1
|
Qian M, Jiang Z, Xu C, Wang L, Hu N. Changes in the gut microbiota and derived fecal metabolites may play a role in tacrolimus-induced diabetes in mice. Future Microbiol 2024:1-10. [PMID: 39711145 DOI: 10.1080/17460913.2024.2444761] [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: 06/30/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024] Open
Abstract
AIMS A notable scarcity of research has focused on examining alterations in gut microbiota and its metabolites within tacrolimus (TAC)-induced diabetes models. METHODS Tacrolimus-induced changes in glucose and lipid metabolism indices were analyzed through different routes of administration. The potential role of gut microbiota and its metabolites in TAC-induced diabetes was investigated using 16S rRNA sequencing and non-targeted metabolomics. RESULTS After intraperitoneal(ip) and oral(po) administration of TAC, the α-diversity index of gut microbiota was significantly increased. The gut microbiota of the three groups of mice was significantly separated, and there were significant changes in composition and functional genes. Fecal metabolites changed significantly after TAC administration by different routes, and 53 metabolites (38 down-regulated and 15 up-regulated) were identified (CON vs. TACip). Similarly, 29 metabolites (8 down-regulated and 21 up-regulated) were identified (CON vs. TACpo). KEGG pathway analysis identified 4 and 13 significantly altered metabolic pathways, respectively. Correlation analysis suggested that microbiota and metabolites were involved in the pathogenesis of TAC-induced diabetes. CONCLUSION This study investigated the alterations in gut microbiota and fecal metabolites in TAC-induced diabetic mice and evaluated the correlation between these changes. These findings provide valuable insights into potential biomarkers in the development of TAC-induced diabetes.
Collapse
Affiliation(s)
- Minyan Qian
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zhenwei Jiang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Caomei Xu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Liying Wang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Nan Hu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, Jiangsu, China
| |
Collapse
|
2
|
Fiordelisi A, Cerasuolo FA, Avvisato R, Buonaiuto A, Maisto M, Bianco A, D'Argenio V, Mone P, Perrino C, D'Apice S, Paolillo R, Pezone A, Varzideh F, Santulli G, Sorriento D, Iaccarino G, Gambardella J. L-Arginine supplementation as mitochondrial therapy in diabetic cardiomyopathy. Cardiovasc Diabetol 2024; 23:450. [PMID: 39707340 DOI: 10.1186/s12933-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 10/23/2024] [Indexed: 12/23/2024] Open
Abstract
In patients with type II diabetes, the development of diabetic cardiomyopathy (DC) is associated with a high risk of mortality. Left ventricular hypertrophy, diastolic dysfunction, and exercise intolerance are the first signs of DC. The underlying mechanisms are not fully elucidated, and there is an urgent need for specific biomarkers and molecular targets for early diagnosis and treatment. Mitochondrial alterations play a key role in the development of DC, and microRNAs regulating mitochondrial function are emerging as potential biomarkers of metabolic stress in DC. L-Arginine (Arg) supplementation has been shown to be an effective strategy for improving mitochondrial function and energetics, with a significant impact on physical performance. The aim of the current study was to evaluate the effects of Arg supplementation on cardiac mitochondrial function, DC development, and relative phenotypes including exercise intolerance. We used db/db mice as a model of type II diabetes, chronically treated with Arg (1 mg/kg/day) for 12 weeks. Arg-treated db/db mice showed preserved diastolic function and left ventricular morphology compared with untreated diabetic mice. Arg supplementation also improved exercise tolerance and the propensity to physical activity. Mitochondrial respiration was significantly increased in cardiomyocytes isolated from treated db/db mice, as well as in diabetic cardiomyocytes treated with Arg in vitro. The improvement of cardiac mitochondrial function in db/db + Arg mice was associated with an increase in PGC-1-alpha levels, mitochondrial biogenesis, recycling, and antioxidant capacity. Arg treatment prevented the accumulation of circulating and cardiac miR-143 in db/db mice, which is an index of metabolic stress and activation of mitochondrial damage mechanisms. In conclusion, Arg supplementation is effective in preventing the development of DC, preserving diastolic function and exercise tolerance by improving mitochondrial fitness and homeostasis. Additionally, miR-143 could potentially be employed to monitor cardiac metabolic stress and the effects of Arg treatment in diabetes.
Collapse
MESH Headings
- Animals
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/prevention & control
- Arginine/metabolism
- Dietary Supplements
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Male
- Ventricular Function, Left/drug effects
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/metabolism
- Exercise Tolerance/drug effects
- Disease Models, Animal
- Mice, Inbred C57BL
- Energy Metabolism/drug effects
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Mice
- Cells, Cultured
Collapse
Affiliation(s)
- Antonella Fiordelisi
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Federica Andrea Cerasuolo
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, Florence, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonietta Buonaiuto
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, Florence, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Antonio Bianco
- Department of Public Health, Federico II University, Naples, Italy
| | - Valeria D'Argenio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
- CEINGE- Advanced Biotechnologies, Naples, Italy
| | - Pasquale Mone
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
- Casa di Cura Privata Montevergine, Mercogliano, Italy
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Stefania D'Apice
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonio Pezone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
| | - Fahimeh Varzideh
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Biology, Federico II University, Naples, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, NY, USA
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Guido Iaccarino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Jessica Gambardella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
- International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy.
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy.
| |
Collapse
|
3
|
Yue L, Li J, Yao M, Song S, Zhang X, Wang Y. Cutting edge of immune response and immunosuppressants in allogeneic and xenogeneic islet transplantation. Front Immunol 2024; 15:1455691. [PMID: 39346923 PMCID: PMC11427288 DOI: 10.3389/fimmu.2024.1455691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
As an effective treatment for diabetes, islet transplantation has garnered significant attention and research in recent years. However, immune rejection and the toxicity of immunosuppressive drugs remain critical factors influencing the success of islet transplantation. While immunosuppressants are essential in reducing immune rejection reactions and can significantly improve the survival rate of islet transplants, improper use of these drugs can markedly increase mortality rates following transplantation. Additionally, the current availability of islet organ donations fails to meet the demand for organ transplants, making xenotransplantation a crucial method for addressing organ shortages. This review will cover the following three aspects: 1) the immune responses occurring during allogeneic islet transplantation, including three stages: inflammation and IBMIR, allogeneic immune response, and autoimmune recurrence; 2) commonly used immunosuppressants in allogeneic islet transplantation, including calcineurin inhibitors (Cyclosporine A, Tacrolimus), mycophenolate mofetil, glucocorticoids, and Bortezomib; and 3) early and late immune responses in xenogeneic islet transplantation and the immune effects of triple therapy (ECDI-fixed donor spleen cells (ECDI-SP) + anti-CD20 + Sirolimus) on xenotransplantation.
Collapse
Affiliation(s)
- Liting Yue
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jisong Li
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Yao
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Xiaoqin Zhang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Wang
- Center of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
4
|
Arcas JM, Oudaha K, González A, Fernández-Trillo J, Peralta FA, Castro-Marsal J, Poyraz S, Taberner F, Sala S, de la Peña E, Gomis A, Viana F. The ion channel TRPM8 is a direct target of the immunosuppressant rapamycin in primary sensory neurons. Br J Pharmacol 2024; 181:3192-3214. [PMID: 38741464 DOI: 10.1111/bph.16402] [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: 09/14/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.
Collapse
Affiliation(s)
- José Miguel Arcas
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Khalid Oudaha
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Alejandro González
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Jorge Fernández-Trillo
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | | - Júlia Castro-Marsal
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Seyma Poyraz
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Francisco Taberner
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Elvira de la Peña
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Ana Gomis
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Félix Viana
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| |
Collapse
|
5
|
Yen NTH, Phat NK, Oh JH, Park SM, Moon KS, Thu VTA, Cho YS, Shin JG, Long NP, Kim DH. Pathway-level multi-omics analysis of the molecular mechanisms underlying the toxicity of long-term tacrolimus exposure. Toxicol Appl Pharmacol 2023; 473:116597. [PMID: 37321324 DOI: 10.1016/j.taap.2023.116597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
Tacrolimus (TAC)-based treatment is associated with nephrotoxicity and hepatotoxicity; however, the underlying molecular mechanisms responsible for this toxicity have not been fully explored. This study elucidated the molecular processes underlying the toxic effects of TAC using an integrative omics approach. Rats were sacrificed after 4 weeks of daily oral TAC administration at a dose of 5 mg/kg. The liver and kidney underwent genome-wide gene expression profiling and untargeted metabolomics assays. Molecular alterations were identified using individual data profiling modalities and further characterized by pathway-level transcriptomics-metabolomics integration analysis. Metabolic disturbances were mainly related to an imbalance in oxidant-antioxidant status, as well as in lipid and amino acid metabolism in the liver and kidney. Gene expression profiles also indicated profound molecular alterations, including in genes associated with a dysregulated immune response, proinflammatory signals, and programmed cell death in the liver and kidney. Joint-pathway analysis indicated that the toxicity of TAC was associated with DNA synthesis disruption, oxidative stress, and cell membrane permeabilization, as well as lipid and glucose metabolism. In conclusion, our pathway-level integration of transcriptome and metabolome and conventional analyses of individual omics profiles, provided a more comprehensive picture of the molecular changes resulting from TAC toxicity. This study also serves as a valuable resource for subsequent investigations aiming to understand the mechanism underlying the molecular toxicology of TAC.
