1
|
Przywara D, Petniak A, Gil-Kulik P. Optimizing Mesenchymal Stem Cells for Regenerative Medicine: Influence of Diabetes, Obesity, Autoimmune, and Inflammatory Conditions on Therapeutic Efficacy: A Review. Med Sci Monit 2024; 30:e945331. [PMID: 39154207 PMCID: PMC11340262 DOI: 10.12659/msm.945331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising tool that may be used in regenerative medicine. Thanks to their ability to differentiate and paracrine signaling, they can be used in the treatment of many diseases. Undifferentiated MSCs can support the regeneration of surrounding tissues through secreted substances and exosomes. This is possible thanks to the production of growth factors. These factors stimulate the growth of neighboring cells, have an anti-apoptotic effect, and support angiogenesis, and MSCs also have an immunomodulatory effect. The level of secreted factors may vary depending on many factors. Apart from the donor's health condition, it is also influenced by the source of MSCs, methods of harvesting, and even the banking of cells. This work is a review of research on how the patient's health condition affects the properties of obtained MSCs. The review discusses the impact of the patient's diabetes, obesity, autoimmune diseases, and inflammation, as well as the impact of the source of MSCs and methods of harvesting and banking cells on the phenotype, differentiation capacity, anti-inflammatory, angiogenic effects, and proliferation potential of MSCs. Knowledge about specific clinical factors allows for better use of the potential of stem cells and more appropriate targeting of procedures for collecting, multiplying, and banking these cells, as well as for their subsequent use. This article aims to review the characteristics, harvesting, banking, and paracrine signaling of MSCs and their role in diabetes, obesity, autoimmune and inflammatory diseases, and potential role in regenerative medicine.
Collapse
|
2
|
Lanci A, Iacono E, Merlo B. Therapeutic Application of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Domestic Animals. Animals (Basel) 2024; 14:2147. [PMID: 39123673 PMCID: PMC11310970 DOI: 10.3390/ani14152147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Recently, the therapeutic potential of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) has been extensively studied in both human and veterinary medicine. EVs are nano-sized particles containing biological components commonly found in other biological materials. For that reason, EV isolation and characterization are critical to draw precise conclusions during their investigation. Research on EVs within veterinary medicine is still considered in its early phases, yet numerous papers were published in recent years. The conventional adult tissues for deriving MSCs include adipose tissue and bone marrow. Nonetheless, alternative sources such as synovial fluid, endometrium, gingiva, and milk have also been intermittently used. Fetal adnexa are amniotic membrane/fluid, umbilical cord and Wharton's jelly. Cells derived from fetal adnexa exhibit an intermediate state between embryonic and adult cells, demonstrating higher proliferative and differentiative potential and longer telomeres compared to cells from adult tissues. Summarized here are the principal and recent preclinical and clinical studies performed in domestic animals such as horse, cattle, dog and cat. To minimize the use of antibiotics and address the serious issue of antibiotic resistance as a public health concern, they will undoubtedly also be utilized in the future to treat infections in domestic animals. A number of concerns, including large-scale production with standardization of EV separation and characterization techniques, must be resolved for clinical application.
Collapse
Affiliation(s)
- Aliai Lanci
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
| | - Eleonora Iacono
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, 40100 Bologna, Italy
| | - Barbara Merlo
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (E.I.); (B.M.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, 40100 Bologna, Italy
| |
Collapse
|
3
|
Huang W, Hong S, Zhu X, Alsaeedi MH, Tang H, Krier JD, Gandhi D, Jordan KL, Saadiq IM, Jiang Y, Eirin A, Lerman LO. Obesity Blunts the Effect of Mesenchymal Stem Cell-Derived Extracellular Vesicles. Kidney Int Rep 2023; 8:1841-1851. [PMID: 37705914 PMCID: PMC10496020 DOI: 10.1016/j.ekir.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/02/2023] [Accepted: 06/12/2023] [Indexed: 09/15/2023] Open
Abstract
Introduction Mesenchymal stem/stromal cell-derived extracellular vesicles (MSC-EVs) are paracrine vectors with therapeutic functions comparable to their parent cells. However, it remains unclear if donor obesity affects their therapeutic functions. We tested the hypothesis that the curative effect of human adipose tissue-derived MSC-EVs (A-MSC-EVs) is blunted by obesity. Methods MSC-EVs were isolated by ultracentrifugation from mesenchymal stem/stromal cells (MSCs) collected from abdominal subcutaneous fat of obese and lean human subjects (obese and lean-MSC-EVs, respectively) and injected into the aorta of mice 2 weeks after renal artery stenosis (RAS) induction. Magnetic resonance imaging studies were conducted 2 weeks after MSC-EVs delivery to determine renal function. The effect of MSC-EVs on tissue injury was assessed by histology and gene expression of inflammatory factors, including interleukin (IL)-1β, IL-6, monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor alpha (TNF-α). Oxidative damage, macrophage infiltration, plasma renin, and hypoxia inducible factor-1α (HIF-1α) were also assessed. Results Tracking showed that MSC-EVs localized in the kidney tissue, including glomeruli and tubules. All MSC-EVs decreased systolic blood pressure (SBP) and plasma renin and improved the poststenotic kidney (STK) volume, but obese-MSC-EVs were less effective than lean-MSC-EVs in improving medullary hypoxia, fibrosis, and tubular injury. Lean-MSC-EVs decreased inflammation, whereas obesity attenuated this effect. Only lean-MSC-EVs decreased STK cortical HIF-1α expression. Conclusion Obesity attenuates the antihypoxia, antifibrosis, antiinflammation, and tubular repair functions of human MSC-EVs in chronic ischemic kidney disease. These observations may have implications for the self-repair potency of obese subjects and for the use of autologous MSC-EVs in regenerative medicine.
