101
|
Ge X, Meng Q, Liu X, Liu J, Ma X, Shi S, Li M, Lin F, Liang X, Gong X, Liu Z, Han W, Zhou X. Alterations of long noncoding RNAs and mRNAs in extracellular vesicles derived from the murine heart post-ischemia-reperfusion injury. J Cell Mol Med 2022; 26:6006-6018. [PMID: 36444487 PMCID: PMC9753460 DOI: 10.1111/jcmm.17617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022] Open
Abstract
Extracellular vesicles (EVs) play important roles in cardiovascular diseases by delivering their RNA cargos. However, the features and possible role of the lncRNAs and mRNAs in cardiac EVs during ischemia-reperfusion (IR) remain unclear. Therefore, we performed RNA sequencing analysis to profile the features of lncRNAs and mRNAs and predicted their potential functions. Here, we demonstrated that the severity of IR injury was significantly correlated with cardiac EV production. RNA sequencing identified 73 significantly differentially expressed (DE) lncRNAs (39 upregulated and 34 downregulated) and 720 DE-mRNAs (317 upregulated and 403 downregulated). Gene Ontology (GO) and pathway analysis were performed to predict the potential functions of the DE-lncRNAs and mRNAs. The lncRNA-miRNA-mRNA ceRNA network showed the possible functions of DE-lncRNAs with DE-mRNAs which are enriched in the pathways of T cell receptor signalling pathway and cell adhesion molecules. Moreover, the expressions of ENSMUST00000146010 and ENSMUST00000180630 were negatively correlated with the severity of IR injury. A significant positive correlation was revealed between TCONS_00010866 expression and the severity of the cardiac injury. These findings revealed the lncRNA and mRNA profiles in the heart derived EVs and provided potential targets and pathways involved in cardiac IR injury.
Collapse
Affiliation(s)
- Xinyu Ge
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Qingshu Meng
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xuan Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Jing Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaoxue Ma
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Shanshan Shi
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Mimi Li
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Fang Lin
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaoting Liang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji UniversityShanghaiChina
| | - Xin Gong
- Department of Heart FailureShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhongmin Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Institute of Stem Cell Research and Clinical TranslationShanghaiChina
| | - Wei Han
- Department of Heart FailureShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaohui Zhou
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
102
|
Dugail I, Le Lay S. Adipocyte-Derived Extracellular Vesicles: Caveolin Matters. Diabetes 2022; 71:2477-2479. [PMID: 36409791 PMCID: PMC9862523 DOI: 10.2337/dbi22-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022]
Affiliation(s)
| | - Soazig Le Lay
- Université de Nantes, CNRS, INSERM, l’Institut du Thorax, Nantes, France
- Université D’Angers, SFR ICAT, Angers, France
- Corresponding author: Soazig Le Lay,
| |
Collapse
|
103
|
Adnani L, Spinelli C, Tawil N, Rak J. Role of extracellular vesicles in cancer-specific interactions between tumour cells and the vasculature. Semin Cancer Biol 2022; 87:196-213. [PMID: 36371024 DOI: 10.1016/j.semcancer.2022.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/25/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
Cancer progression impacts and exploits the vascular system in several highly consequential ways. Among different types of vascular cells, blood cells and mediators that are engaged in these processes, endothelial cells are at the centre of the underlying circuitry, as crucial constituents of angiogenesis, angiocrine stimulation, non-angiogenic vascular growth, interactions with the coagulation system and other responses. Tumour-vascular interactions involve soluble factors, extracellular matrix molecules, cell-cell contacts, as well as extracellular vesicles (EVs) carrying assemblies of molecular effectors. Oncogenic mutations and transforming changes in the cancer cell genome, epigenome and signalling circuitry exert important and often cancer-specific influences upon pathways of tumour-vascular interactions, including the biogenesis, content, and biological activity of EVs and responses of cancer cells to them. Notably, EVs may carry and transfer bioactive, oncogenic macromolecules (oncoproteins, RNA, DNA) between tumour and vascular cells and thereby elicit unique functional changes and forms of vascular growth and remodeling. Cancer EVs influence the state of the vasculature both locally and systemically, as exemplified by cancer-associated thrombosis. EV-mediated communication pathways represent attractive targets for therapies aiming at modulation of the tumour-vascular interface (beyond angiogenesis) and could also be exploited for diagnostic purposes in cancer.
Collapse
Affiliation(s)
- Lata Adnani
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Cristiana Spinelli
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Nadim Tawil
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Janusz Rak
- McGill University and Research Institute of the McGill University Health Centre, Canada; Department of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
104
|
Shao X, Gong W, Wang Q, Wang P, Shi T, Mahmut A, Qin J, Yao Y, Yan W, Chen D, Chen X, Jiang Q, Guo B. Atrophic skeletal muscle fibre-derived small extracellular vesicle miR-690 inhibits satellite cell differentiation during ageing. J Cachexia Sarcopenia Muscle 2022; 13:3163-3180. [PMID: 36237168 PMCID: PMC9745557 DOI: 10.1002/jcsm.13106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sarcopenia is a common and progressive skeletal muscle disorder characterized by atrophic muscle fibres and contractile dysfunction. Accumulating evidence shows that the number and function of satellite cells (SCs) decline and become impaired during ageing, which may contribute to impaired regenerative capacity. A series of myokines/small extracellular vesicles (sEVs) released from muscle fibres regulate metabolism in muscle and extramuscular tissues in an autocrine/paracrine/endocrine manner during muscle atrophy. It is still unclear whether myokines/sEVs derived from muscle fibres can affect satellite cell function during ageing. METHODS Aged mice were used to investigate changes in the myogenic capacity of SCs during ageing-induced muscle atrophy. The effects of atrophic myotube-derived sEVs on satellite cell differentiation were investigated by biochemical methods and immunofluorescence staining. Small RNA sequencing was performed to identify differentially expressed sEV microRNAs (miRNAs) between the control myotubes and atrophic myotubes. The target genes of the miRNA were predicted by bioinformatics analysis and verified by luciferase activity assays. The effects of identified miRNA on the myogenic capacity of SCs in vivo were investigated by intramuscular injection of adeno-associated virus (AAV) to overexpress or silence miRNA in skeletal muscle. RESULTS Our study showed that the myogenic capacity of SCs was significantly decreased (50%, n = 6, P < 0.001) in the tibialis anterior muscle of aged mice. We showed that atrophic myotube-derived sEVs inhibited satellite cell differentiation in vitro (n = 3, P < 0.001) and in vivo (35%, n = 6, P < 0.05). We also found that miR-690 was the most highly enriched miRNA among all the screened sEV miRNAs in atrophic myotubes [Log2 (Fold Change) = 7, P < 0.001], which was verified in the atrophic muscle of aged mice (threefold, n = 6, P < 0.001) and aged men with mean age of 71 ± 5.27 years (2.8-fold, n = 10, P < 0.001). MiR-690 can inhibit myogenic capacity of SCs by targeting myocyte enhancer factor 2, including Mef2a, Mef2c and Mef2d, in vitro (n = 3, P < 0.05) and in vivo (n = 6, P < 0.05). Specific silencing of miR-690 in the muscle can promote satellite cell differentiation (n = 6, P < 0.001) and alleviate muscle atrophy in aged mice (n = 6, P < 0.001). CONCLUSIONS Our study demonstrated that atrophic muscle fibre-derived sEV miR-690 may inhibit satellite cell differentiation by targeting myocyte enhancer factor 2 during ageing.
Collapse
Affiliation(s)
- Xiaoyan Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Wang Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qianjin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Pu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Abdurahman Mahmut
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jianghui Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenjin Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Dongyang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
105
|
Liao ZZ, Ran L, Qi XY, Wang YD, Wang YY, Yang J, Liu JH, Xiao XH. Adipose endothelial cells mastering adipose tissues metabolic fate. Adipocyte 2022; 11:108-119. [PMID: 35067158 PMCID: PMC8786343 DOI: 10.1080/21623945.2022.2028372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Dynamic communication within adipose tissue depends on highly vascularized structural characteristics to maintain systemic metabolic homoeostasis. Recently, it has been noted that adipose endothelial cells (AdECs) act as essential bridges for biological information transmission between adipose-resident cells. Hence, paracrine regulators that mediate crosstalk between AdECs and adipose stromal cells were summarized. We also highlight the importance of AdECs to maintain adipocytes metabolic homoeostasis by regulating insulin sensitivity, lipid turnover and plasticity. The differential regulation of AdECs in adipose plasticity often depends on vascular density and metabolic states. Although choosing pro-angiogenic or anti-angiogenic therapies for obesity is still a matter of debate in clinical settings, the growing numbers of drugs have been confirmed to play an anti-obesity effect by affecting vascularization. Pharmacologic angiogenesis intervention has great potential as therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Zhe-Zhen Liao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Ran
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiao-Yan Qi
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
106
|
AlZaim I, Eid AH, Abd-Elrahman KS, El-Yazbi AF. Adipose Tissue Mitochondrial Dysfunction and Cardiometabolic Diseases: On the Search for Novel Molecular Targets. Biochem Pharmacol 2022; 206:115337. [DOI: 10.1016/j.bcp.2022.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
107
|
Rajput A, Varshney A, Bajaj R, Pokharkar V. Exosomes as New Generation Vehicles for Drug Delivery: Biomedical Applications and Future Perspectives. Molecules 2022; 27:7289. [PMID: 36364116 PMCID: PMC9658823 DOI: 10.3390/molecules27217289] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022] Open
Abstract
Currently, particular interest among the scientific community is focused on exploring the use of exosomes for several pharmaceutical and biomedical applications. This is due to the identification of the role of exosomes as an excellent intercellular communicator by delivering the requisite cargo comprising of functional proteins, metabolites and nucleic acids. Exosomes are the smallest extracellular vesicles (EV) with sizes ranging from 30-100 nm and are derived from endosomes. Exosomes have similar surface morphology to cells and act as a signal transduction channel between cells. They encompass different biomolecules, such as proteins, nucleic acids and lipids, thus rendering them naturally as an attractive drug delivery vehicle. Like the other advanced drug delivery systems, such as polymeric nanoparticles and liposomes to encapsulate drug substances, exosomes also gained much attention in enhancing therapeutic activity. Exosomes present many advantages, such as compatibility with living tissues, low toxicity, extended blood circulation, capability to pass contents from one cell to another, non-immunogenic and special targeting of various cells, making them an excellent therapeutic carrier. Exosome-based molecules for drug delivery are still in the early stages of research and clinical trials. The problems and clinical transition issues related to exosome-based drugs need to be overcome using advanced tools for better understanding and systemic evaluation of exosomes. In this current review, we summarize the most up-to-date knowledge about the complex biological journey of exosomes from biogenesis and secretion, isolation techniques, characterization, loading methods, pharmaceutical and therapeutic applications, challenges and future perspectives of exosomes.