Collapse
Affiliation(s)
- Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Ky Phat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Jung-Hwa Oh
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Se-Myo Park
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kyoung-Sik Moon
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Vo Thuy Anh Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Yong-Soon Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| |
Collapse
|
6
|
Shin JH, Bozadjieva-Kramer N, Seeley RJ. Reg3γ: current understanding and future therapeutic opportunities in metabolic disease. Exp Mol Med 2023; 55:1672-1677. [PMID: 37524871 PMCID: PMC10474034 DOI: 10.1038/s12276-023-01054-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/01/2023] [Indexed: 08/02/2023] Open
Abstract
Regenerating family member gamma, Reg3γ (the mouse homolog of human REG3A), belonging to the antimicrobial peptides (AMPs), functions as a part of the host immune system to maintain spatial segregation between the gut bacteria and the host in the intestine via bactericidal activity. There is emerging evidence that gut manipulations such as bariatric surgery, dietary supplementation or drug treatment to produce metabolic benefits alter the gut microbiome. In addition to changes in a wide range of gut hormones, these gut manipulations also induce the expression of Reg3γ in the intestine. Studies over the past decades have revealed that Reg3γ not only plays a role in the gut lumen but can also contribute to host physiology through interaction with the gut microbiota. Herein, we discuss the current knowledge regarding the biology of Reg3γ, its role in various metabolic functions, and new opportunities for therapeutic strategies to treat metabolic disorders.
Collapse
Affiliation(s)
- Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Vidigal AC, de Lucena DD, Beyerstedt S, Rangel ÉB. A comprehensive update of the metabolic and toxicological considerations for immunosuppressive drugs used during pancreas transplantation. Expert Opin Drug Metab Toxicol 2023; 19:405-427. [PMID: 37542452 DOI: 10.1080/17425255.2023.2243808] [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: 03/26/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Despite significant advancements in immunosuppressive regimens and surgical techniques, the prevalence of adverse events related to immunosuppression remains a major challenge affecting the long-term survival rates of pancreas and kidney allografts. AREAS COVERED This article presents a comprehensive review of the literature and knowledge (Jan/2012-Feb/2023) concerning glucose metabolism disorders and nephrotoxicity associated with tacrolimus and mammalian target of rapamycin inhibitors (mTORi). Novel signaling pathways potentially implicated in these adverse events are discussed. Furthermore, we extensively examine the findings from clinical trials evaluating the efficacy and safety of tacrolimus, mTORi, and steroid minimization. EXPERT OPINION Tacrolimus-based regimens continue to be the standard treatment following pancreas transplants. However, prolonged use of tacrolimus and mTORi may lead to hyperglycemia and nephrotoxicity. Understanding and interpreting experimental data, particularly concerning novel signaling pathways beyond calcineurin-NFAT and mTOR pathways, can offer valuable insights for therapeutic interventions to mitigate hyperglycemia and nephrotoxicity. Additionally, critically analyzing clinical trial results can identify opportunities for personalized safety-based approaches to minimize side effects. It is imperative to conduct randomized-controlled studies to assess the impact of mTORi use and steroid-free protocols on pancreatic allograft survival. Such studies will aid in tailoring treatment strategies for improved transplant outcomes.
Collapse
Affiliation(s)
- Ana Cláudia Vidigal
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Débora D de Lucena
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
| | - Stephany Beyerstedt
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| | - Érika B Rangel
- Nephrology Division, Department of Medicine, Federal University of São Paulo, SP, Brazil
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, SP, São Paulo, Brazil
| |
Collapse
|
8
|
Luo M, Santulli G. Editorial: The link between obesity, type 2 diabetes, and mitochondria. Front Endocrinol (Lausanne) 2023; 14:1229935. [PMID: 37409237 PMCID: PMC10319143 DOI: 10.3389/fendo.2023.1229935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023] Open
Affiliation(s)
- Moulun Luo
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY, United States
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
9
|
Gambardella J, Fiordelisi A, Cerasuolo FA, Buonaiuto A, Avvisato R, Viti A, Sommella E, Merciai F, Salviati E, Campiglia P, D’Argenio V, Parisi S, Bianco A, Spinelli L, Di Vaia E, Cuocolo A, Pisani A, Riccio E, Di Risi T, Ciccarelli M, Santulli G, Sorriento D, Iaccarino G. Experimental evidence and clinical implications of Warburg effect in the skeletal muscle of Fabry disease. iScience 2023; 26:106074. [PMID: 36879801 PMCID: PMC9984560 DOI: 10.1016/j.isci.2023.106074] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 01/24/2023] [Indexed: 01/30/2023] Open
Abstract
Skeletal muscle (SM) pain and fatigue are common in Fabry disease (FD). Here, we undertook the investigation of the energetic mechanisms related to FD-SM phenotype. A reduced tolerance to aerobic activity and lactate accumulation occurred in FD-mice and patients. Accordingly, in murine FD-SM we detected an increase in fast/glycolytic fibers, mirrored by glycolysis upregulation. In FD-patients, we confirmed a high glycolytic rate and the underutilization of lipids as fuel. In the quest for a tentative mechanism, we found HIF-1 upregulated in FD-mice and patients. This finding goes with miR-17 upregulation that is responsible for metabolic remodeling and HIF-1 accumulation. Accordingly, miR-17 antagomir inhibited HIF-1 accumulation, reverting the metabolic-remodeling in FD-cells. Our findings unveil a Warburg effect in FD, an anaerobic-glycolytic switch under normoxia induced by miR-17-mediated HIF-1 upregulation. Exercise-intolerance, blood-lactate increase, and the underlying miR-17/HIF-1 pathway may become useful therapeutic targets and diagnostic/monitoring tools in FD.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | | | - Antonietta Buonaiuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Alessandro Viti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | | | | | | | | | - Valeria D’Argenio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Rome, Italy
- CEINGE- Advanced Biotechnologies, Naples, Italy
| | - Silvia Parisi
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Naples, Italy
| | - Antonio Bianco
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Letizia Spinelli
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Eugenio Di Vaia
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonio Pisani
- Department of Public Health, Federico II University, Naples, Italy
| | - Eleonora Riccio
- Department of Public Health, Federico II University, Naples, Italy
| | | | - Michele Ciccarelli
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation, Albert Einstein College of Medicine, New York, NY, USA
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Research on Hypertension and Related Conditions (CIRIAPA), Federico II University, Naples, Italy
| |
Collapse
|
10
|
Kale A, Rogers NM. No Time to Die-How Islets Meet Their Demise in Transplantation. Cells 2023; 12:cells12050796. [PMID: 36899932 PMCID: PMC10000424 DOI: 10.3390/cells12050796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Islet transplantation represents an effective treatment for patients with type 1 diabetes mellitus (T1DM) and severe hypoglycaemia unawareness, capable of circumventing impaired counterregulatory pathways that no longer provide protection against low blood glucose levels. The additional beneficial effect of normalizing metabolic glycaemic control is the minimisation of further complications related to T1DM and insulin administration. However, patients require allogeneic islets from up to three donors, and the long-term insulin independence is inferior to that achieved with solid organ (whole pancreas) transplantation. This is likely due to the fragility of islets caused by the isolation process, innate immune responses following portal infusion, auto- and allo-immune-mediated destruction and β-cell exhaustion following transplantation. This review covers the specific challenges related to islet vulnerability and dysfunction that affect long-term cell survival following transplantation.
Collapse
Affiliation(s)
- Atharva Kale
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Renal and Transplant Unit, Westmead Hospital, Westmead, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
11
|
Mone P, Varzideh F, Jankauskas SS, Pansini A, Lombardi A, Frullone S, Santulli G. SGLT2 Inhibition via Empagliflozin Improves Endothelial Function and Reduces Mitochondrial Oxidative Stress: Insights From Frail Hypertensive and Diabetic Patients. Hypertension 2022; 79:1633-1643. [PMID: 35703100 PMCID: PMC9642044 DOI: 10.1161/hypertensionaha.122.19586] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Frailty is a multidimensional condition often diagnosed in older adults with hypertension and diabetes, and both these conditions are associated with endothelial dysfunction and oxidative stress. We investigated the functional role of the SGLT2 (sodium glucose cotransporter 2) inhibitor empagliflozin in frail diabetic and hypertensive older adults. METHODS We studied the effects of empagliflozin in consecutive hypertensive and diabetic older patients with frailty presenting at the ASL (local health unit of the Italian Ministry of Health) of Avellino, Italy, from March 2021 to January 2022. Moreover, we performed in vitro experiments in human endothelial cells to measure cell viability, permeability, mitochondrial Ca2+, and oxidative stress. RESULTS We evaluated 407 patients; 325 frail elders with diabetes successfully completed the study. We propensity-score matched 75 patients treated with empagliflozin and 75 with no empagliflozin. We observed a correlation between glycemia and Montreal Cognitive Assessment (MoCA) score and between glycemia and 5-meter gait speed (5mGS). At 3-month follow-up, we detected a significant improvement in the MoCA score and in the 5mGS in patients receiving empagliflozin compared with non-treated subjects. Mechanistically, we demonstrate that empagliflozin significantly reduces mitochondrial Ca2+ overload and reactive oxygen species production triggered by high glucose in human endothelial cells, attenuates cellular permeability, and improves cell viability in response to oxidative stress. CONCLUSIONS Taken together, our data indicate that empagliflozin reduces frailty in diabetic and hypertensive patients, most likely by decreasing the mitochondrial generation of reactive oxygen species in endothelial cells.
