCs are extremely susceptible to mitochondrial compromise due to their marked bioenergetic requirements and morphology. There is increasing interest in therapies targeting mitochondrial health as a method of preventing visual loss in managing glaucoma. The bioenergetic profile of Tenon's ocular fibroblasts from glaucoma patients and controls was investigated using the Seahorse XF24 analyser. Impaired mitochondrial cellular bioenergetics was detected in glaucomatous ocular fibroblasts including basal respiration, maximal respiration and spare capacity. Spare respiratory capacity levels reflect mitochondrial bio-energetic adaptability in response to pathophysiological stress. Basal oxidative stress was elevated in glaucomatous Tenon's ocular fibroblasts and hydrogen peroxide (H2O2) induced reactive oxygen species (ROS) simulated the glaucomatous condition in normal Tenon's ocular fibroblasts. This work supports the role of therapeutic interventions to target oxidative stress or provide mitochondrial energetic support in glaucoma.
Collapse
Affiliation(s)
- Neeru A Vallabh
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L69 3BX, United Kingdom; St. Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, L7 8XP, United Kingdom
| | - Jane Armstrong
- Institute of Systems, Molecular and Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, L69 7BE, United Kingdom
| | - Gabriela Czanner
- School of Computer Science and Mathematics, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, United Kingdom; Faculty of Informatics and Information Technology, Slovak University of Technology, 842 16, Bratislava, Slovakia
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Anshoo Choudhary
- St. Paul's Eye Unit, Royal Liverpool University Hospital, Liverpool, L7 8XP, United Kingdom
| | - David N Criddle
- Institute of Systems, Molecular and Integrative Biology, Biosciences Building, University of Liverpool, Liverpool, L69 7BE, United Kingdom
| | - Colin E Willoughby
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L69 3BX, United Kingdom; Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, BT52 1SA, United Kingdom.
| |
Collapse
|
37
|
Divakaruni AS, Jastroch M. A practical guide for the analysis, standardization and interpretation of oxygen consumption measurements. Nat Metab 2022; 4:978-994. [PMID: 35971004 PMCID: PMC9618452 DOI: 10.1038/s42255-022-00619-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/17/2022] [Indexed: 12/14/2022]
Abstract
Measurement of oxygen consumption is a powerful and uniquely informative experimental technique. It can help identify mitochondrial mechanisms of action following pharmacologic and genetic interventions, and characterize energy metabolism in physiology and disease. The conceptual and practical benefits of respirometry have made it a frontline technique to understand how mitochondrial function can interface with-and in some cases control-cell physiology. Nonetheless, an appreciation of the complexity and challenges involved with such measurements is required to avoid common experimental and analytical pitfalls. Here we provide a practical guide to oxygen consumption measurements covering the selection of experimental models and instrumentation, as well as recommendations for the collection, interpretation and normalization of data. These guidelines are provided with the intention of aiding experimental design and enhancing the overall reputability, transparency and reliability of oxygen consumption measurements.
Collapse
Affiliation(s)
- Ajit S Divakaruni
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| |
Collapse
|
38
|
Hong SG, Shin J, Aldokhayyil M, Brown MD, Park JY. Mitochondrial and Metabolic Adaptations to Exercise-Induced Fluid Shear Stress in Endothelial Cells. Exerc Sport Sci Rev 2022; 50:145-155. [PMID: 35152237 PMCID: PMC9203873 DOI: 10.1249/jes.0000000000000289] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent studies have greatly advanced our understanding of the central role of mitochondria on endothelial function. Here, we propose a hypothesis that unidirectional laminar (pulsatile) flow and disturbed laminar (oscillatory) flow may differentially modulate mitochondrial phenotypes in the context of their bioenergetic, signaling, and biosynthetic functions, providing novel insights into subcellular mechanisms underlying how exercise benefits the improvement of vascular health.
