1
|
Imaralu OE, Aluganti Narasimhulu C, Singal PK, Singla DK. Role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in diabetic complications. Can J Physiol Pharmacol 2024; 102:14-25. [PMID: 37748207 DOI: 10.1139/cjpp-2023-0223] [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: 09/27/2023]
Abstract
Cardiovascular disease (CVD) complications have remained a major cause of death among patients with diabetes. Hence, there is a need for effective therapeutics against diabetes-induced CVD complications. Since its discovery, proprotein convertase subtilisin/kexin type 9 (PCSK9) has been reported to be involved in the pathology of various CVDs, with studies showing a positive association between plasma levels of PCSK9, hyperglycemia, and dyslipidemia. PCSK9 regulates lipid homeostasis by interacting with low-density lipoprotein receptors (LDLRs) present in hepatocytes and subsequently induces LDLR degradation via receptor-mediated endocytosis, thereby reducing LDL uptake from circulation. In addition, PCSK9 also induces pro-inflammatory cytokine expression and apoptotic cell death in diabetic-CVD. Furthermore, therapies designed to inhibit PCSK9 effectively reduces diabetic dyslipidemia with clinical studies reporting reduced cardiovascular events in patients with diabetes and no significant adverse effect on glycemic controls. In this review, we discuss the role of PCSK9 in the pathogenesis of diabetes-induced CVD and the potential mechanisms by which PCSK9 inhibition reduces cardiovascular events in diabetic patients.
Collapse
Affiliation(s)
- Omonzejie E Imaralu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Chandrakala Aluganti Narasimhulu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Pawan K Singal
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| |
Collapse
|
2
|
Zhang H, Shen Y, Kim IM, Weintraub NL, Tang Y. The Impaired Bioenergetics of Diabetic Cardiac Microvascular Endothelial Cells. Front Endocrinol (Lausanne) 2021; 12:642857. [PMID: 34054724 PMCID: PMC8160466 DOI: 10.3389/fendo.2021.642857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/06/2021] [Indexed: 01/22/2023] Open
Abstract
Diabetes causes hyperglycemia, which can create a stressful environment for cardiac microvascular endothelial cells (CMECs). To investigate the impact of diabetes on the cellular metabolism of CMECs, we assessed glycolysis by quantifying the extracellular acidification rate (ECAR), and mitochondrial oxidative phosphorylation (OXPHOS) by measuring cellular oxygen consumption rate (OCR), in isolated CMECs from wild-type (WT) hearts and diabetic hearts (db/db) using an extracellular flux analyzer. Diabetic CMECs exhibited a higher level of intracellular reactive oxygen species (ROS), and significantly reduced glycolytic reserve and non-glycolytic acidification, as compared to WT CMECs. In addition, OCR assay showed that diabetic CMECs had increased maximal respiration, and significantly reduced non-mitochondrial oxygen consumption and proton leak. Quantitative PCR (qPCR) showed no difference in copy number of mitochondrial DNA (mtDNA) between diabetic and WT CMECs. In addition, gene expression profiling analysis showed an overall decrease in the expression of essential genes related to β-oxidation (Sirt1, Acox1, Acox3, Hadha, and Hadhb), tricarboxylic acid cycle (TCA) (Idh-3a and Ogdh), and electron transport chain (ETC) (Sdhd and Uqcrq) in diabetic CMECs compared to WT CMECs. Western blot confirmed that the protein expression of Hadha, Acox1, and Uqcrq was decreased in diabetic CMECs. Although lectin staining demonstrated no significant difference in capillary density between the hearts of WT mice and db/db mice, diabetic CMECs showed a lower percentage of cell proliferation by Ki67 staining, and a higher percentage of cellular apoptosis by TUNEL staining, compared with WT CMECs. In conclusion, excessive ROS caused by hyperglycemia is associated with impaired glycolysis and mitochondrial function in diabetic CMECs, which in turn may reduce proliferation and promote CMEC apoptosis.