Collapse
Affiliation(s)
- Pasquale Mone
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
- Asl Avellino
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
| | | | | | - Angela Lombardi
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
| | | | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Albert Einstein College of Medicine, New York, NY
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY
| |
Collapse
|
12
|
Li J, Yan H, Xiang R, Yang W, Ye J, Yin R, Yang J, Chi Y. ATP Secretion and Metabolism in Regulating Pancreatic Beta Cell Functions and Hepatic Glycolipid Metabolism. Front Physiol 2022; 13:918042. [PMID: 35800345 PMCID: PMC9253475 DOI: 10.3389/fphys.2022.918042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetes (DM), especially type 2 diabetes (T2DM) has become one of the major diseases severely threatening public health worldwide. Islet beta cell dysfunctions and peripheral insulin resistance including liver and muscle metabolic disorder play decisive roles in the pathogenesis of T2DM. Particularly, increased hepatic gluconeogenesis due to insulin deficiency or resistance is the central event in the development of fasting hyperglycemia. To maintain or restore the functions of islet beta cells and suppress hepatic gluconeogenesis is crucial for delaying or even stopping the progression of T2DM and diabetic complications. As the key energy outcome of mitochondrial oxidative phosphorylation, adenosine triphosphate (ATP) plays vital roles in the process of almost all the biological activities including metabolic regulation. Cellular adenosine triphosphate participates intracellular energy transfer in all forms of life. Recently, it had also been revealed that ATP can be released by islet beta cells and hepatocytes, and the released ATP and its degraded products including ADP, AMP and adenosine act as important signaling molecules to regulate islet beta cell functions and hepatic glycolipid metabolism via the activation of P2 receptors (ATP receptors). In this review, the latest findings regarding the roles and mechanisms of intracellular and extracellular ATP in regulating islet functions and hepatic glycolipid metabolism would be briefly summarized and discussed.
Collapse
Affiliation(s)
- Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Han Yan
- Key Laboratory of Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Rui Xiang
- Key Laboratory of Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Weili Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jingjing Ye
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), National Center for Trauma Medicine, Trauma Medicine Center, Peking University People’s Hospital, Beijing, China
| | - Ruili Yin
- Beijing Key Laboratory of Diabetes Prevention and Research, Center for Endocrine Metabolic and Immune Disease, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jichun Yang
- Key Laboratory of Cardiovascular Science of the Ministry of Education, Center for Non-coding RNA Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- *Correspondence: Jichun Yang, ; Yujing Chi,
| | - Yujing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, China
- *Correspondence: Jichun Yang, ; Yujing Chi,
| |
Collapse
|
13
|
Li S, Zhou H, Xie M, Zhang Z, Gou J, Yang J, Tian C, Ma K, Wang CY, Lu Y, Li Q, Peng W, Xiang M. Regenerating islet-derived protein 3 gamma (Reg3g) ameliorates tacrolimus-induced pancreatic β-cell dysfunction in mice by restoring mitochondrial function. Br J Pharmacol 2022; 179:3078-3095. [PMID: 35060126 DOI: 10.1111/bph.15803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Tacrolimus a first-line medication used after transplantation can induce β-cell dysfunction, causing new-onset diabetes mellitus (NODM). Regenerating islet-derived protein 3 gamma (Reg3g), a member of the pancreatic regenerative gene family, has been reported to improve type 1 diabetes by promoting β-cell regeneration. We aim to investigate the role of Reg3g in reversing tacrolimus-induced β-cell dysfunction and NODM in mice. EXPERIMENTAL APPROACH Circulating REG3A (the human homologue of mouse Reg3g) in heart transplantation patients treated with tacrolimus was detected. The glucose-stimulated insulin secretion and mitochondrial functions, including mitochondria membrane potential (MMP), mitochondria calcium, ATP production, oxygen consumption rate and mitochondrial morphology were investigated in β-cells. Additionally, effects of Reg3g on tacrolimus-induced NODM in mice were analysed. KEY RESULTS Circulating REG3A level in heart transplantation patients with NODM significantly decreased compared with those without diabetes. Tacrolimus down-regulated Reg3g via inhibiting STAT3-mediated transcription activation. Moreover, Reg3g restored glucose-stimulated insulin secretion suppressed by tacrolimus in β-cells by improving mitochondrial functions, including increased MMP, mitochondria calcium uptake, ATP production, oxygen consumption rate and contributing to an intact mitochondrial morphology. Mechanistically, Reg3g increased accumulation of pSTAT3(Ser727) in mitochondria by activating ERK1/2-STAT3 signalling pathway, leading to restoration of tacrolimus-induced mitochondrial impairment. Reg3g overexpression also effectively mitigated tacrolimus-induced NODM in mice. CONCLUSION AND IMPLICATIONS Reg3g can significantly ameliorate tacrolimus-induced β-cell dysfunction by restoring mitochondrial function in a pSTAT3(Ser727)-dependent manner. Our observations identify a novel Reg3g-mediated mechanism that is involved in tacrolimus-induced NODM and establish the novel role of Reg3g in reversing tacrolimus-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyuan Xie
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zijun Zhang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Gou
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Ma
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yi Lu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Peng
- Department of General Practice, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Gambardella J, Jankauskas SS, D'Ascia SL, Sardu C, Matarese A, Minicucci F, Mone P, Santulli G. Glycation of ryanodine receptor in circulating lymphocytes predicts the response to cardiac resynchronization therapy. J Heart Lung Transplant 2022; 41:438-441. [PMID: 35042640 PMCID: PMC8977242 DOI: 10.1016/j.healun.2021.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 01/02/2023] Open
Abstract
Finding reliable parameters to identify patients with heart failure (HF) that will respond to cardiac resynchronization therapy (CRT) represents a major challenge. We and others have observed post-translational modifications of Ryanodine Receptor (RyR) in several tissues (including skeletal muscle and circulating lymphocytes) of patients with advanced HF. We designed a prospective study to test the hypothesis that RyR1 glycation in circulating lymphocytes could predict CRT responsiveness in patients with non-ischemic HF. We enrolled 94 patients who underwent CRT and 30 individuals without HF, examining RyR1 glycation in peripheral lymphocytes at enrollment and after 1 year. We found that baseline RyR1 glycation independently predicts CRT response at 1 year after adjusting for age, diabetes, QRS duration and morphology, echocardiographic dyssynchrony, and hypertension. Moreover, RyR1 glycation in circulating lymphocytes significantly correlated with pathologic intracellular calcium leak. Taken together, our data show for the first time that RyR1 glycation in circulating lymphocytes represents a novel biomarker to predict CRT responsiveness.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, University of Naples "Federico II" and International Translational Research and Medical Education (ITME) Consortium, Naples, Italy
| | - Stanislovas S Jankauskas
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York
| | | | | | | | - Fabio Minicucci
- Naples Local Health Unit (ASL) of the Italian Ministry of Health, Naples, Italy
| | - Pasquale Mone
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York; Department of Advanced Biomedical Sciences, University of Naples "Federico II" and International Translational Research and Medical Education (ITME) Consortium, Naples, Italy; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation (INI), Norman Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York City, New York.
| |
Collapse
|
15
|
San Martín A, Arce-Molina R, Aburto C, Baeza-Lehnert F, Barros LF, Contreras-Baeza Y, Pinilla A, Ruminot I, Rauseo D, Sandoval PY. Visualizing physiological parameters in cells and tissues using genetically encoded indicators for metabolites. Free Radic Biol Med 2022; 182:34-58. [PMID: 35183660 DOI: 10.1016/j.freeradbiomed.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023]
Abstract
The study of metabolism is undergoing a renaissance. Since the year 2002, over 50 genetically-encoded fluorescent indicators (GEFIs) have been introduced, capable of monitoring metabolites with high spatial/temporal resolution using fluorescence microscopy. Indicators are fusion proteins that change their fluorescence upon binding a specific metabolite. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides. They permit monitoring relative levels, concentrations, and fluxes in living systems. At a minimum they report relative levels and, in some cases, absolute concentrations may be obtained by performing ad hoc calibration protocols. Proper data collection, processing, and interpretation are critical to take full advantage of these new tools. This review offers a survey of the metabolic indicators that have been validated in mammalian systems. Minimally invasive, these indicators have been instrumental for the purposes of confirmation, rebuttal and discovery. We envision that this powerful technology will foster metabolic physiology.