Collapse
Affiliation(s)
| | | | | | - Varsha Pokharkar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharti Vidyapeeth Deemed University, Erandwane, Pune 411038, Maharashtra, India
| |
Collapse
|
108
|
Yue B, Wang H, Cai X, Wang J, Chai Z, Peng W, Shu S, Fu C, Zhong J. Adipose-Secreted Exosomes and Their Pathophysiologic Effects on Skeletal Muscle. Int J Mol Sci 2022; 23:12411. [PMID: 36293266 PMCID: PMC9604254 DOI: 10.3390/ijms232012411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 04/30/2024] Open
Abstract
Due to its prominent secretory activity, adipose tissue (AT) is now considered a major player in the crosstalk between organs, especially with skeletal muscle. In which, exosomes are effective carriers for the intercellular material transfer of a wide range of molecules that can influence a series of physiological and pathological processes in recipient cells. Considering their underlying roles, the regulatory mechanisms of adipose-secreted exosomes and their cellular crosstalk with skeletal muscle have received great attention in the field. In this review, we describe what is currently known of adipose-secreted exosomes, as well as their applications in skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Jiabo Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| | - Wei Peng
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Shi Shu
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Changqi Fu
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China
| |
Collapse
|
109
|
Brasil BB, Masaji S, Martins BT, Jiang H, Song N, Athena A S, Lucas B, François M, Wei-Jun Q, Rohit KN, Ronald KC. Apolipoprotein C3 and circulating mediators of preadipocyte proliferation in states of lipodystrophy. Mol Metab 2022; 64:101572. [PMID: 35964946 PMCID: PMC9418991 DOI: 10.1016/j.molmet.2022.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023] Open
Abstract
Adipogenesis is a complex process controlled by intrinsic and extrinsic factors that regulate preadipocyte proliferation, adipogenic capacity and maturation of metabolic function. Here we show that insulin and IGF-1 receptors are essential for mature adipocyte survival and that deletion of both IR and IGF1R specifically in fat using a tamoxifen inducible-AdipoQ-Cre (Ai-DKO) leads to rapid and severe loss of adipocytes in all depots, associated with a metabolic syndrome characterized by hypertriglyceridemia, hyperglycemia, hyperinsulinemia, fatty liver, and pancreatic beta cell proliferation. In this model, this pathological phenotype reverses over a few weeks, in large part, due to preadipocyte proliferation and adipose tissue regeneration. Incubation of preadipocytes with serum from the Ai-DKO mice in vitro stimulates cell proliferation, and this effect can be mimicked by conditioned media from liver slices of Ai-DKO mice, but not by media of cultured Ai-DKO adipocytes, indicating a hepatic origin of the growth factor. Proteomic analysis of serum reveals apolipoprotein C3 (APOC3), a protein secreted by liver, as one of the most upregulated proteins in the Ai-DKO mice. In vitro, purified and delipidated APOC3 stimulates preadipocyte proliferation, however, knockdown of hepatic APOC3 in vivo in Ai-DKO mice is not sufficient to block adipose regeneration. Thus, lipodystrophy is associated with presence of increased preadipocyte-stimulating growth factors in serum. Our study indicates that APOC3 is one contributing factor to preadipocyte proliferation, however, other still-unidentified circulating growth factors are also likely present in Ai-DKO mice. Identification of these factors may provide a new approach to regulation of adipose mass in health and disease.
Collapse
Affiliation(s)
- Brandao Bruna Brasil
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Sakaguchi Masaji
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Batista Thiago Martins
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Hu Jiang
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Dept. of Medicine, BIDMC, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Nie Song
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Schepmoes Athena A
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | | | - Moreau François
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Qian Wei-Jun
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Kulkarni N Rohit
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Dept. of Medicine, BIDMC, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Kahn C Ronald
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
110
|
Abstract
It is important to understand how different human organs coordinate and interact with each other. Since obesity and cardiac disease frequently coincide, the crosstalk between adipose tissues and heart has drawn attention. We appreciate that specific peptides/proteins, lipids, nucleic acids, and even organelles shuttle between the adipose tissues and heart. These bioactive components can profoundly affect the metabolism of cells in distal organs, including heart. Importantly, this process can be dysregulated under pathophysiological conditions. This also opens the door to efforts targeting these mediators as potential therapeutic strategies to treat patients who manifest diabetes and cardiovascular disease. Here, we summarize the recent progress toward a better understanding of how the adipose tissues and heart interact with each other.
Collapse
|
111
|
Brown Adipose Tissue Sheds Extracellular Vesicles That Carry Potential Biomarkers of Metabolic and Thermogenesis Activity Which Are Affected by High Fat Diet Intervention. Int J Mol Sci 2022; 23:ijms231810826. [PMID: 36142750 PMCID: PMC9504916 DOI: 10.3390/ijms231810826] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Brown adipose tissue (BAT) is a key target for the development of new therapies against obesity due to its role in promoting energy expenditure; BAT secretory capacity is emerging as an important contributor to systemic effects, in which BAT extracellular vesicles (EVs) (i.e., batosomes) might be protagonists. EVs have emerged as a relevant cellular communication system and carriers of disease biomarkers. Therefore, characterization of the protein cargo of batosomes might reveal their potential as biomarkers of the metabolic activity of BAT. In this study, we are the first to isolate batosomes from lean and obese Sprague–Dawley rats, and to establish reference proteome maps. An LC-SWATH/MS analysis was also performed for comparisons with EVs secreted by white adipose tissue (subcutaneous and visceral WAT), and it showed that 60% of proteins were exclusive to BAT EVs. Precisely, batosomes of lean animals contain proteins associated with mitochondria, lipid metabolism, the electron transport chain, and the beta-oxidation pathway, and their protein cargo profile is dramatically affected by high fat diet (HFD) intervention. Thus, in obesity, batosomes are enriched with proteins involved in signal transduction, cell communication, the immune response, inflammation, thermogenesis, and potential obesity biomarkers including UCP1, Glut1, MIF, and ceruloplasmin. In conclusion, the protein cargo of BAT EVs is affected by the metabolic status and contains potential biomarkers of thermogenesis activity.
Collapse
|
112
|
Paterson E, Blenkiron C, Danielson K, Henry C. Recommendations for extracellular vesicle miRNA biomarker research in the endometrial cancer context. Transl Oncol 2022; 23:101478. [PMID: 35820359 PMCID: PMC9284453 DOI: 10.1016/j.tranon.2022.101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/06/2022] [Accepted: 07/03/2022] [Indexed: 11/05/2022] Open
Abstract
Endometrial cancer (EC) is the most common gynaecological malignancy in the developed world, and concerningly incidence is rising, particularly in younger people. Therefore, there is increased interest in novel diagnostic and prognostic biomarkers. Extracellular vesicles (EVs) are membrane-bound particles present in bodily fluids that have the potential to facilitate non-invasive, early diagnosis of EC and could aid with monitoring of recurrence and treatment response. EV cargo provides molecular insight into the tumor, with the lipid bilayer providing stability for RNA species usually prone to degradation. miRNAs have recently become a focus for EV biomarker research due to their ability to regulate cancer related pathways and influence cancer development and progression. This review evaluates the current literature on EV miRNA biomarkers with a focus on EC, and discusses the challenges facing this research. This review finally highlights areas of focus for EV miRNA biomarker research going forward, such as standardization of normalization approaches, sample storage and processing, extensive reporting of methodologies and moving away from single miRNA biomarkers.
Collapse
Affiliation(s)
- Emily Paterson
- Department of Obstetrics, Gynaecology and Women's Health, University of Otago, Wellington, New Zealand
| | - Cherie Blenkiron
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kirsty Danielson
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand
| | - Claire Henry
- Department of Obstetrics, Gynaecology and Women's Health, University of Otago, Wellington, New Zealand.
| |
Collapse
|
113
|
Watanabe S, Sudo Y, Makino T, Kimura S, Tomita K, Noguchi M, Sakurai H, Shimizu M, Takahashi Y, Sato R, Yamauchi Y. Skeletal muscle releases extracellular vesicles with distinct protein and microRNA signatures that function in the muscle microenvironment. PNAS NEXUS 2022; 1:pgac173. [PMID: 36714847 PMCID: PMC9802077 DOI: 10.1093/pnasnexus/pgac173] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/24/2022] [Indexed: 05/25/2023]
Abstract
Extracellular vesicles (EVs) contain various regulatory molecules and mediate intercellular communications. Although EVs are secreted from various cell types, including skeletal muscle cells, and are present in the blood, their identity is poorly characterized in vivo, limiting the identification of their origin in the blood. Since skeletal muscle is the largest organ in the body, it could substantially contribute to circulating EVs as their source. However, due to the lack of defined markers that distinguish skeletal muscle-derived EVs (SkM-EVs) from others, whether skeletal muscle releases EVs in vivo and how much SkM-EVs account for plasma EVs remain poorly understood. In this work, we perform quantitative proteomic analyses on EVs released from C2C12 cells and human iPS cell-derived myocytes and identify potential marker proteins that mark SkM-EVs. These markers we identified apply to in vivo tracking of SkM-EVs. The results show that skeletal muscle makes only a subtle contribution to plasma EVs as their source in both control and exercise conditions in mice. On the other hand, we demonstrate that SkM-EVs are concentrated in the skeletal muscle interstitium. Furthermore, we show that interstitium EVs are highly enriched with the muscle-specific miRNAs and repress the expression of the paired box transcription factor Pax7, a master regulator for myogenesis. Taken together, our findings confirm previous studies showing that skeletal muscle cells release exosome-like EVs with specific protein and miRNA profiles in vivo and suggest that SkM-EVs mainly play a role within the muscle microenvironment where they accumulate.
Collapse
Affiliation(s)
- Sho Watanabe
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yuri Sudo
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Takumi Makino
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Satoshi Kimura
- Technology Advancement Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Kenji Tomita
- Technology Advancement Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Makoto Noguchi
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Makoto Shimizu
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Yu Takahashi
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Ryuichiro Sato
- Laboratory of Food Biochemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- Nutri-Life Science Laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | | |
Collapse
|
114
|
Nicholson RJ, Norris MK, Poss AM, Holland WL, Summers SA. The Lard Works in Mysterious Ways: Ceramides in Nutrition-Linked Chronic Disease. Annu Rev Nutr 2022; 42:115-144. [PMID: 35584813 PMCID: PMC9399075 DOI: 10.1146/annurev-nutr-062220-112920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Diet influences onset, progression, and severity of several chronic diseases, including heart failure, diabetes, steatohepatitis, and a subset of cancers. The prevalence and clinical burden of these obesity-linked diseases has risen over the past two decades. These metabolic disorders are driven by ectopic lipid deposition in tissues not suited for fat storage, leading to lipotoxic disruption of cell function and survival. Sphingolipids such as ceramides are among the most deleterious and bioactive metabolites that accrue, as they participate in selective insulin resistance, dyslipidemia, oxidative stress and apoptosis. This review discusses our current understanding of biochemical pathways controlling ceramide synthesis, production and action; influences of diet on ceramide levels; application of circulating ceramides as clinical biomarkers of metabolic disease; and molecular mechanisms linking ceramides to altered metabolism and survival of cells. Development of nutritional or pharmacological strategies to lower ceramides could have therapeutic value in a wide range of prevalent diseases.