Collapse
Affiliation(s)
- Soon-Gook Hong
- Department of Kinesiology & Cardiovascular Research Center, Temple University, Philadelphia, PA
| | - Junchul Shin
- Department of Kinesiology & Cardiovascular Research Center, Temple University, Philadelphia, PA
| | | | | | - Joon-Young Park
- Department of Kinesiology & Cardiovascular Research Center, Temple University, Philadelphia, PA
| |
Collapse
|
39
|
Kovacevic B, Ionescu CM, Jones M, Wagle SR, Lewkowicz M, Đanić M, Mikov M, Mooranian A, Al-Salami H. The Effect of Deoxycholic Acid on Chitosan-Enabled Matrices for Tissue Scaffolding and Injectable Nanogels. Gels 2022; 8:gels8060358. [PMID: 35735702 PMCID: PMC9222767 DOI: 10.3390/gels8060358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023] Open
Abstract
The pathophysiology of a multitude of diseases is influenced by bioenergetic dysfunction. Healthy mitochondria are presented as essential for the regulation and function of multiple cell types, including the cells of relevance for this research: pancreatic beta cells, muscle cells, and liver cells. Hence, effects of hydrogels (particularly nanogels) on bioenergetics needs to be taken into account when designing optimum delivery matrices. Several polymers have been suggested for use in hydrogels and nanogels, with focus on chitosan due to its range of beneficial properties. Bile acids have emerged as beneficial excipients, including deoxycholic acid, which can increase membrane permeability of cells. Nanogels were manufactured containing various concentrations of chitosan and deoxycholic acid in addition to the staple sodium alginate. Nanogels then underwent an array of analysis including rheological studies and in vitro cell work assessing viability, hypoxia, and the bioenergetic profiles. Overall, deoxycholic acid showed enhanced gel strength although this resulted in slightly lower cell viability and impacted bioenergetic profiles. Results from this study showed the benefits of deoxycholic acid; however, this was found to be less suitable for cell delivery matrices and is perhaps more beneficial for drug-delivery systems.
Collapse
Affiliation(s)
- Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
| | - Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
| | - Michael Lewkowicz
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
| | - Maja Đanić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, 21101 Novi Sad, Serbia; (M.Đ.); (M.M.)
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, 21101 Novi Sad, Serbia; (M.Đ.); (M.M.)
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
- Correspondence: (A.M.); (H.A.-S.)
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia; (B.K.); (C.M.I.); (M.J.); (S.R.W.); (M.L.)
- Hearing Therapeutics Department, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Perth, WA 6009, Australia
- Correspondence: (A.M.); (H.A.-S.)
| |
Collapse
|
40
|
Mitochondrial health quality control: measurements and interpretation in the framework of predictive, preventive, and personalized medicine. EPMA J 2022; 13:177-193. [PMID: 35578648 PMCID: PMC9096339 DOI: 10.1007/s13167-022-00281-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022]
Abstract
Mitochondria are the “gatekeeper” in a wide range of cellular functions, signaling events, cell homeostasis, proliferation, and apoptosis. Consequently, mitochondrial injury is linked to systemic effects compromising multi-organ functionality. Although mitochondrial stress is common for many pathomechanisms, individual outcomes differ significantly comprising a spectrum of associated pathologies and their severity grade. Consequently, a highly ambitious task in the paradigm shift from reactive to predictive, preventive, and personalized medicine (PPPM/3PM) is to distinguish between individual disease predisposition and progression under circumstances, resulting in compromised mitochondrial health followed by mitigating measures tailored to the individualized patient profile. For the successful implementation of PPPM concepts, robust parameters are essential to quantify mitochondrial health sustainability. The current article analyses added value of Mitochondrial Health Index (MHI) and Bioenergetic Health Index (BHI) as potential systems to quantify mitochondrial health relevant for the disease development and its severity grade. Based on the pathomechanisms related to the compromised mitochondrial health and in the context of primary, secondary, and tertiary care, a broad spectrum of conditions can significantly benefit from robust quantification systems using MHI/BHI as a prototype to be further improved. Following health conditions can benefit from that: planned pregnancies (improved outcomes for mother and offspring health), suboptimal health conditions with reversible health damage, suboptimal life-style patterns and metabolic syndrome(s) predisposition, multi-factorial stress conditions, genotoxic environment, ischemic stroke of unclear aetiology, phenotypic predisposition to aggressive cancer subtypes, pathologies associated with premature aging and neuro/degeneration, acute infectious diseases such as COVID-19 pandemics, among others.