Collapse
Affiliation(s)
- Haitao Zhang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Il-man Kim
- Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- *Correspondence: Yaoliang Tang,
| |
Collapse
|
3
|
MMP9 mediates acute hyperglycemia-induced human cardiac stem cell death by upregulating apoptosis and pyroptosis in vitro. Cell Death Dis 2020; 11:186. [PMID: 32170070 PMCID: PMC7070071 DOI: 10.1038/s41419-020-2367-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022]
Abstract
Providing a conducive microenvironment is critical to increase survival of transplanted stem cells in regenerative therapy. Hyperglycemia promotes stem cell death impairing cardiac regeneration in the diabetic heart. Understanding the molecular mechanisms of high glucose-induced stem cell death is important for improving cardiac regeneration in diabetic patients. Matrix metalloproteinase-9 (MMP9), a collagenase, is upregulated in the diabetic heart, and ablation of MMP9 decreases infarct size in the non-diabetic myocardial infarction heart. In the present study, we aim to investigate whether MMP9 is a mediator of hyperglycemia-induced cell death in human cardiac stem cells (hCSCs) in vitro. We created MMP9−/− hCSCs to test the hypothesis that MMP9 mediates hyperglycemia-induced oxidative stress and cell death via apoptosis and pyroptosis in hCSCs, which is attenuated by the lack of MMP9. We found that hyperglycemia induced oxidative stress and increased cell death by promoting pyroptosis and apoptosis in hCSCs, which was prevented in MMP9−/− hCSCs. These findings revealed a novel intracellular role of MMP9 in mediating stem cell death and provide a platform to assess whether MMP9 inhibition could improve hCSCs survival in stem cell therapy at least in acute hyperglycemic microenvironment.
Collapse
|
4
|
Peng BY, Dubey NK, Mishra VK, Tsai FC, Dubey R, Deng WP, Wei HJ. Addressing Stem Cell Therapeutic Approaches in Pathobiology of Diabetes and Its Complications. J Diabetes Res 2018; 2018:7806435. [PMID: 30046616 PMCID: PMC6036791 DOI: 10.1155/2018/7806435] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 04/19/2018] [Accepted: 05/27/2018] [Indexed: 12/14/2022] Open
Abstract
High morbidity and mortality of diabetes mellitus (DM) throughout the human population is a serious threat which needs to be addressed cautiously. Type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) are most prevalent forms. Disruption in insulin regulation and resistance leads to increased formation and accumulation of advanced end products (AGEs), which further enhance oxidative and nitrosative stress leading to microvascular (retinopathy, neuropathy, and nephropathy) and macrovascular complications. These complications affect the normal function of organ and tissues and may cause life-threatening disorders, if hyperglycemia persists and improperly controlled. Current and traditional treatment procedures are only focused on to regulate the insulin level and do not cure the diabetic complications. Pancreatic transplantation seemed a viable alternative; however, it is limited due to lack of donors. Cell-based therapy such as stem cells is considered as a promising therapeutic agent against DM and diabetic complications owing to their multilineage differentiation and regeneration potential. Previous studies have demonstrated the various impacts of both pluripotent and multipotent stem cells on DM and its micro- and macrovascular complications. Therefore, this review summarizes the potential of stem cells to treat DM and its related complications.
Collapse
Affiliation(s)
- Bou-Yue Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Department of Dentistry, Taipei Medical University Hospital, Taipei City 110, Taiwan
| | - Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala, India
| | - Feng-Chou Tsai
- Department of Stem Cell Research, Cosmetic Clinic Group, Taipei City 110, Taiwan
| | - Rajni Dubey
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei City 106, Taiwan
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Hong-Jian Wei
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
| |
Collapse
|
5
|
Mayorga ME, Kiedrowski M, McCallinhart P, Forudi F, Ockunzzi J, Weber K, Chilian W, Penn MS, Dong F. Role of SDF-1:CXCR4 in Impaired Post-Myocardial Infarction Cardiac Repair in Diabetes. Stem Cells Transl Med 2017; 7:115-124. [PMID: 29119710 PMCID: PMC5746149 DOI: 10.1002/sctm.17-0172] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/06/2017] [Indexed: 12/13/2022] Open
Abstract