Collapse
Affiliation(s)
- Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Mina H. Alsaeedi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - James D. Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Deep Gandhi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kyra L. Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Ishran M. Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Yamei Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Hong S, Huang W, Zhu X, Tang H, Krier JD, Xing L, Lu B, Gandhi D, Jordan KL, Saadiq IM, Lerman A, Eirin A, Lerman LO. Obesity blunts amelioration of cardiac hypertrophy and fibrosis by human mesenchymal stem/stromal cell-derived extracellular vesicles. Am J Physiol Heart Circ Physiol 2023; 325:H163-H171. [PMID: 37294895 PMCID: PMC10312317 DOI: 10.1152/ajpheart.00676.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/12/2023] [Accepted: 06/01/2023] [Indexed: 06/11/2023]
Abstract
Renovascular hypertension (RVH) can induce cardiac damage that is reversible using adipose tissue-derived mesenchymal stromal/stem cells (A-MSCs). However, A-MSCs isolated from patients with obesity are less effective than lean-A-MSC in blunting hypertensive cardiomyopathy in mice with RVH. We tested the hypothesis that this impairment extends to their obese A-MSC-extracellular vesicles (EVs) progeny. MSCs were harvested from the subcutaneous fat of obese and lean human subjects, and their EVs were collected and injected into the aorta of mice 2 wk after renal artery stenosis or sham surgery. Cardiac left ventricular (LV) function was studied with MRI 2 wk later, and myocardial tissue ex vivo. Blood pressure, LV myocardial wall thickness, mass, and fibrosis that were elevated in RVH mice were suppressed only by lean EVs. Hence, human A-MSC-derived lean EVs are more effective than obese EVs in blunting hypertensive cardiac injury in RVH mice. These observations highlight impaired paracrine repair potency of endogenous MSCs in patients with obesity.NEW & NOTEWORTHY Injection of A-MSC-derived EVs harvested from patients who are lean can resolve myocardial injury in mice with experimental renovascular hypertension more effectively than A-MSC-derived EVs from patients with obesity. These observations underscore and might have important ramifications for the self-healing capacity of patients with obesity and for the use of autologous EVs as a regenerative tool.
Collapse
Affiliation(s)
- Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Li Xing
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Bo Lu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Deep Gandhi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
5
|
Glasstetter LM, Oderinde TS, Mirchandani M, Rajagopalan KS, Barsom SH, Thaler R, Siddiqi S, Zhu XY, Tang H, Jordan KL, Saadiq IM, van Wijnen AJ, Eirin A, Lerman LO. Obesity and dyslipidemia are associated with partially reversible modifications to DNA hydroxymethylation of apoptosis- and senescence-related genes in swine adipose-derived mesenchymal stem/stromal cells. Stem Cell Res Ther 2023; 14:143. [PMID: 37231414 PMCID: PMC10214739 DOI: 10.1186/s13287-023-03372-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Obesity dysregulates key biological processes underlying the functional homeostasis, fate decisions, and reparative potential of mesenchymal stem/stromal cells (MSCs). Mechanisms directing obesity-induced phenotypic alterations in MSCs remain unclear, but emerging drivers include dynamic modification of epigenetic marks, like 5-hydroxymethylcytosine (5hmC). We hypothesized that obesity and cardiovascular risk factors induce functionally relevant, locus-specific changes in 5hmC of swine adipose-derived MSCs and evaluated their reversibility using an epigenetic modulator, vitamin-C. METHODS Female domestic pigs were fed a 16-week Lean or Obese diet (n = 6 each). MSCs were harvested from subcutaneous adipose tissue, and 5hmC profiles were examined through hydroxymethylated DNA immunoprecipitation sequencing (hMeDIP-seq) followed by an integrative (hMeDIP and mRNA sequencing) gene set enrichment analysis. For clinical context, we compared 5hmC profiles of adipose tissue-derived human MSCs harvested from patients with obesity and healthy controls. RESULTS hMeDIP-seq revealed 467 hyper- (fold change ≥ 1.4; p-value ≤ 0.05) and 591 hypo- (fold change ≤ 0.7; p-value ≤ 0.05) hydroxymethylated loci in swine Obese- versus Lean-MSCs. Integrative hMeDIP-seq/mRNA-seq analysis identified overlapping dysregulated gene sets and discrete differentially hydroxymethylated loci with functions related to apoptosis, cell proliferation, and senescence. These 5hmC changes were associated with increased senescence in cultured MSCs (p16/CDKN2A immunoreactivity, senescence-associated β-galactosidase [SA-β-Gal] staining), were partly reversed in swine Obese-MSCs treated with vitamin-C, and shared common pathways with 5hmC changes in human Obese-MSCs. CONCLUSIONS Obesity and dyslipidemia are associated with dysregulated DNA hydroxymethylation of apoptosis- and senescence-related genes in swine and human MSCs, potentially affecting cell vitality and regenerative functions. Vitamin-C may mediate reprogramming of this altered epigenomic landscape, providing a potential strategy to improve the success of autologous MSC transplantation in obese patients.