Collapse
Affiliation(s)
- A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile.
| | - R Arce-Molina
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - C Aburto
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | | | - L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - Y Contreras-Baeza
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - A Pinilla
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | - D Rauseo
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Universidad Austral de Chile, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| |
Collapse
|
16
|
Effects of Chronic Supplementation of L-Arginine on Physical Fitness in Water Polo Players. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6684568. [PMID: 33815657 PMCID: PMC7994081 DOI: 10.1155/2021/6684568] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/04/2021] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Background Ergogenic nutritional supplementation is sought by professional athletes for improving physical performance; nevertheless, scientific evidence to support the chronic use of L-Arginine among water polo players is missing. Methods Seventeen male professional water polo players were randomly assigned to assume 5 grams per day of L-Arginine (n = 9) or placebo (n = 8) for 4 weeks. The players' fitness level was assessed in the maximal speed swimming test. Ear lobe blood samples taken before and after the effort for serum lactate content were analyzed. A speed-to-lactate ratio was generated at the baseline and after 4 weeks of treatment. We also tested the effects of L-Arginine in vitro, measuring NO production, mitochondrial respiration, and gene expression in human fibroblasts. Results L-Arginine did not modify BMI, muscle strength, and maximal speed at 200 meters after 4 weeks. However, L-Arginine ameliorated oxidative metabolism to exercise as suggested by the statistically significant lower lactate-to-speed ratio, which was not observed in placebo-treated controls. In vitro, L-Arginine induced the expression of a key regulator of mitochondrial biogenesis (PGC1α) and genes encoding for complex I and increased the production of nitric oxide and the maximal oxygen consumption rate. Conclusions Chronic L-Arginine is safe and effective in ameliorating the oxidative metabolism of professional water polo players, through a mechanism of enhanced mitochondrial function.
Collapse
|
17
|
Nian Y, Iske J, Maenosono R, Minami K, Heinbokel T, Quante M, Liu Y, Azuma H, Yang J, Abdi R, Zhou H, Elkhal A, Tullius SG. Targeting age-specific changes in CD4 + T cell metabolism ameliorates alloimmune responses and prolongs graft survival. Aging Cell 2021; 20:e13299. [PMID: 33497523 PMCID: PMC7884034 DOI: 10.1111/acel.13299] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 11/16/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Age impacts alloimmunity. Effects of aging on T-cell metabolism and the potential to interfere with immunosuppressants have not been explored yet. Here, we dissected metabolic pathways of CD4+ and CD8+ T cells in aging and offer novel immunosuppressive targets. Upon activation, CD4+ T cells from old mice failed to exhibit adequate metabolic reprogramming resulting into compromised metabolic pathways, including oxidative phosphorylation (OXPHOS) and glycolysis. Comparable results were also observed in elderly human patients. Although glutaminolysis remained the dominant and age-independent source of mitochondria for activated CD4+ T cells, old but not young CD4+ T cells relied heavily on glutaminolysis. Treating young and old murine and human CD4+ T cells with 6-diazo-5-oxo-l-norleucine (DON), a glutaminolysis inhibitor resulted in significantly reduced IFN-γ production and compromised proliferative capacities specifically of old CD4+ T cells. Of translational relevance, old and young mice that had been transplanted with fully mismatched skin grafts and treated with DON demonstrated dampened Th1- and Th17-driven alloimmune responses. Moreover, DON diminished cytokine production and proliferation of old CD4+ T cells in vivo leading to a significantly prolonged allograft survival specifically in old recipients. Graft prolongation in young animals, in contrast, was only achieved when DON was applied in combination with an inhibition of glycolysis (2-deoxy-d-glucose, 2-DG) and OXPHOS (metformin), two alternative metabolic pathways. Notably, metabolic treatment had not been linked to toxicities. Remarkably, immunosuppressive capacities of DON were specific to CD4+ T cells as adoptively transferred young CD4+ T cells prevented immunosuppressive capacities of DON on allograft survival in old recipients. Depletion of CD8+ T cells did not alter transplant outcomes in either young or old recipients. Taken together, our data introduce an age-specific metabolic reprogramming of CD4+ T cells. Targeting those pathways offers novel and age-specific approaches for immunosuppression.
Collapse
Affiliation(s)
- Yeqi Nian
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Second Xiangya Hospital Central South University Changsha China
- Department of Kidney Transplantation Tianjin First Central Hospital Nankai University Tianjin China
| | - Jasper Iske
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Institute of Transplant Immunology Hannover Medical School Hannover Germany
| | - Ryoichi Maenosono
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Osaka Medical College Osaka Japan
| | - Koichiro Minami
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Urology Osaka Medical College Osaka Japan
| | - Timm Heinbokel
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Department of Pathology Charité – Universitätsmedizin Berlin Berlin Germany
| | - Markus Quante
- Department of General, Visceral‐ and Transplant Surgery University Hospital Tübingen Tubingen Germany
| | - Yang Liu
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
- Institute of Hepatobiliary Diseases Zhongnan Hospital of Wuhan University Wuhan China
| | | | - Jinrui Yang
- Department of Urology Second Xiangya Hospital Central South University Changsha China
| | - Reza Abdi
- Renal Division Transplantation Research Center Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Hao Zhou
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory Brigham and Women's Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
18
|
Selective Targeting of Cancerous Mitochondria and Suppression of Tumor Growth Using Redox-Active Treatment Adjuvant. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6212935. [PMID: 33204397 PMCID: PMC7652615 DOI: 10.1155/2020/6212935] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Redox-active substances and their combinations, such as of quinone/ascorbate and in particular menadione/ascorbate (M/A; also named Apatone®), attract attention with their unusual ability to kill cancer cells without affecting the viability of normal cells as well as with the synergistic anticancer effect of both molecules. So far, the primary mechanism of M/A-mediated anticancer effects has not been linked to the mitochondria. The aim of our study was to clarify whether this “combination drug” affects mitochondrial functionality specifically in cancer cells. Studies were conducted on cancer cells (Jurkat, Colon26, and MCF7) and normal cells (normal lymphocytes, FHC, and MCF10A), treated with different concentrations of menadione, ascorbate, and/or their combination (2/200, 3/300, 5/500, 10/1000, and 20/2000 μM/μM of M/A). M/A exhibited highly specific and synergistic suppression on cancer cell growth but without adversely affecting the viability of normal cells at pharmacologically attainable concentrations. In M/A-treated cancer cells, the cytostatic/cytotoxic effect is accompanied by (i) extremely high production of mitochondrial superoxide (up to 15-fold over the control level), (ii) a significant decrease of mitochondrial membrane potential, (iii) a decrease of the steady-state levels of ATP, succinate, NADH, and NAD+, and (iv) a decreased expression of programed cell death ligand 1 (PD-L1)—one of the major immune checkpoints. These effects were dose dependent. The inhibition of NQO1 by dicoumarol increased mitochondrial superoxide and sensitized cancer cells to M/A. In normal cells, M/A induced relatively low and dose-independent increase of mitochondrial superoxide and mild oxidative stress, which seems to be well tolerated. These data suggest that all anticancer effects of M/A result from a specific mechanism, tightly connected to the mitochondria of cancer cells. At low/tolerable doses of M/A (1/100-3/300 μM/μM) attainable in cancer by oral and parenteral administration, M/A sensitized cancer cells to conventional anticancer drugs, exhibiting synergistic or additive cytotoxicity accompanied by impressive induction of apoptosis. Combinations of M/A with 13 anticancer drugs were investigated (ABT-737, barasertib, bleomycin, BEZ-235, bortezomib, cisplatin, everolimus, lomustine, lonafarnib, MG-132, MLN-2238, palbociclib, and PI-103). Low/tolerable doses of M/A did not induce irreversible cytotoxicity in cancer cells but did cause irreversible metabolic changes, including: (i) a decrease of succinate and NADH, (ii) depolarization of the mitochondrial membrane, and (iii) overproduction of superoxide in the mitochondria of cancer cells only. In addition, M/A suppressed tumor growth in vivo after oral administration in mice with melanoma and the drug downregulated PD-L1 in melanoma cells. Experimental data suggest a great potential for beneficial anticancer effects of M/A through increasing the sensitivity of cancer cells to conventional anticancer therapy, as well as to the immune system, while sparing normal cells. We hypothesize that M/A-mediated anticancer effects are triggered by redox cycling of both substances, specifically within dysfunctional mitochondria. M/A may also have a beneficial effect on the immune system, making cancer cells “visible” and more vulnerable to the native immune response.
Collapse
|
19
|
miR-7 Regulates GLP-1-Mediated Insulin Release by Targeting β-Arrestin 1. Cells 2020; 9:cells9071621. [PMID: 32640511 PMCID: PMC7407368 DOI: 10.3390/cells9071621] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) has been shown to potentiate glucose-stimulated insulin secretion binding GLP-1 receptor on pancreatic β cells. β-arrestin 1 (βARR1) is known to regulate the desensitization of GLP-1 receptor. Mounting evidence indicates that microRNAs (miRNAs, miRs) are fundamental in the regulation of β cell function and insulin release. However, the regulation of GLP-1/βARR1 pathways by miRs has never been explored. Our hypothesis is that specific miRs can modulate the GLP-1/βARR1 axis in β cells. To test this hypothesis, we applied a bioinformatic approach to detect miRs that could target βARR1; we identified hsa-miR-7-5p (miR-7) and we validated the specific interaction of this miR with βARR1. Then, we verified that GLP-1 was indeed able to regulate the transcription of miR-7 and βARR1, and that miR-7 significantly regulated GLP-1-induced insulin release and cyclic AMP (cAMP) production in β cells. Taken together, our findings indicate, for the first time, that miR-7 plays a functional role in the regulation of GLP-1-mediated insulin release by targeting βARR1. These results have a decisive clinical impact given the importance of drugs modulating GLP-1 signaling in the treatment of patients with type 2 diabetes mellitus.