Collapse
|
41
|
Pyrroloquinoline quinone (PQQ) protects mitochondrial function of HEI-OC1 cells under premature senescence. NPJ AGING 2022; 8:3. [PMID: 35927260 PMCID: PMC9158787 DOI: 10.1038/s41514-022-00083-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 03/14/2022] [Indexed: 12/13/2022]
Abstract
The aim of this study was to investigate the effects of pyrroloquinoline quinone (PQQ), an oxidoreductase cofactor, on the H2O2-induced premature senescence model in HEI-OC1 auditory cells and to elucidate its mechanism of action in vitro. Cells were treated with PQQ for 1 day before H2O2 (100 μM) exposure. Mitochondrial respiratory capacity was damaged in this premature senescence model but was restored in cells pretreated with PQQ (0.1 nM or 1.0 nM). A decrease in mitochondrial potential, the promotion of mitochondrial fusion and the accelerated movement of mitochondria were all observed in PQQ-pretreated cells. The protein expression of sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) were significantly decreased under H2O2 exposure while they were increased with PQQ pretreatment, and PGC-1α acetylation was significantly decreased. In conclusion, PQQ has a protective effect on the premature senescence model of HEI-OC1 auditory cells and is associated with the SIRT1/PGC-1α signaling pathway, mitochondrial structure, and mitochondrial respiratory capacity.
Collapse
|
42
|
EBI2 is a negative modulator of brown adipose tissue energy expenditure in mice and human brown adipocytes. Commun Biol 2022; 5:280. [PMID: 35351968 PMCID: PMC8964700 DOI: 10.1038/s42003-022-03201-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/28/2022] [Indexed: 11/16/2022] Open
Abstract
Pharmacological activation of brown adipose tissue (BAT) is an attractive approach for increasing energy expenditure to counteract obesity. Given the side-effects of known activators of BAT, we studied inhibitors of BAT as a novel, alternative concept to regulate energy expenditure. We focused on G-protein-coupled receptors that are one of the major targets of clinically used drugs. Here, we identify GPR183, also known as EBI2, as the most highly expressed inhibitory G-protein-coupled receptor in BAT among the receptors examined. Activation of EBI2 using its endogenous ligand 7α,25-dihydroxycholesterol significantly decreases BAT-mediated energy expenditure in mice. In contrast, mice deficient for EBI2 show increased energy dissipation in response to cold. Interestingly, only thermogenic adipose tissue depots — BAT and subcutaneous white adipose tissue —respond to 7α,25-dihydroxycholesterol treatment/EBI2 activation but not gonadal white fat, which has the lowest thermogenic capacity. EBI2 activation in brown adipocytes significantly reduces norepinephrine-induced cAMP production, whereas pharmacological inhibition or genetic ablation of EBI2 results in an increased response. Importantly, EBI2 significantly inhibits norepinephrine-induced activation of human brown adipocytes. Our data identify the 7α,25-dihydroxycholesterol/EBI2 signaling pathway as a so far unknown BAT inhibitor. Understanding the inhibitory regulation of BAT might lead to novel pharmacological approaches to increase the activity of thermogenic adipose tissue and whole body energy expenditure in humans. Francesca Copperi et al. evaluate the role of the Gi-protein coupled receptor, EBI2, on regulation of thermogenic activity in murine and human adipocytes. They report that loss of Ebi2 in mice increases brown adipocyte energy expenditure in response to cold exposure, providing insight into ways to potentially modulate energy expenditure in humans.