Collapse
Affiliation(s)
- Logan M Glasstetter
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Tomiwa S Oderinde
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mohit Mirchandani
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Samer H Barsom
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Sarosh Siddiqi
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
6
|
Zhang Y, Huang X, Sun T, Shi L, Liu B, Hong Y, Fu QL, Zhang Y, Li X. MicroRNA-19b-3p dysfunction of mesenchymal stem cell-derived exosomes from patients with abdominal aortic aneurysm impairs therapeutic efficacy. J Nanobiotechnology 2023; 21:135. [PMID: 37101174 PMCID: PMC10131394 DOI: 10.1186/s12951-023-01894-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
Senescence of vascular smooth muscle cells (VSMCs) contributes to the formation of abdominal aortic aneurysm (AAA). Although mesenchymal stem cell exosomes (MSC-EXO) have been confirmed to restrict the development of AAA, their biological activity depends largely on the physiological state of the MSCs. This study aimed to compare the effects of adipose-derived MSC-EXO from healthy donors (HMEXO) and AAA patients (AMEXO) on senescence of VSMCs in AAA and explore the underlying mechanisms. An ApoE-/- mouse model of AAA was used to investigate the therapeutic effects of HMEXO, AMEXO or miR-19b-3p-AMEXO on AAA development. This in vitro model of AAA was established by treating VSMCs with Ang II (Angiotensin II). The senescence of VSMCs was determined by senescence-associated β-galactosidase (SA-β-gal) staining. The morphology of mitochondria in VSMCs was examined by MitoTracker staining. HMEXO exhibited superior capacity compared with AMEXO to inhibit VSMC senescence and attenuate AAA formation in Ang II-treated ApoE-/- mice. In vitro, both AMEXO and HMEXO inhibited Ang II-induced VSMC senescence via downregulation of mitochondrial fission. Notably, compared with HMEXO, the ability of AMEXO to inhibit VSMC senescence was significantly decreased. miRNA sequencing and the expression of miR-19b-3p was significantly decreased in AMEXO compared with HMEXO. Luciferase assay suggested that MST4 (Mammalian sterile-20-like kinase 4) is a potential target of miR-19b-3p. Mechanistically, miR-19b-3p in HMEXO ameliorated VSMC senescence by inhibiting mitochondrial fission via regulation of the MST4/ERK/Drp1 signaling pathway. Overexpression of miR-19b-3p in AMEXO improved their beneficial effect on AAA formation. Our study reveals that MSC-exosomal miR-19b-3p exerts protective effects against Ang II-induced AAA and VSMC senescence via regulation of the MST4/ERK/Drp1 pathway. The pathological state of AAA patients alters the miRNA components of AMEXO and impairs their therapeutic benefits.
Collapse
Affiliation(s)
- Yuxiao Zhang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoran Huang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Tucheng Sun
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Linli Shi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Baojuan Liu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Xin Li
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Hong S, Zhu XY, Jiang Y, Zhang L, Tang H, Jordan KL, Saadiq IM, Huang W, Lerman A, Eirin A, Lerman LO. Autologous Extracellular Vesicles Attenuate Cardiac Injury in Experimental Atherosclerotic Renovascular Disease More Effectively Than Their Parent Mesenchymal Stem/Stromal Cells. Stem Cell Rev Rep 2023; 19:700-712. [PMID: 36344721 PMCID: PMC10073252 DOI: 10.1007/s12015-022-10473-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Abstract
Atherosclerotic renovascular disease (RVD) leads to hypertension, chronic kidney disease (CKD), and heart disease. Intrarenal delivery of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (EVs) attenuate renal injury and suppress release of inflammatory cytokines in porcine RVD. We hypothesized that this strategy would also be useful for cardioprotection. Pigs with renovascular hypertension and metabolic syndrome were studied 4 weeks after treatment with a single intrarenal infusion of autologous MSCs, EVs, or vehicle. Cardiac structure and function were assessed in vivo, and myocardial remodeling and expression of the pro-fibrotic factor growth factor receptor-bound protein-2 (Grb2) were measured ex-vivo. Inflammatory cytokine levels were measured in the systemic circulation and myocardial tissue. Blood pressure was elevated in all RVD groups, but serum creatinine increased in RVD and decreased in both RVD + MSCs and RVD + EVs. RVD-induced diastolic dysfunction (lower E/A ratio) was normalized in both MSCs- and EVs- treated pigs. Intrarenal delivery of MSCs and EVs also attenuated RVD-induced myocardial fibrosis, collagen deposition, and Grb2 expression, yet EVs restored capillary density and inflammation more effectively than MSCs. These observations suggest that autologous EVs attenuate cardiac injury in experimental RVD more effectively than their parent MSCs.
Collapse
Affiliation(s)
- Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yamei Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lei Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
8
|
Alberti G, Russo E, Corrao S, Anzalone R, Kruzliak P, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. Current Perspectives on Adult Mesenchymal Stromal Cell-Derived Extracellular Vesicles: Biological Features and Clinical Indications. Biomedicines 2022; 10:2822. [PMID: 36359342 PMCID: PMC9687875 DOI: 10.3390/biomedicines10112822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 08/10/2023] Open
Abstract
Extracellular vesicles (EVs) constitute one of the main mechanisms by which cells communicate with the surrounding tissue or at distance. Vesicle secretion is featured by most cell types, and adult mesenchymal stromal cells (MSCs) of different tissue origins have shown the ability to produce them. In recent years, several reports disclosed the molecular composition and suggested clinical indications for EVs derived from adult MSCs. The parental cells were already known for their roles in different disease settings in regulating inflammation, immune modulation, or transdifferentiation to promote cell repopulation. Interestingly, most reports also suggested that part of the properties of parental cells were maintained by isolated EV populations. This review analyzes the recent development in the field of cell-free therapies, focusing on several adult tissues as a source of MSC-derived EVs and the available clinical data from in vivo models.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Simona Corrao
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Rita Anzalone
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Peter Kruzliak
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | | | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
9
|
Jiang Y, Hong S, Zhu X, Zhang L, Tang H, Jordan KL, Saadiq IM, Huang W, Lerman A, Eirin A, Lerman LO. IL-10 partly mediates the ability of MSC-derived extracellular vesicles to attenuate myocardial damage in experimental metabolic renovascular hypertension. Front Immunol 2022; 13:940093. [PMID: 36203611 PMCID: PMC9530748 DOI: 10.3389/fimmu.2022.940093] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) obtain properties of immunomodulation and tissue repair from their parental mesenchymal stem cells (MSCs), and upon delivery may be associated with fewer adverse events. EVs derived from adipose-tissue MSCs restored kidney function by attenuating kidney inflammation in a swine model of metabolic syndrome (MetS) and renal artery stenosis via anti-inflammatory pathways. EVs also ameliorated myocardial injury in renovascular hypertension (RVH) secondary to inflammation in cardiorenal disease, but the mechanisms regulating this effect are unknown. We hypothesize that the anti-inflammatory cytokine interleukin (IL)-10 mediates the reparative effects of EVs on cardiovascular complications in a preclinical swine model with coexisting MetS and RVH. Twenty-three pigs established as Lean controls or RVH models were observed for 16 weeks. At 12 weeks RVH subgroups received an intrarenal delivery of 1011 either wildtype (WT) EVs or EVs after IL-10 knockdown (KD) (RVH+WT-EVs or RVH+IL-10-KD-EVs, respectively). Cardiac and renal function were studied in-vivo and myocardial tissue injury in-vitro 4 weeks later. RVH pigs showed myocardial inflammation, fibrosis, and left ventricular diastolic dysfunction. WT-EVs attenuated these impairments, increased capillary density, and decreased myocardial inflammation in-vivo. In-vitro, co-incubation with IL-10-containing WT-EVs decreased activated T-cells proliferation and endothelial cells inflammation and promoted their migration. Contrarily, these cardioprotective effects were largely blunted using IL-10-KD-EVs. Thus, the anti-inflammatory and pro-angiogenic effects of EVs in RVH may be partly attributed to their cargo of anti-inflammatory IL-10. Early intervention of IL-10-containing EVs may be helpful to prevent cardiovascular complications of MetS concurrent with RVH.