Diabetes is a risk factor for worse outcomes following acute myocardial infarction (AMI). In this study, we tested the hypothesis that SDF‐1:CXCR4 expression is compromised in post‐AMI in diabetes, and that reversal of this defect can reverse the adverse effects of diabetes. Mesenchymal stem cells (MSC) isolated from green fluorescent protein (GFP) transgenic mice (control MSC) were induced to overexpress stromal cell‐derived factor‐1 (SDF‐1). SDF‐1 expression in control MSC and SDF‐1‐overexpressing MSC (SDF‐1:MSC) were quantified using enzyme‐linked immunosorbent assay (ELISA). AMI was induced on db/db and control mice. Mice were randomly selected to receive infusion of control MSC, SDF‐1:MSC, or saline into the border zone after AMI. Serial echocardiography was used to assess cardiac function. SDF‐1 and CXCR4 mRNA expression in the infarct zone of db/db mice and control mice were quantified. Compared to control mice, SDF‐1 levels were decreased 82%, 91%, and 45% at baseline, 1 day and 3 days post‐AMI in db/db mice, respectively. CXCR4 levels are increased 233% at baseline and 54% 5 days post‐AMI in db/db mice. Administration of control MSC led to a significant improvement in ejection fraction (EF) in control mice but not in db/db mice 21 days after AMI. In contrast, administration of SDF‐1:MSC produced a significant improvement in EF in both control mice and db/db mice 21 days after AMI. The SDF‐1:CXCR4 axis is compromised in diabetes, which appears to augment the deleterious consequences of AMI. Over‐express of SDF‐1 expression in diabetes rescues cardiac function post AMI. Our results suggest that modulation of SDF‐1 may improve post‐AMI cardiac repair in diabetes. stemcellstranslationalmedicine2018;7:115–124
Collapse
Affiliation(s)
- Maritza E Mayorga
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Matthew Kiedrowski
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Patricia McCallinhart
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Farhad Forudi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Jeremiah Ockunzzi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Kristal Weber
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - William Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Marc S Penn
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA.,Summa Cardiovascular Institute, Summa Health System, Akron, Ohio, USA
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
6
|
Fibroblast Growth Factor-9 Activates c-Kit Progenitor Cells and Enhances Angiogenesis in the Infarcted Diabetic Heart. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5810908. [PMID: 26682010 PMCID: PMC4670684 DOI: 10.1155/2016/5810908] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/09/2015] [Indexed: 12/20/2022]
Abstract
We hypothesized that fibroblast growth factor-9 (FGF-9) would enhance angiogenesis via activating c-kit positive stem cells in the infarcted nondiabetic and diabetic heart. In brief, animals were divided into three groups: Sham, MI, and MI+FGF-9. Two weeks following MI or sham surgery, our data suggest that treatment with FGF-9 significantly diminished vascular apoptosis compared to the MI group in both C57BL/6 and db/db mice (p < 0.05). Additionally, the number of c-kit+ve/SM α-actin+ve cells and c-kit+ve/CD31+ve cells were greatly enhanced in the MI+FGF-9 groups relative to the MI suggesting FGF-9 enhances c-Kit cell activation and their differentiation into vascular smooth muscle cells and endothelial cells, respectively (p < 0.05). Histology shows that the total number of vessels were quantified for all groups and our data suggest that the FGF-9 treated groups had significantly more vessels than their MI counterparts (p < 0.05). Finally, echocardiographic data suggests a significant improvement in left ventricular output, as indicated by fractional shortening and ejection fraction in both nondiabetic and diabetic animals treated with FGF-9 (p < 0.05). Overall, our data suggests FGF-9 has the potential to attenuate vascular cell apoptosis, activate c-Kit progenitor cells, and enhance angiogenesis and neovascularization in C57BL/6 and db/db mice leading to improved cardiac function.
Collapse
|
7
|
Abstract
The prevalence of diabetes continues to increase world-wide and is a leading cause of morbidity, mortality, and rapidly rising health care costs. Although strict glucose control combined with good pharmacological and non-pharmacologic interventions can increase diabetic patient life span, the frequency and mortality of myocardial ischemia and infarction remain drastically increased in diabetic patients. Therefore, more effective therapeutic approaches are urgently needed. Over the past 15 years, cellular repair of the injured adult heart has become the focus of a rapidly expanding broad spectrum of pre-clinical and clinical research. Recent clinical trials have achieved favorable initial endpoints with improvements in cardiac function and clinical symptoms following cellular therapy. Due to the increased risk of cardiac disease, cardiac regeneration may be one strategy to treat patients with diabetic cardiomyopathy and/or myocardial infarction. However, pre-clinical studies suggest that the diabetic myocardium may not be a favorable environment for the transplantation and survival of stem cells due to altered kinetics in cellular homing, survival, and in situ remodeling. Therefore, unique conditions in the diabetic myocardium will require novel solutions in order to increase the efficiency of cellular repair following ischemia and/or infarction. This review briefly summarizes some of the recent advances in cardiac regeneration in non-diabetic conditions and then provides an overview of some of the issues related to diabetes that must be addressed in the coming years.