Collapse
Affiliation(s)
- Rebekah J. Nicholson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, Utah, USA,Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, Utah, USA
| | - Marie K. Norris
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, Utah, USA,Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, Utah, USA
| | - Annelise M. Poss
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, Utah, USA,Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, Utah, USA
| | - William L. Holland
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, Utah, USA,Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, Utah, USA
| | - Scott A. Summers
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, Utah, USA,Diabetes and Metabolism Research Center, University of Utah College of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
115
|
Xiong T, Rodriguez Paris V, Edwards MC, Hu Y, Cochran BJ, Rye KA, Ledger WL, Padmanabhan V, Handelsman DJ, Gilchrist RB, Walters KA. Androgen signaling in adipose tissue, but less likely skeletal muscle, mediates development of metabolic traits in a PCOS mouse model. Am J Physiol Endocrinol Metab 2022; 323:E145-E158. [PMID: 35658542 DOI: 10.1152/ajpendo.00418.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common, multifactorial disorder characterized by endocrine, reproductive, and metabolic dysfunction. As the etiology of PCOS is unknown, there is no cure and symptom-oriented treatments are suboptimal. Hyperandrogenism is a key diagnostic trait, and evidence suggests that androgen receptor (AR)-mediated actions are critical to PCOS pathogenesis. However, the key AR target sites involved remain to be fully defined. Adipocyte and muscle dysfunction are proposed as important sites involved in the manifestation of PCOS traits. We investigated the role of AR signaling in white adipose tissue (WAT), brown adipose tissue (BAT), and skeletal muscle in the development of PCOS in a hyperandrogenic PCOS mouse model. As expected, dihydrotestosterone (DHT) exposure induced key reproductive and metabolic PCOS traits in wild-type (WT) females. Transplantation of AR-insensitive (AR-/-) WAT or BAT from AR knockout females (ARKO) into DHT-treated WT mice ameliorated some metabolic PCOS features, including increased body weight, adiposity, and adipocyte hypertrophy, but not reproductive PCOS traits. In contrast, DHT-treated ARKO female mice transplanted with AR-responsive (AR+/+) WAT or BAT continued to resist developing PCOS traits. DHT-treated skeletal muscle-specific AR knockout females (SkMARKO) displayed a comparable phenotype with that of DHT-treated WT females, with full development of PCOS traits. Taken together, these findings infer that both WAT and BAT, but less likely skeletal muscle, are key sites of AR-mediated actions involved in the experimental pathogenesis of metabolic PCOS traits. These data further support targeting adipocyte AR-driven pathways in future research aimed at developing novel therapeutic interventions for PCOS.NEW & NOTEWORTHY Hyperandrogenism is a key feature in the pathogenesis of polycystic ovary syndrome (PCOS); however, the tissue sites of androgen receptor (AR) signaling are unclear. In this study, AR signaling in white and brown adipose tissue, but less likely in skeletal muscle, was found to be involved in the development of metabolic PCOS traits, highlighting the importance of androgen actions in adipose tissue and obesity in the manifestation of metabolic disturbances.
Collapse
Affiliation(s)
- Ting Xiong
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Valentina Rodriguez Paris
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Melissa C Edwards
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Ying Hu
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Blake J Cochran
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - William L Ledger
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | | | - David J Handelsman
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Robert B Gilchrist
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Kirsty A Walters
- Fertility and Research Centre, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Andrology Laboratory, ANZAC Research Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
116
|
Dong X, Zhu S, Liu J, Dong Z, Guan F, Xu A, Zhao J, Ge J. Ameliorating mechanism of nuciferine on high-fat diet-induced dyslipidemia and hepatic steatosis by regulating intestinal absorption and serum extracellular vesicles in rats. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
117
|
Kim B, Arany Z. Endothelial Lipid Metabolism. Cold Spring Harb Perspect Med 2022; 12:a041162. [PMID: 35074792 PMCID: PMC9310950 DOI: 10.1101/cshperspect.a041162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Endothelial cells (ECs) line all vessels of all vertebrates and are fundamental to organismal metabolism. ECs rely on their metabolism both to transport nutrients in and out of underlying parenchyma, and to support their own cellular activities, including angiogenesis. ECs primarily consume glucose, and much is known of how ECs transport and consume glucose and other carbohydrates. In contrast, how lipids are transported, and the role of lipids in normal EC function, has garnered less attention. We review here recent developments on the role of lipids in endothelial metabolism, with a focus on lipid uptake and transport in quiescent endothelium, and the use of lipid pathways during angiogenesis.
Collapse
Affiliation(s)
- Boa Kim
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Zolt Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
118
|
Endothelial Nitric Oxide Synthase in the Perivascular Adipose Tissue. Biomedicines 2022; 10:biomedicines10071754. [PMID: 35885059 PMCID: PMC9313312 DOI: 10.3390/biomedicines10071754] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 01/08/2023] Open
Abstract
Perivascular adipose tissue (PVAT) is a special type of ectopic fat depot that adheres to most vasculatures. PVAT has been shown to exert anticontractile effects on the blood vessels and confers protective effects against metabolic and cardiovascular diseases. PVAT plays a critical role in vascular homeostasis via secreting adipokine, hormones, and growth factors. Endothelial nitric oxide synthase (eNOS; also known as NOS3 or NOSIII) is well-known for its role in the generation of vasoprotective nitric oxide (NO). eNOS is primarily expressed, but not exclusively, in endothelial cells, while recent studies have identified its expression in both adipocytes and endothelial cells of PVAT. PVAT eNOS is an important player in the protective role of PVAT. Different studies have demonstrated that, under obesity-linked metabolic diseases, PVAT eNOS may be even more important than endothelium eNOS in obesity-induced vascular dysfunction, which may be attributed to certain PVAT eNOS-specific functions. In this review, we summarized the current understanding of eNOS expression in PVAT, its function under both physiological and pathological conditions and listed out a few pharmacological interventions of interest that target eNOS in PVAT.
Collapse
|
119
|
Abstract
While most tissues exhibit their greatest growth during development, adipose tissue is capable of additional massive expansion in adults. Adipose tissue expandability is advantageous when temporarily storing fuel for use during fasting, but becomes pathological upon continuous food intake, leading to obesity and its many comorbidities. The dense vasculature of adipose tissue provides necessary oxygen and nutrients, and supports delivery of fuel to and from adipocytes under fed or fasting conditions. Moreover, the vasculature of adipose tissue comprises a major niche for multipotent progenitor cells, which give rise to new adipocytes and are necessary for tissue repair. Given the multiple, pivotal roles of the adipose tissue vasculature, impairments in angiogenic capacity may underlie obesity-associated diseases such as diabetes and cardiometabolic disease. Exciting new studies on the single-cell and single-nuclei composition of adipose tissues in mouse and humans are providing new insights into mechanisms of adipose tissue angiogenesis. Moreover, new modes of intercellular communication involving micro vesicle and exosome transfer of proteins, nucleic acids and organelles are also being recognized to play key roles. This review focuses on new insights on the cellular and signaling mechanisms underlying adipose tissue angiogenesis, and on their impact on obesity and its pathophysiological consequences.
Collapse
|
120
|
Bie N, Yong T, Wei Z, Gan L, Yang X. Extracellular vesicles for improved tumor accumulation and penetration. Adv Drug Deliv Rev 2022; 188:114450. [PMID: 35841955 DOI: 10.1016/j.addr.2022.114450] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/26/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs), including microparticles and exosomes, have emerged as potential tools for tumor targeting delivery during the past years. Recently, mass of strategies are applied to assist EVs to accumulate and penetrate into deep tumor sites. In this review, EVs from different cells with unique innate characters and engineered approaches (e.g. chemical engineering, genetical engineering and biomimetic engineering) as drug delivery systems to enhance tumor accumulation and penetration are summarized. Meanwhile, efficient biological function modulation (e.g. extracellular matrix degradation, mechanical property regulation and transcytosis) is introduced to facilitate tumor accumulation and penetration of EVs. Finally, the prospects and challenges on further clinical applications of EVs are discussed.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
121
|
Chan W, Chow FWN, Tsang CC, Liu X, Yao W, Chan TTY, Siu GKH, Ho AYM, Luk KS, Lau SKP, Woo PCY. Induction of amphotericin B resistance in susceptible Candida auris by extracellular vesicles. Emerg Microbes Infect 2022; 11:1900-1909. [PMID: 35786393 PMCID: PMC9341352 DOI: 10.1080/22221751.2022.2098058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Drug resistance derived from extracellular vesicles (EVs) is an increasingly important research area but has seldom been described regarding fungal pathogens. Here, we characterized EVs derived from a triazole-resistant but amphotericin B-susceptible strain of Candida auris. Nano- to microgram concentrations of C. auris EVs prepared from both broth and solid agar cultures could robustly increase the yeast’s survival against both pure and clinical amphotericin B formulations in a dose-dependent manner, resulting in up to 16-fold changes of minimum inhibitory concentration. Meanwhile, this effect was not observed upon addition of these EVs to C. albicans, nor upon addition of C. albicans EVs to C. auris. No change in susceptibilities was observed upon EV treatment for fluconazole, voriconazole, micafungin, and flucytosine. Mass spectrometry indicated the presence of immunogenic-/drug resistance-implicated proteins in C. auris EVs, including alcohol dehydrogenase 1 as well as C. albicans Mp65-like and Xog1-like proteins in high quantities. Based on these observations, we propose a potential species-specific role for EVs in amphotericin B resistance in C. auris. These observations may provide critical insights into treatment of multidrug-resistant C. auris.