Collapse
Affiliation(s)
- Yamei Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Siting Hong
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Lei Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Kyra L. Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Ishran M. Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Weijun Huang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
10
|
Cluzel GL, Ryan PM, Herisson FM, Caplice NM. High-fidelity porcine models of metabolic syndrome: a contemporary synthesis. Am J Physiol Endocrinol Metab 2022; 322:E366-E381. [PMID: 35224983 DOI: 10.1152/ajpendo.00413.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This review aims to describe and compare porcine models of metabolic syndrome. This syndrome and its associated secondary comorbidities are set to become the greatest challenge to healthcare providers and policy makers in the coming century. However, an incomplete understanding of the pathogenesis has left significant knowledge gaps in terms of efficacious therapeutics. To further our comprehension and, in turn, management of metabolic syndrome, appropriate high-fidelity models of the disease complex are of great importance. In this context, our review aims to assess the most promising porcine models of metabolic syndrome currently available for their similarity to the human phenotype. In addition, we aim to highlight the strengths and shortcomings of each model in an attempt to identify the most appropriate application of each. Although no porcine model perfectly recapitulates the human metabolic syndrome, several pose satisfactory approximations. The Ossabaw miniature swine in particular represents a highly translatable model that develops each of the core parameters of the syndrome with many of the associated secondary comorbidities. Future high-fidelity porcine models of metabolic syndrome need to focus on secondary sequelae replication, which may require extended induction period to reveal.
Collapse
Affiliation(s)
- Gaston L Cluzel
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul M Ryan
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Florence M Herisson
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Noel M Caplice
- Centre for Research in Vascular Biology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
11
|
Effects of obesity on reparative function of human adipose tissue-derived mesenchymal stem cells on ischemic murine kidneys. Int J Obes (Lond) 2022; 46:1222-1233. [PMID: 35256761 PMCID: PMC9156526 DOI: 10.1038/s41366-022-01103-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/01/2023]
Abstract
Introduction: Obesity is a health burden that impairs cellular processes. Mesenchymal stem/stromal cells (MSCs) are endowed with reparative properties and can ameliorate renal injury. Obesity impairs human MSC function in-vitro, but its effect on their in-vivo reparative potency remains unknown. Subjects and Methods: Abdominal adipose tissue-derived MSC were harvested from patients without (‘lean’) or with obesity (‘obese’) (body mass index<30 or ≥30kg/m2, respectively) during kidney donation or bariatric surgery, respectively. MSC (5x105/200μL) or vehicle were then injected into 129S1 mice 2 weeks after renal artery stenosis (RAS) or sham surgery (n=8/group). Two weeks later, mice underwent magnetic resonance imaging to assess renal perfusion and oxygenation in-vivo, and kidneys then harvested for ex-vivo studies. Results: Similar numbers of lean and obese-MSCs engrafted in stenotic mouse kidneys. Vehicle-treated RAS mice had reduced stenotic-kidney cortical and medullary perfusion and oxygenation. Lean (but not obese) MSC normalized ischemic kidney cortical perfusion, whereas both effectively mitigated renal hypoxia. Serum creatinine and blood pressure were elevated in RAS mice and lowered only by lean-MSC. Both types of MSCs alleviated stenotic-kidney fibrosis, but lean-MSC more effectively than obese-MSC. MSC senescence-associated beta-gal activity, and gene expression of p16, p21, and vascular endothelial growth factor correlated with recipient kidney perfusion and tissue injury, linking MSC characteristics with their in-vivo reparative capacity. Discussion: Human obesity impairs the reparative properties of adipose-tissue-derived MSCs, possibly by inducing cellular senescence. Dysfunction and senescence of the endogenous MSC repair system in patients with obesity may warrant targeting interventions to restore MSC vitality.
