251
|
Cianciolo G, Capelli I, Cappuccilli M, Scrivo A, Donadei C, Marchetti A, Rucci P, La Manna G. Is chronic kidney disease-mineral and bone disorder associated with the presence of endothelial progenitor cells with a calcifying phenotype? Clin Kidney J 2017; 10:389-396. [PMID: 28616217 PMCID: PMC5466108 DOI: 10.1093/ckj/sfw145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/02/2016] [Indexed: 01/21/2023] Open
Abstract
Background: Chronic kidney disease-mineral and bone disorder (CKD-MBD) has been implicated in vascular calcification pathogenesis. CKD-MBD results in alterations in the number and function of circulating endothelial progenitor cells (EPCs), physiological regulators of angiogenesis and vessel repair, commonly defined as proangiogenic progenitor cells (PACs) by the antigen pattern CD34+CD133+KDR+CD45– and putative EPCs by the pattern CD34+CD133−KDR+CD45–. These cells might acquire a calcifying phenotype in CKD-MBD, expressing mineralization biomarkers. We investigated the expression of vitamin D receptor (VDR) and osteocalcin (OC) on EPCs of healthy individuals and haemodialysis patients, and their possible associations with circulating biomarkers of inflammation and vascular calcification. Methods: We compared EPC counts, expressing VDR or OC, in 23 healthy subjects versus 53 haemodialysis patients, 17 of them without vitamin D receptor agonist (VDRA) therapy and 35 treated with calcitriol (n = 17) or paricalcitol (n = 18). The correlations with serum levels of inflammatory and calcification indexes were also analysed. Results: All subsets expressing VDR or OC were significantly higher in haemodialysis patients compared with healthy controls, but PACs were increased only in VDRA treatment subgroup, while putative EPCs showed a similar rise also in untreated patients. In VDRA-untreated patients, OC+ PACs correlated positively with calcium levels, while in VDRA-treated patients, VDR+ PACs correlated positively with interleukin 6 levels, and OC+ PACs correlated positively 25-hydroxyvitamin D levels. Conclusions: Our data suggest that in CKD-MBD, EPCs undergo an endothelial-to-procalcific shift, representing a risk factor for vascular calcification. A link between mineral disorders and vitamin D replacement therapy emerged, with potential adverse effects for CKD patients.
Collapse
Affiliation(s)
- Giuseppe Cianciolo
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Irene Capelli
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Maria Cappuccilli
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Anna Scrivo
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Chiara Donadei
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Antonio Marchetti
- Department of Biomedical and Neuromotor Sciences, Unit of Hygiene and Biostatistics, University of Bologna, Bologna, Italy
| | - Paola Rucci
- Department of Biomedical and Neuromotor Sciences, Unit of Hygiene and Biostatistics, University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
252
|
Abstract
Chronic obstructive pulmonary disease (COPD) is regarded as a disease of accelerated lung aging. This affliction shows all of the hallmarks of aging, including telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence, and a low-grade chronic inflammation (inflammaging). Many of these pathways are driven by chronic exogenous and endogenous oxidative stress. There is also a reduction in antiaging molecules, such as sirtuins and Klotho, which further accelerate the aging process. COPD is associated with several comorbidities (multimorbidity), such as cardiovascular and metabolic diseases, that share the same pathways of accelerated aging. Understanding these mechanisms has helped identify several novel therapeutic targets, and several drugs and dietary interventions are now in development to treat multimorbidity.
Collapse
Affiliation(s)
- Peter J. Barnes
- National Heart and Lung Institute, Imperial College, London SW3 6LY, United Kingdom
| |
Collapse
|
253
|
Tan Q, Zhang S, Qi X, Zou X, Sun Q. Permanent atrial fibrillation impairs the function of circulating endothelial progenitor cells. Postgrad Med 2017; 129:198-204. [PMID: 28128678 DOI: 10.1080/00325481.2017.1288063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE to determine whether functions of endothelial progenitor cells (EPCs) were impaired in patients with permanent atrial fibrillation (AF). METHODS 35 patients with permanent AF (AF group) and 35 age and sex matched controls (control group) were collected. The numbers of circulating CD34+/KDR+ cells were determined with flow cytometry in the two groups. Cell proliferation, tube formation, nitric oxygen (NO) and vascular endothelial growth factor (VEGF) were assayed. RESULTS the numbers of CD34+/KDR+ cells were lower in the AF group than the control group (20.01 ± 12.66 /105 vs 77.93 ± 58.93 /105,p = 0.022). Colony formation unit (CFU) of EPCs were decreased in AF group compared to the control group(1.76 ± 0.59 CFU vs 3.45 ± 0.82 CFU, p = 0.0000). The AF group had lower cell proliferation ability than control group(0.401 ± 0.113 A vs 0.558 ± 0.130 A, p = 0.004). Tube formation ability was decreased in AF patients compared to controls(434.30 ± 96.22μm/mm2 vs 568.09 ± 196.17μm/mm2,p = 0.041). AF patients had lower VEGF secretion than controls (27.35 ± 9.93 ng/L vs 41.86 ± 7.31 ng/L,p = 0.001),they also had lower NO secretion than controls(16.55 ± 6.92μmol/l vs 23.65 ± 5.48,p = 0.012). CONCLUSIONS proliferation, tube formation and paracrine of EPCs were reduced in patients with permanent AF.