Collapse
|
43
|
Kang M, Kim HW, Yu AR, Yang JS, Lee SH, Lee JW, Yoon HS, Lee BS, Park HW, Lee SK, Lee S, Whang J, Kim JS. Comparison of Macrophage Immune Responses and Metabolic Reprogramming in Smooth and Rough Variant Infections of Mycobacterium mucogenicum. Int J Mol Sci 2022; 23:ijms23052488. [PMID: 35269631 PMCID: PMC8910348 DOI: 10.3390/ijms23052488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
Mycobacterium mucogenicum (Mmuc), a rapidly growing nontuberculous mycobacterium (NTM), can infect humans (posttraumatic wound infections and catheter-related sepsis). Similar to other NTM species, Mmuc exhibits colony morphologies of rough (Mmuc-R) and smooth (Mmuc-S) types. Although there are several case reports on Mmuc infection, no experimental evidence supports that the R-type is more virulent. In addition, the immune response and metabolic reprogramming of Mmuc have not been studied on the basis of morphological characteristics. Thus, a standard ATCC Mmuc strain and two clinical strains were analyzed, and macrophages were generated from mouse bone marrow. Cytokines and cell death were measured by ELISA and FACS, respectively. Mitochondrial respiration and glycolytic changes were measured by XF seahorse. Higher numbers of intracellular bacteria were found in Mmuc-R-infected macrophages than in Mmuc-S-infected macrophages. Additionally, Mmuc-R induced higher levels of the cytokines TNF-α, IL-6, IL-12p40, and IL-10 and induced more BMDM necrotic death. Furthermore, our metabolic data showed marked glycolytic and respiratory differences between the control and each type of Mmuc infection, and changes in these parameters significantly promoted glucose metabolism, extracellular acidification, and oxygen consumption in BMDMs. In conclusion, at least in the strains we tested, Mmuc-R is more virulent, induces a stronger immune response, and shifts bioenergetic metabolism more extensively than the S-type. This study is the first to report differential immune responses and metabolic reprogramming after Mmuc infection and might provide a fundamental basis for additional studies on Mmuc pathogenesis.
Collapse
Affiliation(s)
- Minji Kang
- Department of Medical Science, Chungnam National University, Daejeon 35365, Korea;
| | - Ho Won Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; (H.W.K.); (A.-R.Y.); (J.W.L.); (H.S.Y.)
| | - A-Reum Yu
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; (H.W.K.); (A.-R.Y.); (J.W.L.); (H.S.Y.)
| | - Jeong Seong Yang
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong 28158, Korea; (J.S.Y.); (S.H.L.)
| | - Seung Heon Lee
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong 28158, Korea; (J.S.Y.); (S.H.L.)
| | - Ji Won Lee
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; (H.W.K.); (A.-R.Y.); (J.W.L.); (H.S.Y.)
| | - Hoe Sun Yoon
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; (H.W.K.); (A.-R.Y.); (J.W.L.); (H.S.Y.)
| | - Byung Soo Lee
- Department of Ophthalmology, Konyang University Hospital and College of Medicine, Daejeon 35365, Korea;
| | - Hwan-Woo Park
- Department of Cell Biology, Konyang University College of Medicine, Daejeon 35365, Korea;
| | - Sung Ki Lee
- Department of Obstetrics and Gynecology, Konyang University Hospital, Daejeon 35365, Korea;
| | - Seungwan Lee
- Department of Medical Science, Konyang University, 158 Gwanjeodong-ro, Daejeon 35365, Korea;
| | - Jake Whang
- Korea Mycobacterium Resource Center (KMRC), Department of Research and Development, The Korean Institute of Tuberculosis, Osong 28158, Korea; (J.S.Y.); (S.H.L.)
- Correspondence: (J.W.); (J.-S.K.); Tel.: +82-43-249-4974 (J.W.); +82-42-600-8648 (J.-S.K.)
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea; (H.W.K.); (A.-R.Y.); (J.W.L.); (H.S.Y.)