Collapse
|
12
|
Adipose-derived stem cells and obesity: The spear and shield relationship. Genes Dis 2021; 10:175-186. [PMID: 37013055 PMCID: PMC10066342 DOI: 10.1016/j.gendis.2021.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/11/2021] [Accepted: 09/08/2021] [Indexed: 11/24/2022] Open
Abstract
With the transformation of modern lifestyles and population ageing, obesity has become a global epidemic, as one of the important threat to human health of chronic non-communicable diseases (NCD). Stem cell therapy seems promising as an alternative strategy for managing obesity and related metabolic problems. Adipose tissue-derived stem cells (ADSCs) have received widespread attention, which provides new ideas for the treatment of obesity and various metabolic-related diseases, due to their abundant reserves, easy acquisition, rapid expansion, and multi-directional differentiation potential, low immunogenicity and many other advantages. Accordingly, there seems to be a "shield and spear paradox" in the relationship between ADSCs and obesity. In this review, we emphatically summarized the role of ADSCs in the occurrence and development of obesity and related metabolic disease processes, in order to pave the way for clinical practice.
Collapse
|
13
|
Eirin A, Lerman LO. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles for Chronic Kidney Disease: Are We There Yet? Hypertension 2021; 78:261-269. [PMID: 34176287 DOI: 10.1161/hypertensionaha.121.14596] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) are the most utilized cell type for cellular therapy, partly due to their important proliferative potential and ability to differentiate into various cell types. MSCs produce large amounts of extracellular vesicles (EVs), which carry genetic and protein cargo to mediate MSC paracrine function. Recently, MSC-derived EVs have been successfully used in several preclinical models of chronic kidney disease. However, uncertainty remains regarding EV fate, safety, and long-term effects, which might impose important limitations on their path to clinical translation. This review discusses the therapeutic application of MSC-derived EV therapy for renal disease, with particular emphasis on potential mechanisms of kidney repair and major translational barriers. Emerging evidence indicates that the cargo of MSC-derived EVs is capable of modulating several pathways responsible for renal injury, including inflammation, oxidative stress, apoptosis, fibrosis, and microvascular remodeling. EV-induced modulation of these pathways has been associated with important renoprotective effects in experimental studies. However, scarce clinical data are available, and several challenges need to be addressed as we move toward clinical translation, including standardization of methods for EV isolation and characterization, EV fate, duration of EV effects, and effects of cardiovascular risk factors. MSC-derived EVs have the potential to preserve renal structure and function, but further experimental and clinical evidence is needed to confirm their protective effects in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| |
Collapse
|
14
|
Karbasiafshar C, Sellke FW, Abid MR. Mesenchymal stem cell-derived extracellular vesicles in the failing heart: past, present, and future. Am J Physiol Heart Circ Physiol 2021; 320:H1999-H2010. [PMID: 33861149 PMCID: PMC8163643 DOI: 10.1152/ajpheart.00951.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. Current treatment options include lifestyle changes, medication, and surgical intervention. However, many patients are unsuitable candidates for surgeries due to comorbidities, diffuse coronary artery disease, or advanced stages of heart failure. The search for new treatment options has recently transitioned from cell-based therapies to stem-cell-derived extracellular vesicles (EVs). A number of challenges remain in the EV field, including the effect of comorbidities, characterization, and delivery. However, recent revolutionary developments and insight into the potential of personalizing EV contents by bioengineering methods to alter specific signaling pathways in the ischemic myocardium hold promise. Here, we discuss the past limitations of cell-based therapies and recent EV studies involving in vivo, in vitro, and omics, and future challenges and opportunities in EV-based treatments in CVD.
Collapse
Affiliation(s)
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Department of Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Department of Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
15
|
Gang D, Yu CJ, Zhu S, Zhu P, Nasser MI. Application of mesenchymal stem cell-derived exosomes in kidney diseases. Cell Immunol 2021; 364:104358. [PMID: 33839596 DOI: 10.1016/j.cellimm.2021.104358] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023]
Abstract
Kidney injury (KI) has high morbidity and mortality; there has been no ideal practical treatment available in clinical practice until now. Exosomes are formed from fusing multisubunit body membranes and are secreted into the extracellular matrix, intercellular communication membracusses. As a cell-free treatment, it offers a new approach to the treatment of KI. Exosomes are spherical vesicles with or no separator cup that shapes proteins, and RNA acts on the target cells through various means to promote tissue damage and mitigate apoptosis, both inflammation and oxidative stress. Exosomes derived from mesenchymal stem cells (MSC) have a paracrine function in promoting tissue repair and immune regulation. The MSC-Exos provide specific benefits over the MSCs. The urinary exosomes closely follow the functions and diseases of the kidneys. Though much of the research in this field is only at the preliminary stages, previous research has demonstrated that MSC-Exos damaged tissues to offer proteins, mRNAs, and microRNAs as remedies for kidney injury. Although exosomes' role in tissue repair is currently is greatly debated, several key issues remain unaddressed. This is a summarization of the work done concerning MSC in the treatment of KI.