Collapse
|
20
|
Venado A, Kolaitis NA, Huang CY, Gao Y, Glidden DV, Soong A, Sutter N, Katz PP, Greenland JR, Calabrese DR, Hays SR, Golden JA, Shah RJ, Leard LE, Kukreja J, Deuse T, Wolters PJ, Covinsky K, Blanc PD, Singer JP. Frailty after lung transplantation is associated with impaired health-related quality of life and mortality. Thorax 2020; 75:669-678. [PMID: 32376733 DOI: 10.1136/thoraxjnl-2019-213988] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Lung transplantation and related medications are associated with pathobiological changes that can induce frailty, a state of decreased physiological reserve. Causes of persistent or emergent frailty after lung transplantation, and whether such transplant-related frailty is associated with key outcomes, are unknown. METHODS Frailty and health-related quality of life (HRQL) were prospectively measured repeatedly for up to 3 years after lung transplantation. Frailty, quantified by the Short Physical Performance Battery (SPPB), was tested as a time-dependent binary and continuous predictor. The association of transplant-related frailty with HRQL and mortality was evaluated using mixed effects and Cox regression models, respectively, adjusting for age, sex, ethnicity, diagnosis, and for body mass index and lung function as time-dependent covariates. We tested the association between measures of body composition, malnutrition, renal dysfunction and immunosuppressants on the development of frailty using mixed effects models with time-dependent predictors and lagged frailty outcomes. RESULTS Among 259 adults (56% male; mean age 55.9±12.3 years), transplant-related frailty was associated with lower HRQL. Frailty was also associated with a 2.5-fold higher mortality risk (HR 2.51; 95% CI 1.21 to 5.23). Further, each 1-point worsening in SPPB was associated, on average, with a 13% higher mortality risk (HR 1.13; 95% CI 1.04 to 1.23). Secondarily, we found that sarcopenia, underweight and obesity, malnutrition, and renal dysfunction were associated with the development of frailty after transplant. CONCLUSIONS Transplant-related frailty is associated with lower HRQL and higher mortality in lung recipients. Abnormal body composition, malnutrition and renal dysfunction may contribute to the development of frailty after transplant. Confirming the role of these potential contributors and developing interventions to mitigate frailty may improve lung transplant success.
Collapse
Affiliation(s)
- Aida Venado
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Nicholas A Kolaitis
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Chiung-Yu Huang
- Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Ying Gao
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - David V Glidden
- Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Allison Soong
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Nicole Sutter
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Patricia P Katz
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - John R Greenland
- Medicine, University of California San Francisco, San Francisco, California, USA.,Medicine, VA Medical Center, San Francisco, California, USA
| | - Daniel R Calabrese
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Steven R Hays
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey A Golden
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Rupal J Shah
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lorriana E Leard
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jasleen Kukreja
- Surgery, University of California San Francisco, San Francisco, California, USA
| | - Tobias Deuse
- Surgery, University of California San Francisco, San Francisco, California, USA
| | - Paul J Wolters
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kenneth Covinsky
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Paul D Blanc
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jonathan P Singer
- Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Amgalan D, Garner TP, Pekson R, Jia XF, Yanamandala M, Paulino V, Liang FG, Corbalan JJ, Lee J, Chen Y, Karagiannis GS, Sanchez LR, Liang H, Narayanagari SR, Mitchell K, Lopez A, Margulets V, Scarlata M, Santulli G, Asnani A, Peterson RT, Hazan RB, Condeelis JS, Oktay MH, Steidl U, Kirshenbaum LA, Gavathiotis E, Kitsis RN. A small-molecule allosteric inhibitor of BAX protects against doxorubicin-induced cardiomyopathy. NATURE CANCER 2020; 1:315-328. [PMID: 32776015 PMCID: PMC7413180 DOI: 10.1038/s43018-020-0039-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/29/2020] [Indexed: 12/27/2022]
Abstract
Doxorubicin remains an essential component of many cancer regimens, but its use is limited by lethal cardiomyopathy, which has been difficult to target, owing to pleiotropic mechanisms leading to apoptotic and necrotic cardiac cell death. Here we show that BAX is rate-limiting in doxorubicin-induced cardiomyopathy and identify a small-molecule BAX inhibitor that blocks both apoptosis and necrosis to prevent this syndrome. By allosterically inhibiting BAX conformational activation, this compound blocks BAX translocation to mitochondria, thereby abrogating both forms of cell death. When co-administered with doxorubicin, this BAX inhibitor prevents cardiomyopathy in zebrafish and mice. Notably, cardioprotection does not compromise the efficacy of doxorubicin in reducing leukemia or breast cancer burden in vivo, primarily due to increased priming of mitochondrial death mechanisms and higher BAX levels in cancer cells. This study identifies BAX as an actionable target for doxorubicin-induced cardiomyopathy and provides a prototype small-molecule therapeutic.
Collapse
Affiliation(s)
- Dulguun Amgalan
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Thomas P Garner
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ryan Pekson
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiaotong F Jia
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mounica Yanamandala
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Cardiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor Paulino
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Felix G Liang
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Jose Corbalan
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jaehoon Lee
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yun Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - George S Karagiannis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Luis Rivera Sanchez
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Surgery, Montefiore Medical Center, Bronx, NY, USA
| | - Huizhi Liang
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Swathi-Rao Narayanagari
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Lopez
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Victoria Margulets
- Departments of Physiology and Pathophysiology and Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Marco Scarlata
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aarti Asnani
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Randall T Peterson
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Rachel B Hazan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John S Condeelis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Surgery, Montefiore Medical Center, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maja H Oktay
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ulrich Steidl
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lorrie A Kirshenbaum
- Departments of Physiology and Pathophysiology and Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Evripidis Gavathiotis
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA.
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA.
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Richard N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx NY, USA.
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
22
|
Torabi J, Konicki A, Rocca JP, Ajaimy M, Campbell A, Azzi Y, Pynadath C, Liriano-Ward L, Akalin E, Kinkhabwala M, Graham JA. The use of LCP-Tacrolimus (Envarsus XR) in simultaneous pancreas and kidney (SPK) transplant recipients. Am J Surg 2020; 219:583-586. [PMID: 32122660 DOI: 10.1016/j.amjsurg.2020.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/23/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Extended release LCP-tacrolimus (LCPT) allows once-daily dosing in transplant recipients. The improved bioavailability may be beneficial for simultaneous pancreas-kidney recipients (SPK). METHODS This is a study of 39 SPK recipients on standard immediate-release tacrolimus (IR-TAC, n = 21) or LCPT (n = 18). Coefficient of variability (CV = 100∗standard deviation/mean) was calculated to assess drug levels. Hemoglobin A1c (HbA1c), tacrolimus and creatinine levels were measured postoperatively. RESULTS There was no difference in tacrolimus CV in the IR-TAC and LCPT groups at 1 month or 3 months postoperatively; however, a greater difference was observed at 1 year (41.0 vs. 33.1%; p = 0.19). There were six episodes of acute rejection in the IR-TAC group compared to zero episodes in the LCPT group (p = 0.01). HbA1c was significantly higher in the IR-TAC group compared to LCPT at 3 (5.5 vs. 4.9%, p = 0.01), 6 (5.6 vs. 4.9%, p = 0.01) and 12 months (5.8 vs. 5.1%, p = 0.07). CONCLUSIONS Significantly lower rates of rejection were observed in patients receiving LCPT. The once daily dosing may facilitate medication adherence and result in improved long-term outcomes.
Collapse
Affiliation(s)
- Julia Torabi
- Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Juan P Rocca
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Maria Ajaimy
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Alesa Campbell
- Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Yorg Azzi
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Cindy Pynadath
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Luz Liriano-Ward
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Enver Akalin
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Milan Kinkhabwala
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA
| | - Jay A Graham
- Albert Einstein College of Medicine, Bronx, NY, USA; Montefiore-Einstein Center for Transplantation, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
23
|
Triñanes J, Ten Dijke P, Groen N, Hanegraaf M, Porrini E, Rodriguez-Rodriguez AE, Drachenberg C, Rabelink TJ, de Koning E, Carlotti F, de Vries APJ. Tacrolimus-Induced BMP/SMAD Signaling Associates With Metabolic Stress-Activated FOXO1 to Trigger β-Cell Failure. Diabetes 2020; 69:193-204. [PMID: 31732500 DOI: 10.2337/db19-0828] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/06/2019] [Indexed: 11/13/2022]
Abstract
Active maintenance of β-cell identity through fine-tuned regulation of key transcription factors ensures β-cell function. Tacrolimus, a widely used immunosuppressant, accelerates onset of diabetes after organ transplantation, but underlying molecular mechanisms are unclear. Here we show that tacrolimus induces loss of human β-cell maturity and β-cell failure through activation of the BMP/SMAD signaling pathway when administered under mild metabolic stress conditions. Tacrolimus-induced phosphorylated SMAD1/5 acts in synergy with metabolic stress-activated FOXO1 through formation of a complex. This interaction is associated with reduced expression of the key β-cell transcription factor MAFA and abolished insulin secretion, both in vitro in primary human islets and in vivo in human islets transplanted into high-fat diet-fed mice. Pharmacological inhibition of BMP signaling protects human β-cells from tacrolimus-induced β-cell dysfunction in vitro. Furthermore, we confirm that BMP/SMAD signaling is activated in protocol pancreas allograft biopsies from recipients on tacrolimus. To conclude, we propose a novel mechanism underlying the diabetogenicity of tacrolimus in primary human β-cells. This insight could lead to new treatment strategies for new-onset diabetes and may have implications for other forms of diabetes.