Collapse
Affiliation(s)
- Walton Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Franklin Wang-Ngai Chow
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hunghom, Hong Kong
| | - Chi-Ching Tsang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,School of Medical and Health Sciences, Tung Wah College, Homantin, Hong Kong
| | - Xueyan Liu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Weiming Yao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tony Tat-Yin Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Gilman Kit-Hang Siu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hunghom, Hong Kong
| | - Alex Yat-Man Ho
- Department of Pathology, Princess Margaret Hospital, Kwai Chung, Hong Kong
| | - Kristine Shik Luk
- Department of Pathology, Princess Margaret Hospital, Kwai Chung, Hong Kong
| | - Susanna Kar-Pui Lau
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Patrick Chiu-Yat Woo
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,PhD Program in Translational Medicine and Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.,The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
122
|
Yang X, Zheng E, Chatterjee V, Ma Y, Reynolds A, Villalba N, Wu MH, Yuan SY. Protein palmitoylation regulates extracellular vesicle production and function in sepsis. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e50. [PMID: 38419739 PMCID: PMC10901530 DOI: 10.1002/jex2.50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/16/2022] [Accepted: 06/19/2022] [Indexed: 03/02/2024]
Abstract
Extracellular vesicles (EVs) are bioactive membrane-encapsulated particles generated by a series of events involving membrane budding, fission and fusion. Palmitoylation, mediated by DHHC palmitoyl acyltransferases, is a lipidation reaction that increases protein lipophilicity and membrane localization. Here, we report palmitoylation as a novel regulator of EV formation and function during sepsis. Our results showed significantly decreased circulating EVs in mice with DHHC21 functional deficiency (Zdhhc21dep/dep), compared to wild-type (WT) mice 24 h after septic injury. Furthermore, WT and Zdhhc21dep/dep EVs displayed distinct palmitoyl-proteomic profiles. Ingenuity pathway analysis indicated that sepsis altered several inflammation related pathways expressed in EVs, among which the most significantly activated was the complement pathway; however, this sepsis-induced complement enrichment in EVs was greatly blunted in Zdhhc21dep/dep EVs. Functionally, EVs isolated from WT mice with sepsis promoted neutrophil adhesion, transmigration, and neutrophil extracellular trap production; these effects were significantly attenuated by DHHC21 loss-of-function. Furthermore, Zdhhc21dep/dep mice displayed reduced neutrophil infiltration in lungs and improved survival after CLP challenges. These findings indicate that blocking palmitoylation via DHHC21 functional deficiency can reduce sepsis-stimulated production of complement-enriched EVs and attenuates their effects on neutrophil activity.
Collapse
Affiliation(s)
- Xiaoyuan Yang
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Ethan Zheng
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Victor Chatterjee
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yonggang Ma
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Amanda Reynolds
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Nuria Villalba
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Mack H. Wu
- Department of SurgeryUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Sarah Y. Yuan
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
- Department of SurgeryUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| |
Collapse
|
123
|
Howard J, Wynne K, Moldenhauer E, Clarke P, Maguire C, Bollard S, Yin X, Brennan L, Mooney L, Fitzsimons S, Halasz M, Aluri ER, Brougham DF, Kolch W, Dwyer RM, Potter S, Kelly P, McCann A. A comparative analysis of extracellular vesicles (EVs) from human and feline plasma. Sci Rep 2022; 12:10851. [PMID: 35761023 PMCID: PMC9237114 DOI: 10.1038/s41598-022-14211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles found in all biological fluids, capable of transporting biological material around the body. Extensive research into the physiological role of EVs has led to the development of the Minimal Information for Studies of Extracellular Vesicles (MISEV) framework in 2018. This framework guides the standardisation of protocols in the EV field. To date, the focus has been on EVs of human origin. As comparative medicine progresses, there has been a drive to study similarities between diseases in humans and animals. To successfully research EVs in felines, we must validate the application of the MISEV guidelines in this group. EVs were isolated from the plasma of healthy humans and felines. EV characterisation was carried out according to the MISEV guidelines. Human and feline plasma showed a similar concentration of EVs, comparable expression of known EV markers and analogous particle to protein ratios. Mass spectrometry analyses showed that the proteomic signature of EVs from humans and felines were similar. Asymmetrical flow field flow fractionation, showed two distinct subpopulations of EVs isolated from human plasma, whereas only one subpopulation was isolated from feline plasma. Metabolomic profiling showed similar profiles for humans and felines. In conclusion, isolation, and characterisation of EVs from humans and felines show that MISEV2018 guidelines may also be applied to felines. Potential comparative medicine studies of EVs may provide a model for studying naturally occurring diseases in both humans and felines.
Collapse
Affiliation(s)
- Jane Howard
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin 4, Ireland. .,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Kieran Wynne
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.,Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Paul Clarke
- Postnova Analytics UK Ltd, Malvern Hills Science Park, Malvern, WR14 3SZ, Worcestershire, UK
| | - Ciaran Maguire
- Particular Sciences Ltd, Unit 2 Birch House, Rosemount Business Park, Ballycoolin, Dublin 11, Ireland
| | - Stephanie Bollard
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.,Department of Plastic & Reconstructive Surgery, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Xiaofei Yin
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lorraine Brennan
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Louise Mooney
- College of Health and Agricultural Sciences, UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Sciences, UCD Conway Institute, Belfield, Dublin 4, Ireland
| | - Melinda Halasz
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.,Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ester Rani Aluri
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dermot F Brougham
- UCD School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | - Róisín M Dwyer
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, H91 V4AY, Ireland
| | - Shirley Potter
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin 4, Ireland.,Department of Plastic & Reconstructive Surgery, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Pamela Kelly
- College of Health and Agricultural Sciences, UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Amanda McCann
- UCD School of Medicine, College of Health and Agricultural Sciences, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
124
|
Zhao X, Yang X, An Z, Liu L, Yong J, Xing H, Huang R, Tian J, Song X. Pathophysiology and molecular mechanism of caveolin involved in myocardial protection strategies in ischemic conditioning. Biomed Pharmacother 2022; 153:113282. [PMID: 35750009 DOI: 10.1016/j.biopha.2022.113282] [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: 04/27/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022] Open
Abstract
Multiple pathophysiological pathways are activated during the process of myocardial injury. Various cardioprotective strategies protect the myocardium from ischemia, infarction, and ischemia/reperfusion (I/R) injury through different targets, yet the clinical translation remains limited. Caveolae and its structure protein, caveolins, have been suggested as a bridge to transmit damage-preventing signals and mediate the protection of ultrastructure in cardiomyocytes under pathological conditions. In this review, we first briefly introduce caveolae and caveolins. Then we review the cardioprotective strategies mediated by caveolins through various pathophysiological pathways. Finally, some possible research directions are proposed to provide future experiments and clinical translation perspectives targeting caveolin based on the investigative evidence.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Jingwen Yong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Haoran Xing
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, 95th Yong An Road, Xuan Wu District, Beijing 100050, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| |
Collapse
|
125
|
Crewe C. The challenges of interrogating adipose tissue extracellular vesicle functions in physiology. Commun Biol 2022; 5:581. [PMID: 35701664 PMCID: PMC9197966 DOI: 10.1038/s42003-022-03511-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
Recent developments in adipose tissue-derived extracellular vesicle (EV) research is highlighted, assessing current adipose tissue EV research strategies and obstacles the field faces.
Collapse
Affiliation(s)
- Clair Crewe
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
126
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
127
|
Zocchi M, Della Porta M, Lombardoni F, Scrimieri R, Zuccotti GV, Maier JA, Cazzola R. A Potential Interplay between HDLs and Adiponectin in Promoting Endothelial Dysfunction in Obesity. Biomedicines 2022; 10:1344. [PMID: 35740366 PMCID: PMC9220412 DOI: 10.3390/biomedicines10061344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 12/11/2022] Open
Abstract
Obesity is an epidemic public health problem that has progressively worsened in recent decades and is associated with low-grade chronic inflammation (LGCI) in metabolic tissues and an increased risk of several diseases. In particular, LGCI alters metabolism and increases cardiovascular risk by impairing endothelial function and altering the functions of adiponectin and high-density lipoproteins (HDLs). Adiponectin is an adipokine involved in regulating energy metabolism and body composition. Serum adiponectin levels are reduced in obese individuals and negatively correlate with chronic sub-clinical inflammatory markers. HDLs are a heterogeneous and complex class of lipoproteins that can be dysfunctional in obesity. Adiponectin and HDLs are strictly interdependent, and the maintenance of their interplay is essential for vascular function. Since such a complex network of interactions is still overlooked in clinical settings, this review aims to highlight the mechanisms involved in the impairment of the HDLs/adiponectin axis in obese patients to predict the risk of cardiovascular diseases and activate preventive countermeasures. Here, we provide a narrative review of the role of LGCI in altering HDLs, adiponectin and endothelial functions in obesity to encourage new studies about their synergic effects on cardiovascular health and disease.
Collapse
Affiliation(s)
- Monica Zocchi
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| | - Matteo Della Porta
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| | - Federico Lombardoni
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| | - Roberta Scrimieri
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
- Department of Pediatrics, Ospedale dei Bambini, 20154 Milan, Italy
| | - Jeanette A. Maier
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| | - Roberta Cazzola
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy; (M.Z.); (M.D.P.); (F.L.); (R.S.); (G.V.Z.); (J.A.M.)
| |
Collapse
|
128
|
Delgadillo-Velázquez J, Mendivil-Alvarado H, Coronado-Alvarado CD, Astiazaran-Garcia H. Extracellular Vesicles from Adipose Tissue Could Promote Metabolic Adaptation through PI3K/Akt/mTOR. Cells 2022; 11:cells11111831. [PMID: 35681526 PMCID: PMC9180692 DOI: 10.3390/cells11111831] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 01/25/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles secreted by cells under physiological and pathological conditions, such as metabolic diseases. In this context, EVs are considered potential key mediators in the physiopathology of obesity. It has been reported that EVs derived from adipose tissue (ADEVs) contribute to the development of a local inflammatory response that leads to adipose tissue dysfunction. In addition, it has been proposed that EVs are associated with the onset and progression of several obesity-related metabolic diseases such as insulin resistance. In particular, characterizing the molecular fingerprint of obesity-related ADEVs can provide a bigger picture that better reflects metabolic adaptation though PI3K/Akt/mTOR. Hence, in this review we describe the possible crosstalk communication of ADEVs with metabolically active organs and the intracellular response in the insulin signaling pathway.
Collapse
Affiliation(s)
- Jaime Delgadillo-Velázquez
- Coordination of Nutrition, Research Center for Food and Development (CIAD), Ave. Gustavo E. Astiazarán #46, Hermosillo 83304, Mexico; (J.D.-V.); (H.M.-A.); (C.D.C.-A.)
| | - Herminia Mendivil-Alvarado
- Coordination of Nutrition, Research Center for Food and Development (CIAD), Ave. Gustavo E. Astiazarán #46, Hermosillo 83304, Mexico; (J.D.-V.); (H.M.-A.); (C.D.C.-A.)
| | - Carlos Daniel Coronado-Alvarado
- Coordination of Nutrition, Research Center for Food and Development (CIAD), Ave. Gustavo E. Astiazarán #46, Hermosillo 83304, Mexico; (J.D.-V.); (H.M.-A.); (C.D.C.-A.)
| | - Humberto Astiazaran-Garcia
- Coordination of Nutrition, Research Center for Food and Development (CIAD), Ave. Gustavo E. Astiazarán #46, Hermosillo 83304, Mexico; (J.D.-V.); (H.M.-A.); (C.D.C.-A.)