Collapse
Affiliation(s)
- Deng Gang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Chang Jiang Yu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
16
|
Metabolic Syndrome Alters the Cargo of Mitochondria-Related microRNAs in Swine Mesenchymal Stem Cell-Derived Extracellular Vesicles, Impairing Their Capacity to Repair the Stenotic Kidney. Stem Cells Int 2020; 2020:8845635. [PMID: 33281903 PMCID: PMC7685840 DOI: 10.1155/2020/8845635] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Background Coexisting metabolic syndrome (MetS) and renal artery stenosis (RAS) are linked to poor renal outcomes. Mesenchymal stem/stromal cell- (MSC-) derived extracellular vesicles (EVs) from lean animals show superior ability to repair the experimental MetS+RAS kidney compared to EVs from MetS pig MSCs. We hypothesized that MetS leads to selective packaging in porcine EVs of microRNAs capable of targeting mitochondrial genes, interfering with their capacity to repair the MetS+RAS kidney. Methods Five groups of pigs (n = 7 each) were studied after 16 weeks of diet-induced MetS and RAS (MetS+RAS) and MetS+RAS 4 weeks after a single intrarenal delivery of EVs harvested from allogeneic adipose tissue-derived MSCs isolated from Lean or MetS pigs, and Lean or MetS sham controls. Single-kidney blood flow (RBF) and glomerular filtration rate (GFR) were assessed in vivo with multidetector CT, whereas EV microRNA cargo, renal tubular mitochondrial structure and bioenergetics, and renal injury pathways were assessed ex vivo. Results microRNA sequencing revealed 19 dysregulated microRNAs capable of targeting several mitochondrial genes in MetS-EVs versus Lean-EVs. Lean- and MetS-EVs were detected in the stenotic kidney 4 weeks after administration. However, only MetS-EVs failed to improve renal mitochondrial density, structure, and function or attenuate oxidative stress, tubular injury, and fibrosis. Furthermore, Lean-EVs but not MetS-EVs restored RBF and GFR in MetS+RAS. Conclusion MetS alters the cargo of mitochondria-related microRNAs in swine MSC-derived EVs, which might impair their capacity to repair the poststenotic kidney in MetS+RAS. These observations may contribute to develop approaches to improve the efficacy of MSC-EVs for patients with MetS.
Collapse
|
17
|
Afarideh M, Thaler R, Khani F, Tang H, Jordan KL, Conley SM, Saadiq IM, Obeidat Y, Pawar AS, Eirin A, Zhu XY, Lerman A, van Wijnen AJ, Lerman LO. Global epigenetic alterations of mesenchymal stem cells in obesity: the role of vitamin C reprogramming. Epigenetics 2020; 16:705-717. [PMID: 32893712 DOI: 10.1080/15592294.2020.1819663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obesity promotes dysfunction and impairs the reparative capacity of mesenchymal stem/stromal cells (MSCs), and alters their transcription, protein content, and paracrine function. Whether these adverse effects are mediated by chromatin-modifying epigenetic changes remains unclear. We tested the hypothesis that obesity imposes global DNA hydroxymethylation and histone tri-methylation alterations in obese swine abdominal adipose tissue-derived MSCs compared to lean pig MSCs. MSCs from female lean (n = 7) and high-fat-diet fed obese (n = 7) domestic pigs were assessed using global epigenetic assays, before and after in-vitro co-incubation with the epigenetic modulator vitamin-C (VIT-C) (50 μg/ml). Dot blotting was used to measure across the whole genome 5-hydroxyemthycytosine (5hmC) residues, and Western blotting to quantify in genomic histone-3 protein tri-methylated lysine-4 (H3K4me3), lysine-9 (H3K9me3), and lysine-27 (H3K27me3) residues. MSC migration and proliferation were studied in-vitro. Obese MSCs displayed reduced global 5hmC and H3K4m3 levels, but comparable H3K9me3 and H3K27me3, compared to lean MSCs. Global 5hmC, H3K4me3, and HK9me3 marks correlated with MSC migration and reduced proliferation, as well as clinical and metabolic characteristics of obesity. Co-incubation of obese MSCs with VIT-C enhanced 5hmC marks, and reduced their global levels of H3K9me3 and H3K27me3. Contrarily, VIT-C did not affect 5hmC, and decreased H3K4me3 in lean MSCs. Obesity induces global genomic epigenetic alterations in swine MSCs, involving primarily genomic transcriptional repression, which are associated with MSC function and clinical features of obesity. Some of these alterations might be reversible using the epigenetic modulator VIT-C, suggesting epigenetic modifications as therapeutic targets in obesity.
Collapse
Affiliation(s)
- Mohsen Afarideh
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, and Department of Biochemistry, and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Farzaneh Khani
- Department of Orthopedic Surgery, and Department of Biochemistry, and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Sabena M Conley
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Yasin Obeidat
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Aditya S Pawar
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, and Department of Biochemistry, and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
18
|
Gardin C, Ferroni L, Chachques JC, Zavan B. Could Mesenchymal Stem Cell-Derived Exosomes Be a Therapeutic Option for Critically Ill COVID-19 Patients? J Clin Med 2020; 9:E2762. [PMID: 32858940 PMCID: PMC7565764 DOI: 10.3390/jcm9092762] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic viral disease originated in Wuhan, China, in December 2019, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The severe form of the disease is often associated with acute respiratory distress syndrome (ARDS), and most critically ill patients require mechanical ventilation and support in intensive care units. A significant portion of COVID-19 patients also develop complications of the cardiovascular system, primarily acute myocardial injury, arrhythmia, or heart failure. To date, no specific antiviral therapy is available for patients with SARS-CoV-2 infection. Exosomes derived from mesenchymal stem cells (MSCs) are being explored for the management of a number of diseases that currently have limited or no therapeutic options, thanks to their anti-inflammatory, immunomodulatory, and pro-angiogenic properties. Here, we briefly introduce the pathogenesis of SARS-CoV-2 and its implications in the heart and lungs. Next, we describe some of the most significant clinical evidence of the successful use of MSC-derived exosomes in animal models of lung and heart injuries, which might strengthen our hypothesis in terms of their utility for also treating critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Morphology, Experimental Medicine and Surgery, University of Ferrara, via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Letizia Ferroni
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Morphology, Experimental Medicine and Surgery, University of Ferrara, via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Juan Carlos Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015 Paris, France;
| | - Barbara Zavan
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy; (C.G.); (L.F.)