Collapse
Affiliation(s)
- Lu Cai
- Kosair Children's Hospital Research Institute, Louisville, KY USA ; Department of Pediatrics, University of Louisville, Louisville, KY USA
| | - Bradley B Keller
- Department of Pediatrics, University of Louisville, Louisville, KY USA ; Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky USA
| |
Collapse
|
8
|
Gili M, Orsello A, Gallo S, Brizzi MF. Diabetes-associated macrovascular complications: cell-based therapy a new tool? Endocrine 2013; 44:557-75. [PMID: 23543434 DOI: 10.1007/s12020-013-9936-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/20/2013] [Indexed: 01/01/2023]
Abstract
Diabetes mellitus and its ongoing macrovascular complications represent one of the major health problems around the world. Rise in obesity and population ages correlate with the increased incidence of diabetes. This highlights the need for novel approaches to prevent and treat this pandemic. The discovery of a reservoir of stem/progenitors in bone marrow and in mesenchymal tissue has attracted interest of both biologists and clinicians. A number of preclinical and clinical trials were developed to explore their potential clinical impact, as target or vehicle, in different clinical settings, including diabetes complications. Currently, bone marrow, peripheral blood, mesenchymal, and adipose tissues have been used as stem/progenitor cell sources. However, evidences have been provided that both bone marrow and circulating progenitor cells are dysfunctional in diabetes. These observations along with the growing advantages in genetic manipulation have spurred researchers to exploit ex vivo manipulated cells to overcome these hurdles. In this article, we provide an overview of data relevant to stem-progenitors potential clinical application in revascularization and/or vascular repair. Moreover, the hurdles at using progenitor cells in diabetic patients will be also discussed.
Collapse
Affiliation(s)
- Maddalena Gili
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | | | | | | |
Collapse
|
9
|
Hughey CC, Ma L, James FD, Bracy DP, Wang Z, Wasserman DH, Rottman JN, Hittel DS, Shearer J. Mesenchymal stem cell transplantation for the infarcted heart: therapeutic potential for insulin resistance beyond the heart. Cardiovasc Diabetol 2013; 12:128. [PMID: 24007410 PMCID: PMC3847505 DOI: 10.1186/1475-2840-12-128] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/30/2013] [Indexed: 12/28/2022] Open
Abstract
Background This study aimed to evaluate the efficacy of mesenchymal stem cell (MSC) transplantation to mitigate abnormalities in cardiac-specific and systemic metabolism mediated by a combination of a myocardial infarction and diet-induced insulin resistance. Methods C57BL/6 mice were high-fat fed for eight weeks prior to induction of a myocardial infarction via chronic ligation of the left anterior descending coronary artery. MSCs were administered directly after myocardial infarction induction through a single intramyocardial injection. Echocardiography was performed prior to the myocardial infarction as well as seven and 28 days post-myocardial infarction. Hyperinsulinemic-euglycemic clamps coupled with 2-[14C]deoxyglucose were employed 36 days post-myocardial infarction (13 weeks of high-fat feeding) to assess systemic insulin sensitivity and insulin-mediated, tissue-specific glucose uptake in the conscious, unrestrained mouse. High-resolution respirometry was utilized to evaluate cardiac mitochondrial function in saponin-permeabilized cardiac fibers. Results MSC administration minimized the decline in ejection fraction following the myocardial infarction. The greater systolic function in MSC-treated mice was associated with increased in vivo cardiac glucose uptake and enhanced mitochondrial oxidative phosphorylation efficiency. MSC therapy promoted reductions in fasting arterial glucose and fatty acid concentrations. Additionally, glucose uptake in peripheral tissues including skeletal muscle and adipose tissue was elevated in MSC-treated mice. Enhanced glucose uptake in these tissues was associated with improved insulin signalling as assessed by Akt phosphorylation and prevention of a decline in GLUT4 often associated with high-fat feeding. Conclusions These studies provide insight into the utility of MSC transplantation as a metabolic therapy that extends beyond the heart exerting beneficial systemic effects on insulin action.