- Correspondence: (J.W.); (J.-S.K.); Tel.: +82-43-249-4974 (J.W.); +82-42-600-8648 (J.-S.K.)
| |
Collapse
|
44
|
Su XX, Chen YR, Wu JQ, Wu XZ, Li KT, Wang XN, Sun JW, Wang H, Ou TM. Design, synthesis, and evaluation of 9-(pyrimidin-2-yl)-9H-carbazole derivatives disrupting mitochondrial homeostasis in human lung adenocarcinoma. Eur J Med Chem 2022; 232:114200. [DOI: 10.1016/j.ejmech.2022.114200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/06/2022] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
|
45
|
Pandey A, Sarkar S, Yadav SK, Yadav SS, Srikrishna S, Siddiqui MH, Parmar D, Yadav S. Studies on Regulation of Global Protein Profile and Cellular Bioenergetics of Differentiating SH-SY5Y Cells. Mol Neurobiol 2022; 59:1799-1818. [PMID: 35025051 DOI: 10.1007/s12035-021-02667-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/25/2021] [Indexed: 01/07/2023]
Abstract
The SH-SY5Y cells differentiated by sequential exposure of retinoic acid (RA) and brain-derived neurotrophic growth factor (BDNF) are a well-employed cellular model for studying the mechanistic aspects of neural development and neurodegeneration. Earlier studies from our lab have identified dramatic upregulation (77 miRNAs) and downregulation (17 miRNAs) of miRNAs in SH-SY5Y cells differentiated with successive exposure of RA + BDNF and demonstrated the essential role of increased levels of P53 proteins in coping with the differentiation-induced changes in protein levels. In continuation to our earlier studies, we have performed unbiased LC-MS/MS global protein profiling of naïve and differentiated SH-SY5Y cells and analyzed the identified proteins in reference to miRNAs identified in our earlier studies to identify the cellular events regulated by both identified miRNAs and proteins. Analysis of LC-MS/MS data has shown a significant increase and decrease in levels of 215 and 163 proteins, respectively, in differentiated SH-SY5Y cells. Integrative analysis of miRNA identified in our previous studies and protein identified in the present study is carried out to discover novel miRNA-protein regulatory modules to elucidate miRNA-protein regulatory relationships of differentiating neurons. In silico network analysis of miRNAs and proteins deregulated upon SH-SY5Y differentiation identified cell cycle, synapse formation, axonogenesis, differentiation, neuron projection, and neurotransmission, as the topmost involved pathways. Further, measuring mitochondrial dynamics and cellular bioenergetics using qPCR and Seahorse XFp Flux Analyzer, respectively, showed that differentiated cells possess increased mitochondrial dynamics and OCR relative to undifferentiated cells. In summary, our studies have identified a novel set of proteins deregulated during neuronal differentiation and establish the role of miRNAs identified in earlier studies in the regulation of proteins identified by LC-MS/MS-based global profiling of differentiating neurons, which will help in future studies related to neural development and neurodegeneration.
Collapse
Affiliation(s)
- Anuj Pandey
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India.,Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sana Sarkar
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India.,Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India
| | - Sanjeev Kumar Yadav
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India
| | - Smriti Singh Yadav
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India
| | - Saripella Srikrishna
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | | | - Devendra Parmar
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India.
| | - Sanjay Yadav
- Systems Toxicology and Health Risk Assessment Group, , CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Uttar Pradesh, Vishvigyan Bhawan, Lucknow, India. .,All India Institute of Medical Sciences (AIIMS), Uttar Pradesh, Raebareli, India.