- Department of Morphology, Experimental Medicine and Surgery, University of Ferrara, via Fossato di Mortara 70, 44121 Ferrara, Italy
| |
Collapse
|
19
|
Song T, Eirin A, Zhu X, Zhao Y, Krier JD, Tang H, Jordan KL, Woollard JR, Taner T, Lerman A, Lerman LO. Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Regulatory T Cells to Ameliorate Chronic Kidney Injury. Hypertension 2020; 75:1223-1232. [PMID: 32223383 DOI: 10.1161/hypertensionaha.119.14546] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic syndrome (MetS) profoundly changes the contents of mesenchymal stem cells and mesenchymal stem cells-derived extracellular vesicles (EVs). The anti-inflammatory TGF-β (transforming growth factor-β) is selectively enriched in EVs from Lean but not from MetS pigs, but the functional impact of this endowment remains unknown. We hypothesized that Lean-EVs more effectively induce regulatory T cells in injured kidneys. Five groups of pigs (n=7 each) were studied after 16 weeks of diet-induced MetS and unilateral renal artery stenosis (RAS; MetS+RAS). Two groups of MetS+RAS were treated 4 weeks earlier with an intrarenal injection of either Lean-EVs or MetS-EVs. MetS+RAS had lower renal volume, renal blood flow, and glomerular filtration rate than MetS pigs. Compared with Lean-EVs, MetS-EVs were less effective in improving renal function and decreasing tubular injury and fibrosis in MetS+RAS. Lean-EVs upregulated TGF-β expression in stenotic kidney and increased regulatory T cells numbers more prominently. Furthermore, markedly upregulated anti-inflammatory M2 macrophages reduced proinflammatory M1 macrophages, and CD8+ T cells were detected in stenotic kidneys treated with Lean-EVs compared with MetS-EVs, and renal vein levels of interleukin-1β were reduced. In vitro, coculture of Lean-EVs with activated T cells led to greater TGF-β-dependent regulatory T cells induction than did MetS-EVs. Therefore, the beneficial effects of mesenchymal stem cells-derived EVs on injured kidneys might be partly mediated by their content of TGF-β signaling components, which permitting increased Treg preponderance. Modulating EV cargo and transforming their functionality might be useful for renal repair.
Collapse
Affiliation(s)
- Turun Song
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Alfonso Eirin
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Xiangyang Zhu
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Yu Zhao
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - James D Krier
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Hui Tang
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Kyra L Jordan
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - John R Woollard
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| | - Timucin Taner
- Department of Transplant Surgery and Immunology (T.T.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Diseases (A.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Division of Nephrology and Hypertension (T.S., A.E., X.Z., Y.Z., J.D.K., H.T., K.L.J., J.R.W., L.O.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
20
|
Pawar AS, Eirin A, Tang H, Zhu XY, Lerman A, Lerman LO. Upregulated tumor necrosis factor-α transcriptome and proteome in adipose tissue-derived mesenchymal stem cells from pigs with metabolic syndrome. Cytokine 2020; 130:155080. [PMID: 32240922 PMCID: PMC7529712 DOI: 10.1016/j.cyto.2020.155080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/04/2020] [Accepted: 03/21/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) have endogenous reparative properties, and may constitute an exogenous therapeutic intervention in patients with chronic kidney disease. The microenvironment of metabolic syndrome (MetS) induces fat inflammation, with abundant expression of tumor necrosis factor (TNF)-α. MetS may also alter the content of adipose tissue-derived MSCs, and we hypothesized that the inflammatory profile of MetS manifests via upregulating MSC mRNAs and proteins of the TNF-α pathway. METHODS Domestic pigs were fed a 16-week Lean or MetS diet (n = 4 each). MSCs were harvested from abdominal subcutaneous fat, and their extracellular vesicles (EVs) isolated. Expression profiles of mRNAs and proteins in MSCs and EVs were obtained by high-throughput sequencing and proteomics. Nuclear translocation of the pro-inflammatory transcription factor (NF)-kB was evaluated in MSC and in pig renal tubular cells (TEC) co-incubated with EVs. RESULTS We found 13 mRNAs and 4 proteins in the TNF-α pathway upregulated in MetS- vs. Lean-MSCs (fold-change > 1.4, p < 0.05), mostly via TNF-α receptor-1 (TNF-R1) signaling. Three mRNAs were upregulated in MetS-EVs. MetS-MSCs, as well as TECs co-incubated with MetS-EVs, showed increased nuclear translocation of NF-kB. Using qPCR, JUNB, MAP2K7 and TRAF2 genes followed the same direction of RNA-sequencing findings. CONCLUSIONS MetS upregulates the TNF-α transcriptome and proteome in swine adipose tissue-derived MSCs, which are partly transmitted to their EV progeny, and are associated with activation of NF-kB in target cells. Hence, the MetS milieu may affect the profile of endogenous MSCs and their paracrine vectors and limit their use as an exogenous regenerative therapy. Anti-inflammatory strategies targeting the TNF-α pathway might be a novel strategy to restore MSC phenotype, and in turn function.
Collapse
Affiliation(s)
- Aditya S Pawar
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States; Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
21
|
Kaddour H, Lyu Y, Welch JL, Paromov V, Mandape SN, Sakhare SS, Pandhare J, Stapleton JT, Pratap S, Dash C, Okeoma CM. Proteomics Profiling of Autologous Blood and Semen Exosomes from HIV-infected and Uninfected Individuals Reveals Compositional and Functional Variabilities. Mol Cell Proteomics 2020; 19:78-100. [PMID: 31676584 PMCID: PMC6944229 DOI: 10.1074/mcp.ra119.001594] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 10/18/2019] [Indexed: 12/18/2022] Open
Abstract
Blood and semen are important body-fluids that carry exosomes for bioinformation transmission. Therefore, characterization of their proteomes is necessary for understanding body-fluid-specific physiologic and pathophysiologic functions. Using systematic multifactorial proteomic profiling, we characterized the proteomes of exosomes and exosome-free fractions from autologous blood and semen from three HIV-uninfected and three HIV-infected participants (total of 24 samples). We identified exosome-based protein signatures specific to blood and semen along with HIV-induced tissue-dependent proteomic perturbations. We validated our findings with samples from 16 additional donors and showed that unlike blood exosomes (BE), semen exosomes (SE) are enriched in clusterin. SE but not BE promote Protein·Nucleic acid binding and increase cell adhesion irrespective of HIV infection. This is the first comparative study of the proteome of autologous BE and SE. The proteins identified may be developed as biomarkers applicable to different fields of medicine, including reproduction and infectious diseases.