Collapse
Affiliation(s)
- Curtis C Hughey
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, 2500 University Drive N,W,, Calgary, AB, Canada, T2N 1N4.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Yan B, Singla DK. Transplanted induced pluripotent stem cells mitigate oxidative stress and improve cardiac function through the Akt cell survival pathway in diabetic cardiomyopathy. Mol Pharm 2013; 10:3425-32. [PMID: 23879836 DOI: 10.1021/mp400258d] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence suggests transplanted stem cells improve left ventricular function in diabetic induced cardiomyopathy (DICM). However, little is known about the mechanisms by which induced pluripotent stem (iPS) cells or factors released from these cells inhibit adverse cardiac remodeling in DICM. The present study was designed to determine molecular mediators and pathways regulated by transplanted iPS cells and their conditioned media (CM) in DICM. Animals were divided into four experimental groups such as control, streptozotocin (STZ), STZ+iPS-CM, and STZ+iPS cells. Experimental diabetes was induced in C57BL/6 mice by intraperitoneal STZ injections (100 mg/kg body weight for 2 consecutive days). Following STZ injections, iPS cells or CM was given intravenously for 3 consecutive days. Animals were humanely killed, and hearts were harvested at D14. Animals transplanted with iPS cells or CM demonstrated a significant reduction in apoptosis, mediated by Akt upregulation and ERK1/2 downregulation, and inhibition of interstitial fibrosis via MMP-9 suppression compared with the STZ group. Oxidative stress was significantly hindered in iPS cell and CM groups as evidenced by diminished pro-oxidant expression and enhanced antioxidant (catalase and MnSOD) concentration. Echocardiography data suggest a significant improvement in cardiac function in cells and CM groups in comparison to STZ. In conclusion, our data strongly suggest that iPS cells and CM attenuate oxidative stress and associated apoptosis and fibrosis. Moreover, we also suggest that increased antioxidant levels, decreased adverse cardiac remodeling, and improved cardiac function is mediated by iPS CM and cells in DICM through multiple autocrine and paracrine mechanisms.
Collapse
Affiliation(s)
- Binbin Yan
- Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida , Orlando, Florida 32816, United States
| | | |
Collapse
|
11
|
Peng Y, Huang S, Cheng B, Nie X, Enhe J, Feng C, Fu X. Mesenchymal stem cells: a revolution in therapeutic strategies of age-related diseases. Ageing Res Rev 2013; 12:103-15. [PMID: 22569401 DOI: 10.1016/j.arr.2012.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/11/2012] [Accepted: 04/23/2012] [Indexed: 02/06/2023]
Abstract
The great evolutionary biologist Theodosius Dobzhansky once said: "Nothing in biology makes sense except in the light of evolution". Aging is a complex biological phenomenon and the factors governing the process of aging and age-related diseases are only beginning to be understood, oxidative stress, telomere shortening in DNA components and genetic changes were shown to be the mainly regulating mechanisms during the recent decades. Although a considerable amount of both animal and clinical data that demonstrate the extensive and safe use of mesenchymal stromal cells (MSCs) is available, the precise summarization and identification of MSCs in age-related diseases remains a challenge. Along this line, this review discussed several typical age-related diseases for which MSCs have been proved to confer protection and put forward a hypothesis for the association among MSCs and age-related diseases from an evolutionary perspective. Above all, we hope further and more research efforts could be aroused to elucidate the role and mechanisms that MSCs involved in the age-related diseases.