| |
Collapse
|
46
|
Mooranian A, Ionescu CM, Walker D, Jones M, Wagle SR, Kovacevic B, Chester J, Foster T, Johnston E, Kuthubutheen J, Brown D, Atlas MD, Mikov M, Al-Salami H. Single-Cellular Biological Effects of Cholesterol-Catabolic Bile Acid-Based Nano/Micro Capsules as Anti-Inflammatory Cell Protective Systems. Biomolecules 2022; 12:biom12010073. [PMID: 35053221 PMCID: PMC8773943 DOI: 10.3390/biom12010073] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/27/2023] Open
Abstract
Recent studies in our laboratories have shown promising effects of bile acids in ➀ drug encapsulation for oral targeted delivery (via capsule stabilization) particularly when encapsulated with Eudragit NM30D® and ➁ viable-cell encapsulation and delivery (via supporting cell viability and biological activities, postencapsulation). Accordingly, this study aimed to investigate applications of bile acid-Eudragit NM30D® capsules in viable-cell encapsulation ready for delivery. Mouse-cloned pancreatic β-cell line was cultured and cells encapsulated using bile acid-Eudragit NM30D® capsules, and capsules' images, viability, inflammation, and bioenergetics of encapsulated cells assessed. The capsules' thermal and chemical stability assays were also assessed to ascertain an association between capsules' stability and cellular biological activities. Bile acid-Eudragit NM30D® capsules showed improved cell viability (e.g., F1 < F2 & F8; p < 0.05), insulin, inflammatory profile, and bioenergetics as well as thermal and chemical stability, compared with control. These effects were formulation-dependent and suggest, overall, that changes in ratios of bile acids to Eudragit NM30D® can change the microenvironment of the capsules and subsequent cellular biological activities.
Collapse
Affiliation(s)
- Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Daniel Walker
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Jacqueline Chester
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Edan Johnston
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | | | - Daniel Brown
- Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia;
| | - Marcus D. Atlas
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
| | - Momir Mikov
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, 21101 Novi Sad, Serbia;
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia; (A.M.); (C.M.I.); (D.W.); (M.J.); (S.R.W.); (B.K.); (J.C.); (T.F.); (E.J.)
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands, Perth, WA 6009, Australia;
- Correspondence: ; Tel.: +61-8-9266-9816; Fax: +61-8-9266-2769
| |
Collapse
|
47
|
Abstract
The adult mammalian heart is recalcitrant to regeneration after injury, in part due to the postmitotic nature of cardiomyocytes. Accumulating evidence suggests that cardiomyocyte proliferation in fetal or neonatal mammals and in regenerative non-mammalian models depends on a conducive metabolic state. Results from numerous studies in adult hearts indicate that conditions of relatively low fatty acid oxidation, low reactive oxygen species generation, and high glycolysis are required for induction of cardiomyocyte proliferation. Glycolysis appears particularly important because it provides branchpoint metabolites for several biosynthetic pathways that are essential for synthesis of nucleotides and nucleotide sugars, amino acids, and glycerophospholipids, all of which are required for daughter cell formation. In addition, the proliferative cardiomyocyte phenotype is supported in part by relatively low oxygen tensions and through the actions of critical transcription factors, coactivators, and signaling pathways that promote a more glycolytic and proliferative cardiomyocyte phenotype, such as hypoxia inducible factor 1α (Hif1α), Yes-associated protein (Yap), and ErbB2. Interventions that inhibit glycolysis or its integrated biosynthetic pathways almost universally impair cardiomyocyte proliferative capacity. Furthermore, metabolic enzymes that augment biosynthetic capacity such as phosphoenolpyruvate carboxykinase 2 and pyruvate kinase M2 appear to be amplifiers of cardiomyocyte proliferation. Collectively, these studies suggest that acquisition of a glycolytic and biosynthetic metabolic phenotype is a sine qua non of cardiomyocyte proliferation. Further knowledge of the regulatory mechanisms that control substrate partitioning to coordinate biosynthesis with energy provision could be leveraged to prompt or augment cardiomyocyte division and to promote cardiac repair.