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora, Hermosillo 83000, Mexico
- Correspondence: ; Tel.: +52-662-1029-701
| |
Collapse
|
129
|
Fridman ES, Ginini L, Gil Z. The Role of Extracellular Vesicles in Metabolic Reprogramming of the Tumor Microenvironment. Cells 2022; 11:cells11091433. [PMID: 35563739 PMCID: PMC9104192 DOI: 10.3390/cells11091433] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment (TME) includes a network of cancerous and non-cancerous cells, together with associated blood vessels, the extracellular matrix, and signaling molecules. The TME contributes to cancer progression during various phases of tumorigenesis, and interactions that take place within the TME have become targets of focus in cancer therapy development. Extracellular vesicles (EVs) are known to be conveyors of genetic material, proteins, and lipids within the TME. One of the hallmarks of cancer is its ability to reprogram metabolism to sustain cell growth and proliferation in a stringent environment. In this review, we provide an overview of TME EV involvement in the metabolic reprogramming of cancer and stromal cells, which favors cancer progression by enhancing angiogenesis, proliferation, metastasis, treatment resistance, and immunoevasion. Targeting the communication mechanisms and systems utilized by TME-EVs is opening a new frontier in cancer therapy.
Collapse
Affiliation(s)
- Eran S. Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion—Israel Institute of Technology, Haifa 31096, Israel; (E.S.F.); (L.G.)
| | - Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion—Israel Institute of Technology, Haifa 31096, Israel; (E.S.F.); (L.G.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel
- Correspondence: ; Tel.: +972-4-854-2480
| |
Collapse
|
130
|
Zhao H, Chen X, Hu G, Li C, Guo L, Zhang L, Sun F, Xia Y, Yan W, Cui Z, Guo Y, Guo X, Huang C, Fan M, Wang S, Zhang F, Tao L. Small Extracellular Vesicles From Brown Adipose Tissue Mediate Exercise Cardioprotection. Circ Res 2022; 130:1490-1506. [PMID: 35387487 DOI: 10.1161/circresaha.121.320458] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Long-term exercise provides reliable cardioprotection via mechanisms still incompletely understood. Although traditionally considered a thermogenic tissue, brown adipose tissue (BAT) communicates with remote organs (eg, the heart) through its endocrine function. BAT expands in response to exercise, but its involvement in exercise cardioprotection remains undefined. OBJECTIVE This study investigated whether small extracellular vesicles (sEVs) secreted by BAT and their contained microRNAs (miRNAs) regulate cardiomyocyte survival and participate in exercise cardioprotection in the context of myocardial ischemia/reperfusion (MI/R) injury. METHODS AND RESULTS Four weeks of exercise resulted in a significant BAT expansion in mice. Surgical BAT ablation before MI/R weakened the salutary effects of exercise. Adeno-associated virus 9 vectors carrying short hairpin RNA targeting Rab27a (a GTPase required for sEV secretion) or control viruses were injected in situ into the interscapular BAT. Exercise-mediated protection against MI/R injury was greatly attenuated in mice whose BAT sEV secretion was suppressed by Rab27a silencing. Intramyocardial injection of the BAT sEVs ameliorated MI/R injury, revealing the cardioprotective potential of BAT sEVs. Discovery-driven experiments identified miR-125b-5p, miR-128-3p, and miR-30d-5p (referred to as the BAT miRNAs) as essential BAT sEV components for mediating cardioprotection. BAT-specific inhibition of the BAT miRNAs prevented their upregulation in plasma sEVs and hearts of exercised mice and attenuated exercise cardioprotection. Mechanistically, the BAT miRNAs cooperatively suppressed the proapoptotic MAPK (mitogen-associated protein kinase) pathway by targeting a series of molecules (eg, Map3k5, Map2k7, and Map2k4) in the signaling cascade. Delivery of BAT sEVs into hearts or cardiomyocytes suppressed MI/R-related MAPK pathway activation, an effect that disappeared with the combined use of the BAT miRNA inhibitors. CONCLUSIONS The sEVs secreted by BAT participate in exercise cardioprotection via delivering the cardioprotective miRNAs into the heart. These results provide novel insights into the mechanisms underlying the BAT-cardiomyocyte interaction and highlight BAT sEVs and their contained miRNAs as alternative candidates for exercise cardioprotection.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Pharmacy, the 960th Hospital of the Logistics Support Force, Jinan, China (H.Z.)
| | - Xiyao Chen
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Department of Geriatrics, The Fourth Military Medical University, Xi'an, China. (X.C.)
| | - Guangyu Hu
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Congye Li
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Lanyan Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fangfang Sun
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yunlong Xia
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Wenjun Yan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ze Cui
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Yongzhen Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.).,Xijing Hospital and Department of Toxicology, School of Public Health, The Fourth Military Medical University, Xi'an, China. (Y.G.)
| | - Xiong Guo
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Chong Huang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Miaomiao Fan
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Shan Wang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Fuyang Zhang
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| | - Ling Tao
- Department of Cardiology, The Fourth Military Medical University, Xi'an, China. (H.Z., X.C., G.H., C.L., L.G., L.Z., F.S., Y.X., W.Y., Z.C., Y.G., X.G., C.H., M.F., S.W., F.Z., L.T.)
| |
Collapse
|
131
|
Ioannidou A, Fisher RM, Hagberg CE. The multifaceted roles of the adipose tissue vasculature. Obes Rev 2022; 23:e13403. [PMID: 34866318 DOI: 10.1111/obr.13403] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 12/30/2022]
Abstract
The prevalence of obesity and its associated pathologies continue to increase, which has led to a renewed interest in our major weight-regulating organ, the white adipose tissue. It has become clear that its development, expansion, and physiological function depend on proper crosstalk between each of its cellular constituents, with a central role for the vascular endothelium lining the blood vessels. Although first considered a mere barrier, the endothelium has emerged as a dynamic unit modulating many critical adipose tissue functions. It not only oversees the uptake of all nutrients to be stored in the adipocytes but also provides an important growth niche for adipocyte progenitors and regulates the expandability of the tissue during overfeeding and obesity. In this review, we describe the reciprocal relationship between endothelial cells, adipocytes, and obesity. We present recent studies that support an important role for endothelial cells as central mediators of many of the physiological and pathological functions of the adipose tissue and highlight several unknown aspects of adipose tissue vascular biology. This new perspective could present exciting opportunities to develop new therapeutic approaches against obesity-related pathologies and is thus of great interest in our increasingly obese society.
Collapse
Affiliation(s)
- Anna Ioannidou
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Rachel M Fisher
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Solna, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
132
|
Shen DD, Pang JR, Bi YP, Zhao LF, Li YR, Zhao LJ, Gao Y, Wang B, Wang N, Wei L, Guo H, Liu HM, Zheng YC. LSD1 deletion decreases exosomal PD-L1 and restores T-cell response in gastric cancer. Mol Cancer 2022; 21:75. [PMID: 35296335 PMCID: PMC8925194 DOI: 10.1186/s12943-022-01557-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/01/2022] [Indexed: 12/20/2022] Open
Abstract
Background Histone lysine-specific demethylase 1 (LSD1) expression has been shown to be significantly elevated in gastric cancer (GC) and may be associated with the proliferation and metastasis of GC. It has been reported that LSD1 repressed tumor immunity through programmed cell death 1 ligand 1 (PD-L1) in melanoma and breast cancer. The role of LSD1 in the immune microenvironment of GC is unknown. Methods Expression LSD1 and PD-L1 in GC patients was analyzed by immunohistochemical (IHC) and Western blotting. Exosomes were isolated from the culture medium of GC cells using an ultracentrifugation method and characterized by transmission electronic microscopy (TEM), nanoparticle tracking analysis (NTA), sucrose gradient centrifugation, and Western blotting. The role of exosomal PD-L1 in T-cell dysfunction was assessed by flow cytometry, T-cell killing and enzyme-linked immunosorbent assay (ELISA). Results Through in vivo exploration, mouse forestomach carcinoma (MFC) cells with LSD1 knockout (KO) showed significantly slow growth in 615 mice than T-cell-deficient BALB/c nude mice. Meanwhile, in GC specimens, expression of LSD1 was negatively correlated with that of CD8 and positively correlated with that of PD-L1. Further study showed that LSD1 inhibited the response of T cells in the microenvironment of GC by inducing the accumulation of PD-L1 in exosomes, while the membrane PD-L1 stayed constant in GC cells. Using exosomes as vehicles, LSD1 also obstructed T-cell response of other cancer cells while LSD1 deletion rescued T-cell function. It was found that while relying on the existence of LSD1 in donor cells, exosomes can regulate MFC cells proliferation with distinct roles depending on exosomal PD-L1-mediated T-cell immunity in vivo. Conclusion LSD1 deletion decreases exosomal PD-L1 and restores T-cell response in GC; this finding indicates a new mechanism with which LSD1 may regulate cancer immunity in GC and provides a new target for immunotherapy against GC. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-022-01557-1.
Collapse
Affiliation(s)
- Dan-Dan Shen
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Jing-Ru Pang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Ya-Ping Bi
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Long-Fei Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Yin-Rui Li
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Li-Juan Zhao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Ya Gao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Bo Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China
| | - Ning Wang
- The School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Hebei, China
| | - Huiqin Guo
- Thoracic Department, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China.
| | - Yi-Chao Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Henan, 450052, Zhengzhou, China. .,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Academy of Medical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China.
| |
Collapse
|
133
|
Bond ST, Calkin AC, Drew BG. Adipose-Derived Extracellular Vesicles: Systemic Messengers and Metabolic Regulators in Health and Disease. Front Physiol 2022; 13:837001. [PMID: 35283789 PMCID: PMC8905439 DOI: 10.3389/fphys.2022.837001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is comprised of a heterogeneous population of cells that co-operate to perform diverse physiological roles including endocrine-related functions. The endocrine role of adipose tissue enables it to communicate nutritional and health cues to other organs, such as the liver, muscle, and brain, in order to regulate appetite and whole body metabolism. Adipose tissue dysfunction, which is often observed in obesity, is associated with changes in the adipose secretome, which can subsequently contribute to disease pathology. Indeed, secreted bioactive factors released from adipose tissue contribute to metabolic homeostasis and likely play a causal role in disease; however, what constitutes the entirety of the adipose tissue secretome is still poorly understood. Recent advances in nanotechnology have advanced this field substantially and have led to the identification of small, secreted particles known as extracellular vesicles (EVs). These small nano-sized lipid envelopes are released by most cell types and are capable of systemically delivering bioactive molecules, such as nucleic acids, proteins, and lipids. EVs interact with target cells to deliver specific cargo that can then elicit effects in various tissues throughout the body. Adipose tissue has recently been shown to secrete EVs that can communicate with the periphery to maintain metabolic homeostasis, or under certain pathological conditions, drive disease. In this review, we discuss the current landscape of adipose tissue-derived EVs, with a focus on their role in the regulation of metabolic homeostasis and disease pathology.