Collapse
|
12
|
Zeng H, Li L, Chen JX. Overexpression of angiopoietin-1 increases CD133+/c-kit+ cells and reduces myocardial apoptosis in db/db mouse infarcted hearts. PLoS One 2012; 7:e35905. [PMID: 22558265 PMCID: PMC3338852 DOI: 10.1371/journal.pone.0035905] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/23/2012] [Indexed: 12/29/2022] Open
Abstract
Hematopoietic progenitor CD133(+)/c-kit(+) cells have been shown to be involved in myocardial healing following myocardial infarction (MI). Previously we demonstrated that angiopoietin-1(Ang-1) is beneficial in the repair of diabetic infarcted hearts. We now investigate whether Ang-1 affects CD133(+)/c-kit(+) cell recruitment to the infarcted myocardium thereby mediating cardiac repair in type II (db/db) diabetic mice. db/db mice were administered either adenovirus Ang-1 (Ad-Ang-1) or Ad-β-gal systemically immediately after ligation of the left anterior descending coronary artery (LAD). Overexpression of Ang-1 resulted in a significant increase in CXCR-4/SDF-1α expression and promoted CD133(+)/c-kit(+), CD133(+)/CXCR-4(+) and CD133(+)/SDF-1α(+) cell recruitment into ischemic hearts. Overexpression of Ang-1 led to significant increases in number of CD31(+) and smooth muscle-like cells and VEGF expression in bone marrow (BM). This was accompanied by significant decreases in cardiac apoptosis and fibrosis and an increase in myocardial capillary density. Ang-1 also upregulated Jagged-1, Notch3 and apelin expression followed by increases in arteriole formation in the infarcted myocardium. Furthermore, overexpression of Ang-1 resulted in a significant improvement of cardiac functional recovery after 14 days of ischemia. Our data strongly suggest that Ang-1 attenuates cardiac apoptosis and promotes cardiac repair by a mechanism involving in promoting CD133(+)/c-kit(+) cells and angiogenesis in diabetic db/db mouse infarcted hearts.
Collapse
Affiliation(s)
- Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Lanfang Li
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| |
Collapse
|
13
|
Neel S, Singla DK. Induced pluripotent stem (iPS) cells inhibit apoptosis and fibrosis in streptozotocin-induced diabetic rats. Mol Pharm 2011; 8:2350-7. [PMID: 21988648 DOI: 10.1021/mp2004675] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent data suggests that transplanted bone marrow stem cells improve cardiac function in streptozotocin induced diabetic cardiomyopathy (SIDC). However, whether modified embryonic stem (ES) cells, induced pluripotent stem (iPS) cells, or factors released from these cells can inhibit apoptosis and fibrosis remains completely unknown. Therefore, we hypothesized that transplanted ES cells overexpressing pancreatic transcription factor 1 a (Ptf1a), a propancreatic endodermal transcription factor, iPS cells, or their respective conditioned media (CM) will attenuate cardiac remodeling and improve cardiac function in SIDC. Experimental diabetes was induced in male Sprague-Dawley rats (8-10 weeks old) by intraperitoneal injections of streptozotocin (STZ) (65 mg/kg body weight). Animals were divided into different groups including control, STZ, stem cells, and CM. Histology, TUNEL, caspase-3 activity, sarcomeric α-actin, and DHE stainings were performed to assess cardiac apoptosis, fibrosis, and oxidative stress. Animals transplanted with ES cells, iPS cells, or both CM showed a significant (p < 0.05) reduction in apoptosis compared with STZ treated animals. Furthermore, our data also shows that active apoptosis was present in cardiac myocytes as confirmed with combined stainings with TUNEL, sarcomeric α-actin, and active caspase-3 antibodies. Increased oxidative stress as evidenced by DHE staining was significantly (p < 0.05) reduced following stem cell or CM transplantation. Moreover, stem cells or CM also attenuated increased interstitial and vascular fibrosis in SIDC hearts. Echocardiography analysis showed a significant (p < 0.05) improvement in fractional shortening in stem cell and CM transplanted groups compared with respective controls. In conclusion, our data suggest that transplanted stem cells or their CM inhibit apoptosis, reduce fibrosis, and improve cardiac function in STZ-treated diabetic rats.
Collapse
Affiliation(s)
- Sarah Neel
- Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32816, United States
| | | |
Collapse
|
14
|
Yan B, Abdelli LS, Singla DK. Transplanted Induced Pluripotent Stem Cells Improve Cardiac Function and Induce Neovascularization in the Infarcted Hearts of db/db Mice. Mol Pharm 2011; 8:1602-10. [DOI: 10.1021/mp2003576] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Binbin Yan
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32817, United States
| | - Latifa S. Abdelli
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32817, United States
| | - Dinender K. Singla
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32817, United States
| |
Collapse
|
15
|
Bibliography-Editors' selection of current word literature. Coron Artery Dis 2010; 22:45-7. [PMID: 21160292 DOI: 10.1097/mca.0b013e328342fc9d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|