Collapse
Affiliation(s)
- Javier Triñanes
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Nathalie Groen
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike Hanegraaf
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Esteban Porrini
- Instituto de Tecnología Biomédicas and Hospital Universitario de Canarias, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Ana E Rodriguez-Rodriguez
- Instituto de Tecnología Biomédicas and Hospital Universitario de Canarias, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD
| | - Ton J Rabelink
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelco de Koning
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Françoise Carlotti
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Aiko P J de Vries
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Transplant Center, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
24
|
Ellis S, Rang C, Kotsimbos T, Keating D, Finlayson F, Stark R, Thyagarajan D, Wilson J. CNS imaging studies in cystic fibrosis patients presenting with sudden neurological events. BMJ Open Respir Res 2019; 6:e000456. [PMID: 31423315 PMCID: PMC6688669 DOI: 10.1136/bmjresp-2019-000456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/13/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
Background Acute neurological events may present as an extrapulmonary complication in patients with cystic fibrosis (CF). These events can be secondary to a range of different aetiologies. Methods A retrospective analysis of 476 medical records of CF patients attending a large teaching hospital between 2000 and 2018 was performed. Patients presenting with acute neurological events who had MRI brain imaging were evaluated. Patients who had headaches without associated neurological symptoms were excluded from this analysis. Results Acute neurological presentations, excluding headaches without associated neurological symptoms, were reported in 27 index patients out of the 476 patients. Of these, 16 patients had MRI brain imaging for review. Three patients suffered pathology secondary to vascular events, both ischaemic and haemorrhagic; four patients had evidence of ischaemia or infarction not consistent with a vascular territory stroke and the remaining patients experienced a range of different neurological events. The most common presentation among these patients was seizure activity, followed by a transient motor or sensory deficit. Conclusions Neurological complications are recognised among individuals with CF. Although rare, they can be secondary to a range of different aetiologies, including dysfunctional cell energetics. Additional studies are required to further evaluate this association.
Collapse
Affiliation(s)
- Samantha Ellis
- Department of Radiology, Alfred Health, Melbourne, Victoria, Australia
| | - Catherine Rang
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Tom Kotsimbos
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Dominic Keating
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Felicity Finlayson
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Richard Stark
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | | | - John Wilson
- Department of Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Wei S, Li T, Xie R, Ye B, Xiang J, Liu K, Chen Z, Gao X. The role of ATF3 in ZnO nanoparticle-induced genotoxicity and cytotoxicity in bronchial epithelial cells. Int J Biochem Cell Biol 2019; 113:95-102. [PMID: 31220582 DOI: 10.1016/j.biocel.2019.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/06/2019] [Accepted: 06/14/2019] [Indexed: 12/15/2022]
Abstract
ZnO nanoparticle (ZnO NP) exposure causes oxidative stress in the respiratory system, leading to pulmonary damage. Activating transcription factor 3 (ATF3) participates in a variety of cellular stress responses. However, the role of ATF3 in ZnO NP genotoxicity and cytotoxicity remains to be explored. Here we reported that ZnO NP treatment dramatically induced the expression of ATF3 in human bronchial epithelial (HBE) cells, which was mediated by the nuclear factor erythroid 2-related factor 2 (Nrf2). ATF3 was required for the repair of ZnO NP-induced DNA damage as gamma foci number increased when endogenous ATF3 was silenced. Moreover, ATF3 also contributed to ZnO NP-induced cell apoptosis. Mechanistic study revealed that ATF3 interacted with the p53 protein and upregulated its expression under ZnO NP treatment. Collectively, our findings demonstrated ATF3 as an important regulator of epithelial homeostasis by promoting both DNA repair and the death of damaged cells under ZnO NP-induced genotoxic stress.
Collapse
Affiliation(s)
- Saisai Wei
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tiezheng Li
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Renxiang Xie
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Bingqi Ye
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jie Xiang
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kangli Liu
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhanghui Chen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Xiangwei Gao
- Institute of Environmental Health, and Sir Run-Run Shaw Hospital, and Institute of Environmental Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
26
|
Matsushima Y, Terada K, Kamei C, Sugimoto Y. Sertraline inhibits nerve growth factor-induced neurite outgrowth in PC12 cells via a mechanism involving the sigma-1 receptor. Eur J Pharmacol 2019; 853:129-135. [PMID: 30902656 DOI: 10.1016/j.ejphar.2019.03.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 03/18/2019] [Indexed: 02/02/2023]
Abstract
The selective serotonin reuptake inhibitors (SSRIs) fluvoxamine and sertraline show a high affinity for sigma-1 receptors. Fluvoxamine enhances nerve growth factor (NGF)-induced neurite outgrowth in PC12 cells via a sigma-1 receptor-mediated mechanism, which suggests that neurogenesis may be involved in the antidepressant action of fluvoxamine. However, the effects of sertraline on neurite outgrowth remain unclear. Here, we report the effects of sertraline on NGF-induced neurite outgrowth in PC12 cells. At concentrations above 0.3 μM, sertraline inhibited neurite outgrowth induced by NGF (50 ng/mL) in PC12 cells in a concentration-dependent manner. At 0.3-3 μM, sertraline inhibited NGF-induced neurite outgrowth; however, had no effect on cell viability. This suggests that at these concentrations, sertraline inhibits NGF-induced neurite outgrowth without causing cell toxicity. Because sertraline has a high affinity for the sigma-1 receptor, we investigated whether this receptor is involved in sertraline's inhibitory effect on NGF-induced neurite outgrowth. The effect was reversed by both the sigma-1 receptor agonist PRE-084 and the sigma-1 receptor antagonist NE-100. These results suggest that sertraline inhibits NGF-induced neurite outgrowth in PC12 cells by acting as an inverse agonist of the sigma-1 receptor in this system.
Collapse
Affiliation(s)
- Yukari Matsushima
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan; Department of Kampo and Natural Product Chemistry, Yokohama University of Pharmacy, 601 Matanocho, Totsuka-ku, Yokohama 245-0066, Japan
| | - Kazuki Terada
- Laboratory of Drug Design and Drug Delivery, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Chiaki Kamei
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Yumi Sugimoto
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1 Kamiohno, Himeji 670-8524, Japan.
| |
Collapse
|
27
|
Luo K, Yu JH, Quan Y, Shin YJ, Lee KE, Kim HL, Ko EJ, Chung BH, Lim SW, Yang CW. Therapeutic potential of coenzyme Q 10 in mitochondrial dysfunction during tacrolimus-induced beta cell injury. Sci Rep 2019; 9:7995. [PMID: 31142763 PMCID: PMC6541596 DOI: 10.1038/s41598-019-44475-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/17/2019] [Indexed: 02/06/2023] Open
Abstract
We previously reported that oxidative stress induced by long-term tacrolimus treatment impairs mitochondrial function in pancreatic beta cells. In this study, we aimed to investigate the therapeutic potential of coenzyme Q10, which is known to be a powerful antioxidant, in mitochondrial dysfunction in tacrolimus-induced diabetic rats. In a rat model of tacrolimus-induced diabetes mellitus, coenzyme Q10 treatment improved pancreatic beta cell function. The administration of coenzyme Q10 improved insulin immunoreactivity within islets, which was accompanied by reductions in oxidative stress and apoptosis. Assessment of the mitochondrial ultrastructure by electron microscopy revealed that coenzyme Q10 treatment increased the size, number, and volume of mitochondria, as well as the number of insulin granules compared with that induced by tacrolimus treatment alone. An in vitro study using a pancreatic beta cell line showed that tacrolimus treatment increased apoptosis and the production of mitochondrial reactive oxygen species, while cotreatment with coenzyme Q10 effectively attenuated these alterations. At the subcellular level, tacrolimus-induced impairment of mitochondrial respiration was significantly improved by coenzyme Q10, as evidenced by the increased mitochondrial oxygen consumption and ATP production. Our data indicate that coenzyme Q10 plays an important role in reducing tacrolimus-induced oxidative stress and protects the mitochondria in pancreatic beta cells. These findings suggest that supplementation with coenzyme Q10 has beneficial effects in tacrolimus-induced diabetes mellitus.