Collapse
Affiliation(s)
- Simon T Bond
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
134
|
He C, Dai M, Zhou X, Long J, Tian W, Yu M. Comparison of two cell-free therapeutics derived from adipose tissue: small extracellular vesicles versus conditioned medium. Stem Cell Res Ther 2022; 13:86. [PMID: 35241142 PMCID: PMC8895642 DOI: 10.1186/s13287-022-02757-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cell-free therapy has been inspired as a promising approach to overcome the limitations of traditional stem cell therapy. However, the therapeutic effect between extracellular vesicles and conditioned medium with the same source had not been compared. Our previous studies have shown that both the conditioned medium of adipose tissue (adipose tissue extract, ATE) and its further purification product small extracellular vesicles (sEV-AT) contributed to adipose tissue regeneration. In this study, we aimed to compare the ATE and sEV-AT in composition, inductivity on cells and de novo adipose regenerative potential. METHODS The characteristics of sEV-AT and ATE were compared through protein and particle yield, particle size distribution and composition. The inductivity of sEV-AT and ATE on cells were compared through co-culture of sEV-AT or ATE with ASC, HUVEC and RAW264.7 in vitro. The capacity of promoting de novo adipogenesis was compared by implanting the silicone tube containing sEV-AT or ATE subcutaneously in vivo. RESULTS More particles and concentrated particle size distribution were detected in sEV-AT. In turn, more soluble factors and multiple peaks in particle size distribution were detected in ATE. In 1662 common proteins of sEV-AT and ATE, there were 984 (59.2%) proteins enriched twice more in sEV-AT than in ATE. With the prerequisite of equivalent protein concentration, sEV-AT outperformed ATE in promoting proliferation, migration and regeneration potential of cells those contributing adipose tissue regeneration in vitro. Furthermore, sEV-AT expedited the de novo adipose tissue regeneration and angiogenesis at the early stage than ATE in vivo, but sEV-AT and ATE group formed similar neoadipose tissue and new vessels at week 12. CONCLUSIONS Our results provided a direct comparison between EV and conditioned medium as cell-free therapeutic strategy. Both sEV and ATE had specific biological signature to facilitate tissue repair. Considering the convenience of extraction and acceptable effect, ATE represented a feasible product of cell-free therapy, providing another option for different situations in clinical application. Furthermore, the complex contents of both sEV-AT and ATE should be studied comprehensively to avoid possible negative effects and to ensure sufficient safety for clinical applications.
Collapse
Affiliation(s)
- Chuan He
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China School of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Minjia Dai
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Department of Stomatology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojie Zhou
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China School of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Long
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Weidong Tian
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China.,Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China School of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei Yu
- State Key Laboratory of Oral Disease and National Clinical Research Center for Oral Diseases and National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, China. .,Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China School of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
135
|
Abstract
Healthy white adipose tissue is dependent on proliferation of endothelial cells to maintain homeostasis or undergo expansion. A new study shows that endothelial cells communicate with adipocytes via polyamines to promote vascularization of adipose tissue, thereby reversing the metabolic effects of obesity.
Collapse
Affiliation(s)
- Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
136
|
Monelli E, Villacampa P, Zabala-Letona A, Martinez-Romero A, Llena J, Beiroa D, Gouveia L, Chivite I, Zagmutt S, Gama-Perez P, Osorio-Conles O, Muixi L, Martinez-Gonzalez A, Castillo SD, Martín-Martín N, Castel P, Valcarcel-Jimenez L, Garcia-Gonzalez I, Villena JA, Fernandez-Ruiz S, Serra D, Herrero L, Benedito R, Garcia-Roves P, Vidal J, Cohen P, Nogueiras R, Claret M, Carracedo A, Graupera M. Angiocrine polyamine production regulates adiposity. Nat Metab 2022; 4:327-343. [PMID: 35288722 DOI: 10.1038/s42255-022-00544-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Reciprocal interactions between endothelial cells (ECs) and adipocytes are fundamental to maintain white adipose tissue (WAT) homeostasis, as illustrated by the activation of angiogenesis upon WAT expansion, a process that is impaired in obesity. However, the molecular mechanisms underlying the crosstalk between ECs and adipocytes remain poorly understood. Here, we show that local production of polyamines in ECs stimulates adipocyte lipolysis and regulates WAT homeostasis in mice. We promote enhanced cell-autonomous angiogenesis by deleting Pten in the murine endothelium. Endothelial Pten loss leads to a WAT-selective phenotype, characterized by reduced body weight and adiposity in pathophysiological conditions. This phenotype stems from enhanced fatty acid β-oxidation in ECs concomitant with a paracrine lipolytic action on adipocytes, accounting for reduced adiposity. Combined analysis of murine models, isolated ECs and human specimens reveals that WAT lipolysis is mediated by mTORC1-dependent production of polyamines by ECs. Our results indicate that angiocrine metabolic signals are important for WAT homeostasis and organismal metabolism.
Collapse
Affiliation(s)
- Erika Monelli
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Pilar Villacampa
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Amaia Zabala-Letona
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Anabel Martinez-Romero
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Judith Llena
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Daniel Beiroa
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Leonor Gouveia
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Iñigo Chivite
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Pau Gama-Perez
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Oscar Osorio-Conles
- Department of Endocrinology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laia Muixi
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Ainara Martinez-Gonzalez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Sandra D Castillo
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Traslational prostate cancer Research lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Pau Castel
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lorea Valcarcel-Jimenez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Josep A Villena
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sonia Fernandez-Ruiz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Dolors Serra
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Laura Herrero
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Pablo Garcia-Roves
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Josep Vidal
- Department of Endocrinology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Rubén Nogueiras
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Galician Agency of Investigation, Xunta de Galicia, La Coruña, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Traslational prostate cancer Research lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
- Ikerbasque; Basque Foundation for Science, Bilbao, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
137
|
Malnutrition and Biomarkers: A Journey through Extracellular Vesicles. Nutrients 2022; 14:nu14051002. [PMID: 35267977 PMCID: PMC8912428 DOI: 10.3390/nu14051002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles (EVs) have been identified as active components in cellular communication, which are easily altered both morphologically and chemically by the cellular environment and metabolic state of the body. Due to this sensitivity to the conditions of the cellular microenvironment, EVs have been found to be associated with disease conditions, including those associated with obesity and undernutrition. The sensitivity that EVs show to changes in the cellular microenvironment could be a reflection of early cellular alterations related to conditions of malnutrition, which could eventually be used in the routine monitoring and control of diseases or complications associated with it. However, little is known about the influence of malnutrition alone; that is, without the influence of additional diseases on the heterogeneity and specific content of EVs. To date, studies in “apparently healthy” obese patients show that there are changes in the size, quantity, and content of EVs, as well as correlations with some metabolic parameters (glucose, insulin, and serum lipids) in comparison with non-obese individuals. In light of these changes, a direct participation of EVs in the development of metabolic and cardiovascular complications in obese subjects is thought to exist. However, the mechanisms through which this process might occur are not yet fully understood. The evidence on EVs in conditions of undernutrition is limited, but it suggests that EVs play a role in the maintenance of homeostasis and muscle repair. A better understanding of how EVs participate in or promote cellular signaling in malnutrition conditions could help in the development of new strategies to treat them and their comorbidities.
Collapse
|
138
|
Jiang P, Ma X, Han S, Ma L, Ai J, Wu L, Zhang Y, Xiao H, Tian M, Tao WA, Zhang S, Chai R. Characterization of the microRNA transcriptomes and proteomics of cochlear tissue-derived small extracellular vesicles from mice of different ages after birth. Cell Mol Life Sci 2022; 79:154. [PMID: 35218422 PMCID: PMC11072265 DOI: 10.1007/s00018-022-04164-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/30/2021] [Accepted: 01/23/2022] [Indexed: 12/22/2022]
Abstract
The cochlea is an important sensory organ for both balance and sound perception, and the formation of the cochlea is a complex developmental process. The development of the mouse cochlea begins on embryonic day (E)9 and continues until postnatal day (P)21 when the hearing system is considered mature. Small extracellular vesicles (sEVs), with a diameter ranging from 30 to 200 nm, have been considered a significant medium for information communication in both physiological and pathological processes. However, there are no studies exploring the role of sEVs in the development of the cochlea. Here, we isolated tissue-derived sEVs from the cochleae of FVB mice at P3, P7, P14, and P21 by ultracentrifugation. These sEVs were first characterized by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Next, we used small RNA-seq and mass spectrometry to characterize the microRNA transcriptomes and proteomes of cochlear sEVs from mice at different ages. Many microRNAs and proteins were discovered to be related to inner ear development, anatomical structure development, and auditory nervous system development. These results all suggest that sEVs exist in the cochlea and are likely to be essential for the normal development of the auditory system. Our findings provide many sEV microRNA and protein targets for future studies of the roles of cochlear sEVs.
Collapse
Affiliation(s)
- Pei Jiang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xiangyu Ma
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shanying Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Leyao Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingru Ai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Leilei Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yuan Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hairong Xiao
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Mengyao Tian
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - W Andy Tao
- Department of Chemistry, Department of Biochemistry, Purdue University, West Lafayette, Indiana, 47907, USA.
- Center for Cancer Research, Purdue University, West Lafayette, Indiana, 47907, USA.
| | - Shasha Zhang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
139
|
Hematopoietic Progenitors and the Bone Marrow Niche Shape the Inflammatory Response and Contribute to Chronic Disease. Int J Mol Sci 2022; 23:ijms23042234. [PMID: 35216355 PMCID: PMC8879433 DOI: 10.3390/ijms23042234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/17/2022] Open
Abstract
It is now well understood that the bone marrow (BM) compartment can sense systemic inflammatory signals and adapt through increased proliferation and lineage skewing. These coordinated and dynamic alterations in responding hematopoietic stem and progenitor cells (HSPCs), as well as in cells of the bone marrow niche, are increasingly viewed as key contributors to the inflammatory response. Growth factors, cytokines, metabolites, microbial products, and other signals can cause dysregulation across the entire hematopoietic hierarchy, leading to lineage-skewing and even long-term functional adaptations in bone marrow progenitor cells. These alterations may play a central role in the chronicity of disease as well as the links between many common chronic disorders. The possible existence of a form of “memory” in bone marrow progenitor cells is thought to contribute to innate immune responses via the generation of trained immunity (also called innate immune memory). These findings highlight how hematopoietic progenitors dynamically adapt to meet the demand for innate immune cells and how this adaptive response may be beneficial or detrimental depending on the context. In this review, we will discuss the role of bone marrow progenitor cells and their microenvironment in shaping the scope and scale of the immune response in health and disease.