Collapse
Affiliation(s)
- Tamer M A Mohamed
- Division of Cardiovascular Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Corresponding authors: Tamer M.A. Mohamed, PhD, Department of Medicine, Division of Cardiovascular Medicine, Institute of Molecular Cardiology, 580 S. Preston Street, Rm 121A, Louisville, KY 40202, USA.
| | - Riham Abouleisa
- Division of Cardiovascular Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - Bradford G Hill
- Division of Environmental Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
- Bradford G. Hill, PhD, Department of Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, 580 S. Preston Street, Rm 321E, Louisville, KY 40202, USA.
| |
Collapse
|
48
|
Tsujioka T, Sasaki D, Takeda A, Harashima H, Yamada Y. Resveratrol-Encapsulated Mitochondria-Targeting Liposome Enhances Mitochondrial Respiratory Capacity in Myocardial Cells. Int J Mol Sci 2021; 23:112. [PMID: 35008537 PMCID: PMC8745115 DOI: 10.3390/ijms23010112] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
The development of drug delivery systems for use in the treatment of cardiovascular diseases is an area of great interest. We report herein on an evaluation of the therapeutic potential of a myocardial mitochondria-targeting liposome, a multifunctional envelope-type nano device for targeting pancreatic β cells (β-MEND) that was previously developed in our laboratory. Resveratrol (RES), a natural polyphenol compound that has a cardioprotective effect, was encapsulated in the β-MEND (β-MEND (RES)), and its efficacy was evaluated using rat myocardioblasts (H9c2 cells). The β-MEND (RES) was readily taken up by H9c2 cells, as verified by fluorescence-activated cell sorter data, and was observed to be colocalized with intracellular mitochondria by confocal laser scanning microscopy. Myocardial mitochondrial function was evaluated by a Seahorse XF Analyzer and the results showed that the β-MEND (RES) significantly activated cellular maximal respiratory capacity. In addition, the β-MEND (RES) showed no cellular toxicity for H9c2 cells as evidenced by Premix WST-1 assays. This is the first report of the use of a myocardial mitochondria-targeting liposome encapsulating RES for activating mitochondrial function, which was clearly confirmed based on analyses using a Seahorse XF Analyzer.
Collapse
Affiliation(s)
- Takao Tsujioka
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan; (T.T.); (D.S.); (A.T.)
| | - Daisuke Sasaki
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan; (T.T.); (D.S.); (A.T.)
| | - Atsuhito Takeda
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan; (T.T.); (D.S.); (A.T.)
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan;
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan;
| |
Collapse
|
49
|
Le HHT, Liu CW, Denaro P, Jousma J, Shao NY, Rahman I, Lee WH. Genome-wide differential expression profiling of lncRNAs and mRNAs in human induced pluripotent stem cell-derived endothelial cells exposed to e-cigarette extract. Stem Cell Res Ther 2021; 12:593. [PMID: 34863290 PMCID: PMC8643021 DOI: 10.1186/s13287-021-02654-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/31/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Electronic-cigarette (e-cig) usage, particularly in the youth population, is a growing concern. It is known that e-cig causes endothelial dysfunction, which is a risk factor for the development of cardiovascular diseases; however, the mechanisms involved remain unclear. We hypothesized that long noncoding RNAs (lncRNAs) may play a role in e-cig-induced endothelial dysfunction. METHODS Here, we identified lncRNAs that are dysregulated in human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) following 24 h of e-cig aerosol extract treatment via microarray analysis. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genome (KEGG) pathway analyses of the dysregulated mRNAs following e-cig exposure and constructed co-expression networks of the top 5 upregulated lncRNAs and the top 5 downregulated lncRNAs and the mRNAs that are correlated with them. Furthermore, the functional effects of knocking down lncRNA lung cancer-associated transcript 1 (LUCAT1) on EC phenotypes were determined as it was one of the significantly upregulated lncRNAs following e-cig exposure based on our profiling. RESULTS 183 lncRNAs and 132 mRNAs were found to be upregulated, whereas 297 lncRNAs and 413 mRNAs were found to be downregulated after e-cig exposure. We also observed that e-cig caused dysregulation of endothelial metabolism resulting in increased FAO activity, higher mitochondrial membrane potential, and decreased glucose uptake and glycolysis. These results suggest that e-cig alters EC metabolism by increasing FAO to compensate for energy deficiency in ECs. Finally, the knockdown of LUCAT1 prevented e-cig-induced EC dysfunction by maintaining vascular barrier, reducing reactive oxygen species level, and increasing migration capacity. CONCLUSION This study identifies an expression profile of differentially expressed lncRNAs and several potential regulators and pathways in ECs exposed to e-cig, which provide insights into the regulation of lncRNAs and mRNAs and the role of lncRNA and mRNA networks in ECs associated e-cig exposure.