Collapse
Affiliation(s)
- Kang Luo
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Yu
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Yi Quan
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Hong Lim Kim
- Integrative Research Support Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| | - Sun Woo Lim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea. .,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Transplant Research Center, The Catholic University of Korea School of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea School of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Zhao H, Tong G, Liu J, Wang J, Zhang H, Bai J, Hou L, Zhang Z. IP3R and RyR channels are involved in traffic-related PM 2.5-induced disorders of calcium homeostasis. Toxicol Ind Health 2019; 35:339-348. [PMID: 31023176 DOI: 10.1177/0748233719843763] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Traffic-related PM2.5 can result in immune system damage and diseases; however, the possible mechanism of its effect remains unclear. Calcium (Ca2+) is a critical signaling molecule in a variety of cells. Indeed, Ca2+ is involved in numerous basic functions, including cell growth and death. In this study, Jurkat T cells were used to explore the possible mechanisms of PM2.5-elicited intracellular Ca2+signal responses. The results indicate that PM2.5 could raise the level of intracellular Ca2+ concentration ([Ca2+]i). The [Ca2+]i in Jurkat T cells significantly decreased after treatment with heparin as an inhibitor of inositol trisphosphate receptors (IP3 R), or procaine as an inhibitor of ryanodine receptors (RyR). The expression of calmodulin (CAM) protein decreased in a time-dependent manner after exposure to PM2.5, whereas the activity of Ca2+-Mg2+-ATPase seemed to show a slight drop trend after exposure to PM2.5. Our findings demonstrate that PM2.5 stimulation to Jurkat T cells would result in an increase in [Ca2+]i, which is modulated by IP3 R and RyR, as well as CAM.
Collapse
Affiliation(s)
- Huichao Zhao
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Guoqiang Tong
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jiejing Liu
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jing Wang
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Hongmei Zhang
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jianying Bai
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Lifang Hou
- 2 Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,3 Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zhihong Zhang
- 1 Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| |
Collapse
|
29
|
Jouve T, Noble J, Rostaing L, Malvezzi P. An update on the safety of tacrolimus in kidney transplant recipients, with a focus on tacrolimus minimization. Expert Opin Drug Saf 2019; 18:285-294. [DOI: 10.1080/14740338.2019.1599858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Thomas Jouve
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, CHU Grenoble-Alpes, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Johan Noble
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, CHU Grenoble-Alpes, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Lionel Rostaing
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, CHU Grenoble-Alpes, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Paolo Malvezzi
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, CHU Grenoble-Alpes, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
30
|
Chareyron I, Wall C, Thevenet J, Santo-Domingo J, Wiederkehr A. Cellular stress is a prerequisite for glucose-induced mitochondrial matrix alkalinization in pancreatic β-cells. Mol Cell Endocrinol 2019; 481:71-83. [PMID: 30476561 DOI: 10.1016/j.mce.2018.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/20/2018] [Accepted: 11/22/2018] [Indexed: 11/24/2022]
Abstract
Changes in mitochondrial and cytosolic pH alter the chemical gradient across the inner mitochondrial membrane. The proton chemical gradient contributes to mitochondrial ATP synthesis as well as the uptake and release of metabolites and ions from the organelle. Here mitochondrial pH and ΔpH were studied for the first time in human pancreatic β-cells. Adenoviruses were used for rat insulin promoter dependent expression of the pH sensor SypHer targeted to either the mitochondrial matrix or the cytosol. The matrix pH in resting human β-cells is low (pH = 7.50 ± SD 0.17) compared to published values in other cell types. Consequently, the ΔpH of β-cells mitochondria is small. Glucose stimulation consistently resulted in acidification of the matrix pH in INS-1E insulinoma cells and β-cells in intact human islets or islet monolayer cultures. We registered acidification with similar kinetics but of slightly smaller amplitude in the cytosol of β-cells, thus glucose stimulation further reduced the ΔpH. Infection of human islets with high levels of adenoviruses caused the mitochondrial pH to increase. The apoptosis inducer and broad-spectrum kinase inhibitor staurosporine had similar effects on pH homeostasis. Although staurosporine alone does not affect the mitochondrial pH, glucose slightly increases the matrix pH of staurosporine treated cells. These two cellular stressors alter the normal mitochondrial pH response to glucose in pancreatic β-cells.
Collapse
Affiliation(s)
- Isabelle Chareyron
- Mitochondrial Function, Nestlé Institute of Health Sciences, 1015, Lausanne, Switzerland
| | - Christopher Wall
- Mitochondrial Function, Nestlé Institute of Health Sciences, 1015, Lausanne, Switzerland
| | - Jonathan Thevenet
- Mitochondrial Function, Nestlé Institute of Health Sciences, 1015, Lausanne, Switzerland
| | - Jaime Santo-Domingo
- Mitochondrial Function, Nestlé Institute of Health Sciences, 1015, Lausanne, Switzerland
| | - Andreas Wiederkehr
- Mitochondrial Function, Nestlé Institute of Health Sciences, 1015, Lausanne, Switzerland.
| |
Collapse
|
31
|
Shu J, Matarese A, Santulli G. Diabetes, body fat, skeletal muscle, and hypertension: The ominous chiasmus? J Clin Hypertens (Greenwich) 2018; 21:239-242. [PMID: 30525276 DOI: 10.1111/jch.13453] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jun Shu
- Department of Medicine, Albert Einstein College of Medicine, Montefiore University Hospital, New York, New York
| | - Alessandro Matarese
- Department of Pneumology and Oncology, AORN "Ospedale dei Colli", Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, Montefiore University Hospital, New York, New York.,Department of Biomedical Advanced Sciences, "Federico II" University, Naples, Italy
| |
Collapse
|
32
|
Saeedi Borujeni MJ, Esfandiary E, Baradaran A, Valiani A, Ghanadian M, Codoñer-Franch P, Basirat R, Alonso-Iglesias E, Mirzaei H, Yazdani A. Molecular aspects of pancreatic β-cell dysfunction: Oxidative stress, microRNA, and long noncoding RNA. J Cell Physiol 2018; 234:8411-8425. [PMID: 30565679 DOI: 10.1002/jcp.27755] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Metabolic syndrome is known as a frequent precursor of type 2 diabetes mellitus (T2D). This disease could affect 8% of the people worldwide. Given that pancreatic β-cell dysfunction and loss have central roles in the initiation and progression of the disease, the understanding of cellular and molecular pathways associated with pancreatic β-cell dysfunction can provide more information about the underlying pathways involved in T2D. Multiple lines evidence indicated that oxidative stress, microRNA, and long noncoding RNA play significant roles in various steps of diseases. Oxidative stress is one of the important factors involved in T2D pathogenesis. This could affect the function and survival of the β cell via activation or inhibition of several processes and targets, such as receptor-signal transduction, enzyme activity, gene expression, ion channel transport, and apoptosis. Besides oxidative stress, microRNAs and noncoding RNAs have emerged as epigenetic regulators that could affect pancreatic β-cell dysfunction. These molecules exert their effects via targeting a variety of cellular and molecular pathways involved in T2D pathogenesis. Here, we summarized the molecular aspects of pancreatic β-cell dysfunction. Moreover, we highlighted the roles of oxidative stress, microRNAs, and noncoding RNAs in pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Mohammad Javad Saeedi Borujeni
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiary
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azar Baradaran
- Department of Pathology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Valiani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mustafa Ghanadian
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Reyhane Basirat
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Amid Yazdani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
33
|
Lombardi A, Tsomos E, Hammerstad SS, Tomer Y. Interferon alpha: The key trigger of type 1 diabetes. J Autoimmun 2018; 94:7-15. [PMID: 30115527 DOI: 10.1016/j.jaut.2018.08.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/04/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
IFNα is a cytokine essential to a vast array of immunologic processes. Its induction early in the innate immune response provides a priming mechanism that orchestrates numerous subsequent pathways in innate and adaptive immunity. Despite its beneficial effects in viral infections IFNα has been reported to be associated with several autoimmune diseases including autoimmune thyroid disease, systemic lupus erythematosus, rheumatoid arthritis, primary biliary cholangitis, and recently emerged as a major cytokine that triggers Type 1 Diabetes. In this review, we dissect the role of IFNα in T1D, focusing on the potential pathophysiological mechanisms involved. Evidence from human and mouse studies indicates that IFNα plays a key role in enhancing islet expression of HLA-I in patients with T1D, thereby increasing autoantigen presentation and beta cell activation of autoreactive cytotoxic CD8 T-lymphocytes. The binding of IFNα to its receptor induces the secretion of chemokines, attracting monocytes, T lymphocytes, and NK cells to the infected tissue triggering autoimmunity in susceptible individuals. Furthermore, IFNα impairs insulin production through the induction of endoplasmic reticulum stress as well as by impairing mitochondrial function. Due to its central role in the early phases of beta cell death, targeting IFNα and its pathways in genetically predisposed individuals may represent a potential novel therapeutic strategy in the very early stages of T1D.
Collapse
Affiliation(s)
- Angela Lombardi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Effie Tsomos
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sara S Hammerstad
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Aker, Oslo, Norway; Department of Pediatrics, Oslo University Hospital, Ulleval, Oslo, Norway
| | - Yaron Tomer
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
34
|
De novo aortopathy and cardiovascular outcomes in paediatric liver transplant recipients. Cardiol Young 2018; 28:986-994. [PMID: 29972110 DOI: 10.1017/s104795111800063x] [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] [Indexed: 11/07/2022]
Abstract
With the increase in long-term survival of post-transplant children, there is a paradigm shift in the emphasis of post-transplant care. We describe de novo cardiovascular abnormalities, which occurred in otherwise asymptomatic paediatric liver transplant recipients, who received liver allografts between 1991 and 2014 at the National University Hospital, Singapore, detected during routine post-transplant monitoring. A total of 96 paediatric liver transplants were performed in 90 children. After transplant, 7/90 (7.8%) recipients were identified with new-onset aortopathy. Glycogen storage disease type I (42.9% versus 2.4%; p<0.001) and recipient Epstein-Barr virus seropositivity (85.7 versus 31.0%, p=0.004) were significant risk factors for aortopathy on univariate analysis. On multivariate analysis, only glycogen storage disease type I remained as the significant risk factor (odds ratio 51.3 [95% confidence intervals: 1.1-2498.1, p=0.047]). Liver transplant is a double-edged sword that reverses certain cardiopulmonary complications of end-stage liver disease but may induce de novo structural cardiac injury in the form of aortic dilation.