Collapse
|
140
|
Stratman AN, Crewe C, Stahl PD. The microenvironment‐ a general hypothesis on the homeostatic function of extracellular vesicles. FASEB Bioadv 2022; 4:284-297. [PMID: 35520390 PMCID: PMC9065581 DOI: 10.1096/fba.2021-00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/09/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022] Open
Abstract
Extracellular vesicles (EVs), exosomes and microvesicles, is a burgeoning field of biological and biomedical research that may change our understanding of cell communication in plants and animals while holding great promise for the diagnosis of disease and the development of therapeutics. However, the challenge remains to develop a general hypothesis about the role of EVs in physiological homeostasis and pathobiology across kingdoms. While they can act systemically, EVs are often seen to operate locally within a microenvironment. This microenvironment is built as a collection of microunits comprised of cells that interact with each other via EV exchange, EV signaling, EV seeding, and EV disposal. We propose that microunits are part of a larger matrix at the tissue level that collectively communicates with the surrounding environment, including other end‐organ systems. Herein, we offer a working model that encompasses the various facets of EV function in the context of the cell biology and physiology of multicellular organisms.
Collapse
Affiliation(s)
- Amber N Stratman
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| | - Clair Crewe
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
- Department of Internal Medicine Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| | - Philip D Stahl
- Department of Cell Biology and Physiology Washington University School of Medicine 660 South Euclid Avenue St. Louis Missouri USA 63110
| |
Collapse
|
141
|
Crewe C, Scherer PE. Intercellular and interorgan crosstalk through adipocyte extracellular vesicles. Rev Endocr Metab Disord 2022; 23:61-69. [PMID: 33447986 DOI: 10.1007/s11154-020-09625-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2020] [Indexed: 12/13/2022]
Abstract
Functional adipose tissue is essential for homeostatic maintenance of systemic metabolism. As such, adipose tissue dysfunction, like that seen in the obese state, directly contributes to system-wide pathological metabolism, leading to the development of type 2 diabetes and other obesity-associated comorbidities. In addition to the storage function of adipocytes, they also secrete numerous factors that robustly regulate metabolism-related pathways throughout the body. Many of these factors, in addition to other signaling proteins, RNA species and lipids, are found in extracellular vesicles (EVs) released from adipocytes. EVs are vesicles with a lipid bilayer, known to carry signaling proteins and lipids, mRNAs and miRNAs. Because of this diverse cargo, EVs can have robust and pleotropic signaling effects depending on the receiving target cells. We are only now starting to understand how adipocyte EVs can modulate metabolism within adipose tissue and beyond. Here, we highlight the current literature that demonstrates EV-mediated crosstalk between adipocytes and other tissues or distal cells. We become increasingly aware of the importance of these adipocyte-derived EV signals that establish a so far underappreciated endocrine system. Adipocyte EVs offer a new avenue for pharmacological manipulation of metabolism to treat obesity-related disease.
Collapse
Affiliation(s)
- Clair Crewe
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Internal Medicine and Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
142
|
Sabaratnam R, Wojtaszewski JFP, Højlund K. Factors mediating exercise-induced organ crosstalk. Acta Physiol (Oxf) 2022; 234:e13766. [PMID: 34981891 DOI: 10.1111/apha.13766] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 10/11/2021] [Accepted: 01/01/2022] [Indexed: 12/21/2022]
Abstract
Exercise activates a plethora of metabolic and signalling pathways in skeletal muscle and other organs causing numerous systemic beneficial metabolic effects. Thus, regular exercise may ameliorate and prevent the development of several chronic metabolic diseases. Skeletal muscle is recognized as an important endocrine organ regulating systemic adaptations to exercise. Skeletal muscle may mediate crosstalk with other organs through the release of exercise-induced cytokines, peptides and proteins, termed myokines, into the circulation. Importantly, other tissues such as the liver and adipose tissue may also release cytokines and peptides in response to exercise. Hence, exercise-released molecules are collectively called exerkines. Moreover, extracellular vesicles (EVs), in the form of exosomes or microvesicles, may carry some of the signals involved in tissue crosstalk. This review focuses on the role of factors potentially mediating crosstalk between muscle and other tissues in response to exercise.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Steno Diabetes Center Odense Odense University Hospital Odense C Denmark
- Section of Molecular Diabetes & Metabolism, Department of Clinical Research & Department of Molecular Medicine University of Southern Denmark Odense C Denmark
| | - Jørgen F. P. Wojtaszewski
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense Odense University Hospital Odense C Denmark
- Section of Molecular Diabetes & Metabolism, Department of Clinical Research & Department of Molecular Medicine University of Southern Denmark Odense C Denmark
| |
Collapse
|
143
|
Gao H, Luo Z, Ji Y, Tang K, Jin Z, Ly C, Sears DD, Mahata S, Ying W. Accumulation of microbial DNAs promotes to islet inflammation and β cell abnormalities in obesity in mice. Nat Commun 2022; 13:565. [PMID: 35091566 PMCID: PMC8799656 DOI: 10.1038/s41467-022-28239-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Various microbial products leaked from gut lumen exacerbate tissue inflammation and metabolic disorders in obesity. Vsig4+ macrophages are key players preventing infiltration of bacteria and their products into host tissues. However, roles of islet Vsig4+ macrophages in the communication between microbiota and β cells in pathogenesis of obesity-associated islet abnormalities are unknown. Here, we find that bacterial DNAs are enriched in β cells of individuals with obesity. Intestinal microbial DNA-containing extracellular vesicles (mEVs) readily pass through obese gut barrier and deliver microbial DNAs into β cells, resulting in elevated inflammation and impaired insulin secretion by triggering cGAS/STING activation. Vsig4+ macrophages prevent mEV infiltration into β cells through a C3-dependent opsonization, whereas loss of Vsig4 leads to microbial DNA enrichment in β cells after mEV treatment. Removal of microbial DNAs blunts mEV effects. Loss of Vsig4+ macrophages leads to microbial DNA accumulation in β cells and subsequently obesity-associated islet abnormalities.
Collapse
Affiliation(s)
- Hong Gao
- Division of Endocrinology & Metabolism, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Zhenlong Luo
- Division of Endocrinology & Metabolism, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Gastroenterology, Tongji Hospital, Tongji medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yudong Ji
- Division of Endocrinology & Metabolism, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care, Union Hospital, Tongji medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Kechun Tang
- VA San Diego Healthcare System, La Jolla, CA, 92093, USA
| | - Zhongmou Jin
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Crystal Ly
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dorothy D Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, 85004, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Family Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, 92093, CA, USA
| | - Sushil Mahata
- Division of Endocrinology & Metabolism, University of California, San Diego, La Jolla, CA, 92093, USA
- VA San Diego Healthcare System, La Jolla, CA, 92093, USA
| | - Wei Ying
- Division of Endocrinology & Metabolism, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
144
|
Manakanatas C, Ghadge SK, Agic A, Sarigol F, Fichtinger P, Fischer I, Foisner R, Osmanagic-Myers S. Endothelial and systemic upregulation of miR-34a-5p fine-tunes senescence in progeria. Aging (Albany NY) 2022; 14:195-224. [PMID: 35020601 PMCID: PMC8791216 DOI: 10.18632/aging.203820] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/25/2021] [Indexed: 11/25/2022]
Abstract
Endothelial defects significantly contribute to cardiovascular pathology in the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS). Using an endothelium-specific progeria mouse model, we identify a novel, endothelium-specific microRNA (miR) signature linked to the p53-senescence pathway and a senescence-associated secretory phenotype (SASP). Progerin-expressing endothelial cells exert profound cell-non-autonomous effects initiating senescence in non-endothelial cell populations and causing immune cell infiltrates around blood vessels. Comparative miR expression analyses revealed unique upregulation of senescence-associated miR34a-5p in endothelial cells with strong accumulation at atheroprone aortic arch regions but also, in whole cardiac- and lung tissues as well as in the circulation of progeria mice. Mechanistically, miR34a-5p knockdown reduced not only p53 levels but also late-stage senescence regulator p16 with no effect on p21 levels, while p53 knockdown reduced miR34a-5p and partially rescued p21-mediated cell cycle inhibition with a moderate effect on SASP. These data demonstrate that miR34a-5p reinforces two separate senescence regulating branches in progerin-expressing endothelial cells, the p53- and p16-associated pathways, which synergistically maintain a senescence phenotype that contributes to cardiovascular pathology. Thus, the key function of circulatory miR34a-5p in endothelial dysfunction-linked cardiovascular pathology offers novel routes for diagnosis, prognosis and treatment for cardiovascular aging in HGPS and potentially geriatric patients.
Collapse
Affiliation(s)
- Christina Manakanatas
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Santhosh Kumar Ghadge
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Azra Agic
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Fatih Sarigol
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Petra Fichtinger
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Irmgard Fischer
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Roland Foisner
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Selma Osmanagic-Myers
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
- Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
145
|
Gan L, Liu D, Xie D, Bond Lau W, Liu J, Christopher TA, Lopez B, Liu L, Hu H, Yao P, He Y, Gao E, Koch WJ, Zhao J, Ma XL, Cao Y, Wang Y. Ischemic Heart-Derived Small Extracellular Vesicles Impair Adipocyte Function. Circ Res 2022; 130:48-66. [PMID: 34763521 DOI: 10.1161/circresaha.121.320157] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Patients with acute myocardial infarction suffer systemic metabolic dysfunction via incompletely understood mechanisms. Adipocytes play critical role in metabolic homeostasis. The impact of acute myocardial infarction upon adipocyte function is unclear. Small extracellular vesicles (sEVs) critically contribute to organ-organ communication. Whether and how small extracellular vesicle mediate post-MI cardiomyocyte/adipocyte communication remain unknown. METHODS Plasma sEVs were isolated from sham control (Pla-sEVSham) or 3 hours after myocardial ischemia/reperfusion (Pla-sEVMI/R) and incubated with adipocytes for 24 hours. Compared with Pla-sEVSham, Pla-sEVMI/R significantly altered expression of genes known to be important in adipocyte function, including a well-known metabolic regulatory/cardioprotective adipokine, APN (adiponectin). Pla-sEVMI/R activated 2 (PERK-CHOP and ATF6 [transcription factor 6]-EDEM [ER degradation enhancing alpha-mannosidase like protein 1] pathways) of the 3 endoplasmic reticulum (ER) stress pathways in adipocytes. These pathological alterations were also observed in adipocytes treated with sEVs isolated from adult cardiomyocytes subjected to in vivo myocardial ischemia/reperfusion (MI/R) (Myo-sEVMI/R). Bioinformatic/RT-qPCR analysis demonstrates that the members of miR-23-27-24 cluster are significantly increased in Pla-sEVMI/R, Myo-sEVMI/R, and adipose tissue of MI/R animals. Administration of cardiomyocyte-specific miR-23-27-24 sponges abolished adipocyte miR-23-27-24 elevation in MI/R animals, supporting the cardiomyocyte origin of adipocyte miR-23-27-24 cluster. In similar fashion to Myo-sEVMI/R, a miR-27a mimic activated PERK-CHOP and ATF6-EDEM-mediated ER stress. Conversely, a miR-27a inhibitor significantly attenuated Myo-sEVMI/R-induced ER stress and restored APN production. RESULTS An unbiased approach identified EDEM3 (ER degradation enhancing alpha-mannosidase like protein 3) as a novel downstream target of miR-27a. Adipocyte EDEM3 deficiency phenocopied multiple pathological alterations caused by Myo-sEVMI/R, whereas EDEM3 overexpression attenuated Myo-sEVMI/R-resulted ER stress. Finally, administration of GW4869 or cardiomyocyte-specific miR-23-27-24 cluster sponges attenuated adipocyte ER stress, improved adipocyte endocrine function, and restored plasma APN levels in MI/R animals. CONCLUSIONS We demonstrate for the first time that MI/R causes significant adipocyte ER stress and endocrine dysfunction by releasing miR-23-27-24 cluster-enriched small extracellular vesicle. Targeting small extracellular vesicle-mediated cardiomyocyte-adipocyte pathological communication may be of therapeutic potential to prevent metabolic dysfunction after MI/R.