Collapse
Affiliation(s)
- Hoai Huong Thi Le
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N 5th Street, Building ABC1, Rm 426, Phoenix, AZ, 85004-2157, USA
| | - Chen-Wei Liu
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N 5th Street, Building ABC1, Rm 426, Phoenix, AZ, 85004-2157, USA
| | - Philip Denaro
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N 5th Street, Building ABC1, Rm 426, Phoenix, AZ, 85004-2157, USA
| | - Jordan Jousma
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Ning-Yi Shao
- Health Sciences, University of Macau, Macau, China
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 N 5th Street, Building ABC1, Rm 426, Phoenix, AZ, 85004-2157, USA.
| |
Collapse
|
50
|
McDowell RE, Ali KF, Lad S, San Martin VT, Bottino R, Walsh M, Stevens T, Wilke W, Kirwan JP, Hatipoglu B. Bioenergetics of Islet Preparations in a Pilot Clinical Trial of Peri-Transplant Hydroxychloroquine for Autologous Islet Transplantation. Cell Transplant 2021; 30:9636897211057440. [PMID: 34757864 PMCID: PMC8586172 DOI: 10.1177/09636897211057440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The inflammatory response is an obstacle to success in both allogeneic and autologous islet transplantation. In autologous islet transplantation (AIT), however, the recipient is also the donor, permitting pretreatment of donor/recipient for a controlled duration prior to transplantation. We sought to exploit this feature of (AIT) by pretreating donor/recipients with chronic pancreatitis undergoing total pancreatectomy and autologous islet transplantation (TPAIT) to test the hypothesis that peri-transplant treatment with the FDA-approved anti-inflammatory hydroxychloroquine (HCQ) improves graft function. In this randomized placebo-controlled pilot clinical study, patients (n = 6) were treated with oral HCQ for 30 days prior to and 90 days after TPAIT. In vivo islet function was assessed via Mixed Meal Tolerance Testing before HCQ treatment, 6- and 12-months after surgery. In vitro islet bioenergetics were assessed at the time of transplantation via extracellular flux analysis of islet preparation samples from the clinical trial cohort and six additional patients (n = 12). Our study shows that HCQ did not alter clinical endpoints, but HCQ-treated patients showed greater spare respiratory capacity (SRC) compared to samples from control patients (P=0.028). Glycolytic metabolism of islet preparations directly correlated with stimulated C-peptide secretion both before and after TPAIT (P=0.01, R2=0.489 and P=0.03, R2=0.674, respectively), and predicted in vivo islet function better than mitochondrial metabolism of islet preps or islet equivalents infused. Overnight culture of islet preparations altered bioenergetic function, significantly decreasing SRC and maximal respiration (P<0.001). In conclusion, while HCQ did not alter clinical outcomes, it was associated with significantly increased SRC in islet preparations. Bioenergetic analyses of islet preparations suggests that culture should be avoided and that glycolysis may be a more sensitive indicator of in vivo islet function than current metrics, including islet oxygen consumption and islet equivalents infused.
Collapse
Affiliation(s)
- Ruth E McDowell
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Biology, Oberlin College, Oberlin, OH, USA
| | - Khawla F Ali
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, USA.,Royal College of Surgeons in Ireland-Medical University of Bahrain, Muharraq, Bahrain
| | - Saloni Lad
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | | | - Rita Bottino
- Institute for Cellular Therapeutics, Allegheny Health Network Research Institute, Pittsburgh, PA, USA.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Matthew Walsh
- Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tyler Stevens
- Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
| | - William Wilke
- Orthopaedic & Rheumatologic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John P Kirwan
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Betul Hatipoglu
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, USA.,Diabetes & Obesity Center, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|