Collapse
|
35
|
Chen HQ, Gao D. Inhibitory effect of microRNA-154 targeting WHSC1 on cell proliferation of human skin squamous cell carcinoma through mediating the P53 signaling pathway. Int J Biochem Cell Biol 2018; 100:22-29. [PMID: 29727714 DOI: 10.1016/j.biocel.2018.04.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Skin squamous cell carcinoma (SCC) is a common, morbid, and frequently lethal malignancy and ranks as the sixth most deadly cancer worldwide. Hence, this study aims to explore the effect of microRNA-154 (miR-154) targeting WHSC1 on proliferation and apoptosis of SCC cells via the P53 signaling pathway. METHODS The targeting relationship between WHSC1 and miR-154 was validated using dual-luciferase reporter assay. Normal human epidermal keratinocytes (NHEK) were included, and SCC A431 and SCC-15 cell lines were cultured and transfected with miR-154 mimic, miR-154 inhibitor or siRNA-WHSC1. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis were used for the miR-154 expression and levels of WHSC1, P53 signaling pathway- and apoptosis-related genes. MTT assay and flow cytometry were applied to determine the cell viability and apoptosis. RESULTS WHSC1 is a target gene of miR-154. MiR-154 negatively regulated WHSC1 expression and inhibited the activation of P53 signaling pathway. In response to miR-154 mimic or siRNA-WHSC1, SCC A431 and SCC-15 cell lines exhibited increased expression of P73, P16 and Bax, decreased expression of WHSC1, P53, c-myc and Bcl-2, as well as attenuated cell viability and enhanced cell apoptosis. The treatment of miR-154 inhibitor reversed the tendency. CONCLUSION These results demonstrate that up-regulation of miR-154 inhibits proliferation and induces apoptosis of human skin SCC cells by down-regulating WHSC1 and blocking the P53 signaling pathway.
Collapse
Affiliation(s)
- Hong-Quan Chen
- Department of Dermatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266003, Shandong Province, PR China
| | - Dong Gao
- Department of Dermatology, Yantai Yu Huang Ding Hospital, No. 20 Yu Huang Ding East Road, Yantai, 264000, Shandong Province, PR China.
| |
Collapse
|
36
|
Liu Y, Chen LY, Zeng H, Ward R, Wu N, Ma L, Mu X, Li QL, Yang Y, An S, Guo XX, Hao Q, Xu TR. Assessing the real-time activation of the cannabinoid CB1 receptor and the associated structural changes using a FRET biosensor. Int J Biochem Cell Biol 2018; 99:114-124. [PMID: 29626639 DOI: 10.1016/j.biocel.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/25/2018] [Accepted: 04/04/2018] [Indexed: 11/18/2022]
Abstract
The cannabinoid receptor 1 (CB1) is mainly expressed in the nervous system and regulates learning, memory processes, pain and energy metabolism. However, there is no way to directly measure its activation. In this study, we constructed a CB1 intramolecular fluorescence resonance energy transfer (FRET) sensor, which could measure CB1 activation by monitoring structural changes between the third intracellular loop and the C-terminal tail. CB1 agonists induced a time- and concentration-dependent increase in the FRET signal, corresponding to a reduction in the distance between the third intracellular loop and the C-terminal tail. This, in turn, mobilized intracellular Ca2+, inhibited cAMP accumulation, and increased phosphorylation of the ERK1/2 MAP kinases. The activation kinetics detected using this method were consistent with those from previous reports. Moreover, the increased FRET signal was markedly inhibited by the CB1 antagonist rimonabant, which also reduced phosphorylation of the ERK1/2 MAP kinases. We mutated a single cysteine residue in the sensor (at position 257 or 264) to alanine. Both mutation reduced the agonist-induced increase in FRET signal and structural changes in the CB1 receptor, which attenuated phosphorylation of the ERK1/2 MAP kinases. In summary, our sensor directly assesses the kinetics of CB1 activation in real-time and can be used to monitor CB1 structure and function.
Collapse
Affiliation(s)
- Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Lu-Yao Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Hong Zeng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Richard Ward
- Center for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Nan Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Li Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Xi Mu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Qiu-Lan Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Qian Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| |
Collapse
|
37
|
Wahl MP, Scalzo RL, Regensteiner JG, Reusch JEB. Mechanisms of Aerobic Exercise Impairment in Diabetes: A Narrative Review. Front Endocrinol (Lausanne) 2018; 9:181. [PMID: 29720965 PMCID: PMC5915473 DOI: 10.3389/fendo.2018.00181] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/21/2022] Open
Abstract
The prevalence of diabetes in the United States and globally has been rapidly increasing over the last several decades. There are now estimated to be 30.3 million people in the United States and 422 million people worldwide with diabetes. Diabetes is associated with a greatly increased risk of cardiovascular mortality, which is the leading cause of death in adults with diabetes. While exercise training is a cornerstone of diabetes treatment, people with diabetes have well-described aerobic exercise impairments that may create an additional diabetes-specific barrier to adding regular exercise to their lifestyle. Physiologic mechanisms linked to exercise impairment in diabetes include insulin resistance, cardiac abnormalities, mitochondrial function, and the ability of the body to supply oxygen. In this paper, we highlight the abnormalities of exercise in type 2 diabetes as well as potential therapeutic approaches.
Collapse
Affiliation(s)
- Matthew P. Wahl
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, CO, United States
- Veterans Administration Eastern Colorado Health Care System, Denver, CO, United States
| | - Rebecca L. Scalzo
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, CO, United States
- Center for Women’s Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Judith G. Regensteiner
- Center for Women’s Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Jane E. B. Reusch
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, CO, United States
- Veterans Administration Eastern Colorado Health Care System, Denver, CO, United States
- Center for Women’s Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- *Correspondence: Jane E. B. Reusch,
| |
Collapse
|
38
|
Santulli G, Lewis D, des Georges A, Marks AR, Frank J. Ryanodine Receptor Structure and Function in Health and Disease. Subcell Biochem 2018; 87:329-352. [PMID: 29464565 PMCID: PMC5936639 DOI: 10.1007/978-981-10-7757-9_11] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ryanodine receptors (RyRs) are ubiquitous intracellular calcium (Ca2+) release channels required for the function of many organs including heart and skeletal muscle, synaptic transmission in the brain, pancreatic beta cell function, and vascular tone. In disease, defective function of RyRs due either to stress (hyperadrenergic and/or oxidative overload) or genetic mutations can render the channels leaky to Ca2+ and promote defective disease-causing signals as observed in heat failure, muscular dystrophy, diabetes mellitus, and neurodegerative disease. RyRs are massive structures comprising the largest known ion channel-bearing macromolecular complex and exceeding 3 million Daltons in molecular weight. RyRs mediate the rapid release of Ca2+ from the endoplasmic/sarcoplasmic reticulum (ER/SR) to stimulate cellular functions through Ca2+-dependent processes. Recent advances in single-particle cryogenic electron microscopy (cryo-EM) have enabled the determination of atomic-level structures for RyR for the first time. These structures have illuminated the mechanisms by which these critical ion channels function and interact with regulatory ligands. In the present chapter we discuss the structure, functional elements, gating and activation mechanisms of RyRs in normal and disease states.
Collapse
Affiliation(s)
- Gaetano Santulli
- The Wu Center for Molecular Cardiology, Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
- The Wilf Family Cardiovascular Research Institute and the Einstein-Mount Sinai Diabetes Research Center, Department of Medicine, Albert Einstein College of Medicine - Montefiore University Hospital, New York, NY, USA
| | - Daniel Lewis
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Amedee des Georges
- Advanced Science Research Center at the Graduate Center of the City University of New York, New York, NY, USA
- Department of Chemistry & Biochemistry, City College of New York, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
39
|
Barra JM, Tse HM. Redox-Dependent Inflammation in Islet Transplantation Rejection. Front Endocrinol (Lausanne) 2018; 9:175. [PMID: 29740396 PMCID: PMC5924790 DOI: 10.3389/fendo.2018.00175] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/03/2018] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease that results in the progressive destruction of insulin-producing pancreatic β-cells inside the islets of Langerhans. The loss of this vital population leaves patients with a lifelong dependency on exogenous insulin and puts them at risk for life-threatening complications. One method being investigated to help restore insulin independence in these patients is islet cell transplantation. However, challenges associated with transplant rejection and islet viability have prevented long-term β-cell function. Redox signaling and the production of reactive oxygen species (ROS) by recipient immune cells and transplanted islets themselves are key players in graft rejection. Therefore, dissipation of ROS generation is a viable intervention that can protect transplanted islets from immune-mediated destruction. Here, we will discuss the newly appreciated role of redox signaling and ROS synthesis during graft rejection as well as new strategies being tested for their efficacy in redox modulation during islet cell transplantation.
Collapse
|