Collapse
Affiliation(s)
- Lu Gan
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China (D.L.)
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Jing Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Bernard Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Lian Liu
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Hang Hu
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Yarong He
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
| | - Erhe Gao
- Laboratory of Emergency Medicine, Department of Emergency Medicine and National Clinical Research Center for Geriatrics, West China Hospital (L.G., L.L., H.H., P.Y., Y.H., Y.C.), Sichuan University, Chengdu, China
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.J.K.)
| | - Walter J Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G., W.J.K.)
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| | - Yu Cao
- Disaster Medical Center (Y.C.), Sichuan University, Chengdu, China
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (L.G., D.L., D.X., W.B.L., J.L., T.A.C., B.L., P.Y., J.Z., X.-L.M., Y.W.)
| |
Collapse
|
146
|
Sun W. Analysis of Single-Cell/Nucleus Transcriptome Data in Adipose Tissue. Methods Mol Biol 2022; 2448:291-306. [PMID: 35167105 DOI: 10.1007/978-1-0716-2087-8_19] [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] [Indexed: 06/14/2023]
Abstract
Adipose tissue is highly heterogeneous and plastic. Recent advances in single-cell/nucleus RNA sequencing technology have helped to study the cellular composition and dynamics of adipose tissue. In this protocol, I outline a typical workflow of analyzing single-cell/nucleus transcriptome data. Specifically, I show an example of how cellular populations are estimated and characterized from a single-nucleus RNAseq data set of frozen archived human adipose tissue.
Collapse
Affiliation(s)
- Wenfei Sun
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
147
|
Le Lay S, Magré J, Prieur X. Not Enough Fat: Mouse Models of Inherited Lipodystrophy. Front Endocrinol (Lausanne) 2022; 13:785819. [PMID: 35250856 PMCID: PMC8895270 DOI: 10.3389/fendo.2022.785819] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Lipodystrophies belong to the heterogenous group of syndromes in which the primary defect is a generalized or partial absence of adipose tissue, which may be congenital or acquired in origin. Lipodystrophy should be considered in patients manifesting the combination of insulin resistance (with or without overt diabetes), dyslipidemia and fatty liver. Lipodystrophies are classified according to the etiology of the disease (genetic or acquired) and to the anatomical distribution of adipose tissue (generalized or partial). The mechanism of adipose tissue loss is specific to each syndrome, depending on the biological function of the mutated gene. Mice models, together with cellular studies have permitted clarification of the mechanisms by which human mutations deeply compromise adipocyte homeostasis. In addition, rodent models have proven to be crucial in deciphering the cardiometabolic consequences of the lack of adipose tissue such as NAFLD, muscle insulin resistance and cardiomyopathy. More precisely, tissue-specific transgenic and knockout mice have brought new tools to distinguish phenotypic traits that are the consequences of lipodystrophy from those that are cell-autonomous. In this review, we discuss the mice models of lipodystrophy including those of inherited human syndromes of generalized and partial lipodystrophy. We present how these models have demonstrated the central role of white adipose tissue in energetic homeostasis in general, including insulin sensitivity and lipid handling in particular. We underscore the differences reported with the human phenotype and discuss the limit of rodent models in recapitulating adipose tissue primary default. Finally, we present how these mice models have highlighted the function of the causative-genes and brought new insights into the pathophysiology of the cardiometabolic complications associated with lipodystrophy.
Collapse
Affiliation(s)
- Soazig Le Lay
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
- Univ Angers, SFR ICAT, Angers, France
| | - Jocelyne Magré
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
| | - Xavier Prieur
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
- *Correspondence: Xavier Prieur,
| |
Collapse
|
148
|
Garcia-Martin R, Wang G, Brandão BB, Zanotto TM, Shah S, Kumar Patel S, Schilling B, Kahn CR. MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 2022; 601:446-451. [PMID: 34937935 PMCID: PMC9035265 DOI: 10.1038/s41586-021-04234-3] [Citation(s) in RCA: 392] [Impact Index Per Article: 130.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 11/12/2021] [Indexed: 11/09/2022]
Abstract
Exosomes and other small extracellular vesicles (sEVs) provide a unique mode of cell-to-cell communication in which microRNAs (miRNAs) produced and released from one cell are taken up by cells at a distance where they can enact changes in gene expression1-3. However, the mechanism by which miRNAs are sorted into exosomes/sEVs or retained in cells remains largely unknown. Here we demonstrate that miRNAs possess sorting sequences that determine their secretion in sEVs (EXOmotifs) or cellular retention (CELLmotifs) and that different cell types, including white and brown adipocytes, endothelium, liver and muscle, make preferential use of specific sorting sequences, thus defining the sEV miRNA profile of that cell type. Insertion or deletion of these CELLmotifs or EXOmotifs in a miRNA increases or decreases retention in the cell of production or secretion into exosomes/sEVs. Two RNA-binding proteins, Alyref and Fus, are involved in the export of miRNAs carrying one of the strongest EXOmotifs, CGGGAG. Increased miRNA delivery mediated by EXOmotifs leads to enhanced inhibition of target genes in distant cells. Thus, this miRNA code not only provides important insights that link circulating exosomal miRNAs to tissues of origin, but also provides an approach for improved targeting in RNA-mediated therapies.
Collapse
Affiliation(s)
- Ruben Garcia-Martin
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Guoxiao Wang
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Bruna B Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Tamires M Zanotto
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Samah Shah
- The Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
149
|
Camino T, Lago-Baameiro N, Bravo SB, Molares-Vila A, Sueiro A, Couto I, Baltar J, Casanueva EF, Pardo M. Human obese white adipose tissue sheds depot-specific extracellular vesicles and reveals candidate biomarkers for monitoring obesity and its comorbidities. Transl Res 2022; 239:85-102. [PMID: 33465489 DOI: 10.1016/j.trsl.2021.01.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 12/23/2022]
Abstract
Extracellular vesicles (EVs) have been recently postulated as key players in metabolic disorders emerging as an alternative way of paracrine/endocrine communication. However, the nature of EVs shed by adipose tissue (AT) and their role in obesity is still very limited. Here, we isolated human morbid obese visceral (VAT) and subcutaneous (SAT) whole AT shed EVs from donors submitted to bariatric surgery to characterize their protein cargo by qualitative and quantitative/SWATH mass spectrometry analysis. We identified 574 different proteins shed by morbid obese VAT and 401 proteins in those from SAT, establishing the first obese AT EV proteome reference map. Only 50% of identified proteins in VAT vesicles were common to those in SAT; additionally, EVs shed by obese VAT showed more AT and obesity-related adipokines than SAT. Functional classification shows that obese VAT vesicles exhibit an enrichment of proteins implicated in AT inflammation and insulin resistance such as TGFBI, CAVN1, CD14, mimecan, thrombospondin-1, FABP-4 or AHNAK. Selected candidate biomarkers from the quantitative-SWATH analysis were validated in EVs from independent morbid obese and from moderate obese to lean individuals showing that morbid obese VAT vesicles are characterized by a diminution of syntenin 1 and the elevation of TGFBI and mimecan. Interestingly, TGFBI and mimecan containing vesicles could be detected and quantified at circulating level in plasma. Thus, a significant elevation of -TGFBI-EVs was detected on those obese patients with a history of T2D compared to nondiabetic, and an augmentation of mimecan-EVs in obese plasma compared to those in healthy lean individuals. Thus, we conclude that obese AT release functional EVs carrying AT and obesity candidate biomarkers which vary regarding the AT of origin. Our findings suggest that circulating EV-TGFBI may facilitate monitoring T2D status in obese patients, and EV-mimecan may be useful to track adiposity, and more precisely, visceral obesity.
Collapse
Affiliation(s)
- Tamara Camino
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Nerea Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Susana Belén Bravo
- Unidad de Proteómica, Instituto de Investigación Sanitaria de Santiago (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Alberto Molares-Vila
- Bioinformatics Platform, Instituto de Investigación Sanitaria de Santiago (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Aurelio Sueiro
- Grupo Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Spain
| | - Iván Couto
- Servicio de Cirugía Plástica y Reparadora, Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Javier Baltar
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; Servicio de Cirugía General, Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain
| | - Eelipe F Casanueva
- Grupo Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Spain; CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Maria Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), Santiago de Compostela, Spain; CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
150
|
Deng H, Chen Y. The role of adipose-derived stem cells-derived extracellular vesicles in the treatment of diabetic foot ulcer: Trends and prospects. Front Endocrinol (Lausanne) 2022; 13:902130. [PMID: 35966086 PMCID: PMC9363610 DOI: 10.3389/fendo.2022.902130] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic foot ulcer(DFU) is one of the most severe chronic complications of type 2 diabetes mellitus, which is mainly caused by peripheral vascular occlusion with various degrees of infection. Treatment of DFU is difficult, and ulcer formation in lower limbs and deep-tissue necrosis might lead to disability or even death. Insulin resistance is the major mechanism of type 2 diabetes mellitus development, largely caused by adipose tissue dysfunction. However, adipose tissue was recently identified as an important endocrine organ that secretes bio-active factors, such as adipokines and extracellular vesicles(EVs). And adipose tissue-derived stem cells(ADSCs) are abundant in adipose tissue and have become a hot topic in the tissue engineering field. In particular, EVs derived from ADSCs contain abundant biomarkers and mediators. These EVs exert significant effects on distant cells and organs, contributing to metabolic homeostasis. In this review, we aim to elaborate on the mechanisms of diabetic non-healing wound development and the role of ADSCs-EVs in wound repair, which might provide a new therapy for treating DFU.
Collapse
Affiliation(s)
- Hongyan Deng
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Internal Medicine, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Laboratory of Endocrinology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
- *Correspondence: Yong Chen,
| |
Collapse
|