Collapse
Affiliation(s)
- Hussein Kaddour
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, New York 11794-8651
| | - Yuan Lyu
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, New York 11794-8651
| | - Jennifer L Welch
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
| | - Victor Paromov
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee 37208
| | - Sammed N Mandape
- College of Medicine, The University of Arizona Health Sciences, Tucson, Arizona 85721
| | - Shruti S Sakhare
- University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Jui Pandhare
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee 37208
| | - Jack T Stapleton
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242-1109
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee 37208
| | - Chandravanu Dash
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, Tennessee 37208
| | - Chioma M Okeoma
- Department of Pharmacology, Stony Brook University Renaissance School of Medicine, Stony Brook, New York 11794-8651.
| |
Collapse
|
22
|
Qiu G, Zheng G, Ge M, Wang J, Huang R, Shu Q, Xu J. Functional proteins of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res Ther 2019; 10:359. [PMID: 31779700 PMCID: PMC6883709 DOI: 10.1186/s13287-019-1484-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/29/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) contain proteins, microRNAs, mRNAs, long non-coding RNAs, and phospholipids, and are a novel mechanism of intercellular communication. It has been proposed that the immunomodulatory and regenerative effects of mesenchymal stem/stromal cells (MSCs) are mainly mediated by soluble paracrine factors and MSC-derived EVs (MSC-EVs). Recent studies suggest that MSC-EVs may serve as a novel and cell-free alternative to whole-cell therapies. The focus of this review is to discuss the functional proteins which facilitate the effects of MSC-EVs. The first section of the review discusses the general functions of EV proteins. Next, we describe the proteomics of MSC-EVs as compared with their parental cells. Then, the review presents the current knowledge that protein contents of MSC-EVs play an essential role in immunomodulation and treatment of various diseases. In summary, functional protein components are at least partially responsible for disease-modulating capacity of MSC-EVs.
Collapse
Affiliation(s)
- Guanguan Qiu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, 312000, Zhejiang, China
| | - Guoping Zheng
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, 312000, Zhejiang, China
| | - Menghua Ge
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, 312000, Zhejiang, China
| | - Jiangmei Wang
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Ruoqiong Huang
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Qiang Shu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, 310051, Zhejiang, China.
| | - Jianguo Xu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, 312000, Zhejiang, China. .,The First Affiliated Hospital of Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
23
|
Eirin A, Lerman LO. Stem cell-derived extracellular vesicles for renal repair: do cardiovascular comorbidities matter? Am J Physiol Renal Physiol 2019; 317:F1414-F1419. [PMID: 31630544 DOI: 10.1152/ajprenal.00434.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicle (EV)-based regenerative therapy has shown promising results in preclinical models of renal disease and might be useful for patients with several forms of chronic kidney disease. However, individuals with chronic kidney disease often present with comorbidities, including obesity, hypertension, diabetes, or even metabolic syndrome, which may alter the endogenous characteristics and impair the reparative capacity of stem cells and their daughter EVs. This brief review summarizes evidence of alterations in the morphology, cargo, and function of mesenchymal stem cells and mesenchymal stem cell-derived EVs in the face of cardiovascular disease. We further discuss the important ramifications for their use in patients with kidney disease.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
24
|
Conley SM, Shook JE, Zhu XY, Eirin A, Jordan KL, Woollard JR, Isik B, Hickson LJ, Puranik AS, Lerman LO. Metabolic Syndrome Induces Release of Smaller Extracellular Vesicles from Porcine Mesenchymal Stem Cells. Cell Transplant 2019; 28:1271-1278. [PMID: 31250656 PMCID: PMC6767891 DOI: 10.1177/0963689719860840] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) belong to the endogenous cellular reparative
system, and can be used exogenously in cell-based therapy. MSCs release extracellular
vesicles (EVs), including exosomes and microvesicles, which mediate some of their
therapeutic activity through intercellular communication. We have previously demonstrated
that metabolic syndrome (MetS) modifies the cargo packed within swine EV, but whether it
influences their phenotypical characteristics remains unclear. This study tested the
hypothesis that MetS shifts the size distribution of MSC-derived EVs. Adipose
tissue-derived MSC-EV subpopulations from Lean (n = 6) and MetS
(n = 6) pigs were characterized for number and size using
nanoparticle-tracking analysis, flow cytometry, and transmission electron microscopy.
Expression of exosomal genes was determined using next-generation RNA-sequencing
(RNA-seq). The number of EV released from Lean and MetS pig MSCs was similar, yet
MetS-MSCs yielded a higher proportion of small-size EVs (202.4 ± 17.7 nm vs. 280.3 ± 15.1
nm), consistent with exosomes. RNA-seq showed that their EVs were enriched with exosomal
markers. Lysosomal activity remained unaltered in MetS-MSCs. Therefore, MetS alters the
size distribution of MSC-derived EVs in favor of exosome release. These observations may
reflect MSC injury and membrane recycling in MetS or increased expulsion of waste
products, and may have important implications for development of adequate cell-based
treatments.
Collapse
Affiliation(s)
- Sabena M Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - John E Shook
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA.,Department of Cardiothoracic Surgery, University of Nebraska Medical Center, Omaha, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - John R Woollard
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Busra Isik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| | - Amrutesh S Puranik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA.,Division of Rheumatology, Colton Center for Autoimmunity, New York University Langone Medical Center, New York, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| |
Collapse
|