Collapse
Affiliation(s)
- Qiang Tan
- a Department of Cardiology , Qinhuangdao First Hospital, Hebei Medical University , Qinhuangdao , Hebei Province , China
| | - Shuangyue Zhang
- a Department of Cardiology , Qinhuangdao First Hospital, Hebei Medical University , Qinhuangdao , Hebei Province , China
| | - Ximing Qi
- a Department of Cardiology , Qinhuangdao First Hospital, Hebei Medical University , Qinhuangdao , Hebei Province , China
| | - Xiaoyi Zou
- a Department of Cardiology , Qinhuangdao First Hospital, Hebei Medical University , Qinhuangdao , Hebei Province , China
| | - Qiang Sun
- a Department of Cardiology , Qinhuangdao First Hospital, Hebei Medical University , Qinhuangdao , Hebei Province , China
| |
Collapse
|
254
|
Patschan D, Schwarze K, Tampe B, Zeisberg M, Patschan S, Müller GA. Endothelial Colony Forming Cells (ECFCs) in murine AKI - implications for future cell-based therapies. BMC Nephrol 2017; 18:53. [PMID: 28166726 PMCID: PMC5294892 DOI: 10.1186/s12882-017-0471-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
Background In recent years, early Endothelial Progenitor Cells (eEPCs) have been proven as effective tool in murine ischemic AKI and in diabetic nephropathy. The mechanisms of eEPC-mediated vasoprotection have been elucidated in detail. Besides producing a diverse range of humoral factors, the cells also act by secreting vasomodulatory microvesicles. Only few data in contrast have been published about the role of so-called Endothelial Colony Forming Cells (ECFCs - late EPCs) in ischemic AKI. We thus aimed to investigate ECFC effects on postischemic kidney function over several weeks. Our special interest focused on endothelial-to-mesenchymal transition (EndoMT), peritubular capillary density (PTCD), endothelial alpha-Tubulin (aT - cytoskeletal integrity), and endothelial p62 (marker of autophagocytic flux). Methods Eight to twelve weeks old male C57Bl/6 N mice were subjected to bilateral renal pedicle clamping for 35 or 45 min, respectively. Donor-derived syngeneic ECFCs (0.5 × 106) were i.v. injected at the end of ischemia. Animals were analyzed 1, 4 and 6 weeks later. Results Cell therapy improved kidney function exclusively at week 1 (35 and 45 min). Ischemia-induced fibrosis was diminished in all experimental groups by ECFCs, while PTCD loss remained unaffected. Significant EndoMT was detected in only two of 6 groups (35 min, week 4 and 45 min, week 6), ECFCs reduced EndoMT only in the latter. Endothelial aT declined under almost all experimental conditions and these effects were further aggravated by ECFCs. p62 was elevated in endothelial cells, more so after 45 than after 35 min of ischemia. Cell therapy did not modulate p62 abundances at any time point. Conclusion A single dose of ECFCs administered shortly post-ischemia is capable to reduce interstitial fibrosis in the mid- to long-term whereas excretory dysfunction is improved only in a transient manner. There are certain differences in renal outcome parameters between eEPCs and ECFC. The latter do not prevent animals from peritubular capillary loss and they also do not further elevate endothelial p62. We conclude that differences between eEPCs and ECFCs result from certain mechanisms by which the cells act around and within vessels. Overall, ECFC treatment was not as efficient as eEPC therapy in preventing mice from ischemia-induced mid- to long-term damage.
Collapse
Affiliation(s)
- D Patschan
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
| | - K Schwarze
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - B Tampe
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - M Zeisberg
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - S Patschan
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - G A Müller
- Clinic of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| |
Collapse
|
255
|
Liang J, Huang W, Cai W, Wang L, Guo L, Paul C, Yu XY, Wang Y. Inhibition of microRNA-495 Enhances Therapeutic Angiogenesis of Human Induced Pluripotent Stem Cells. Stem Cells 2017; 35:337-350. [DOI: 10.1002/stem.2477] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Therapeutic angiogenesis has emerged as a promising strategy to regenerate the damaged blood vessels resulting from ischemic diseases such as myocardial infarction (MI). However, the functional integration of implanted endothelial cells (ECs) in infarcted heart remains challenging. We herein develop an EC generation approach by inhibiting microRNA-495 (miR-495) in human induced pluripotent stem cells (hiPSCs) and assess the angiogenic potential for MI treatment. The anti-angiogenic miR-495 belonging to Dlk1-Dio3 miR cluster was identified through expression profiling and computational analysis. Loss-of-function experiments for miR-495 were performed using a lentiviral transfer of antisense sequence in hiPSCs. The pluripotency of hiPSCs was not impacted by the genetic modification. Induced with differentiation medium, miR-495 inhibition enhanced the expression of EC genes of hiPSCs, as well as the yield of ECs. Newly derived ECs displayed prominent angiogenic characteristics including tube formation, cell migration, and proliferation. Mechanistically, miR-495 mediated the expression of endothelial or angiogenic genes by directly targeting vascular endothelial zinc finger 1. After transplantation in immunodeficient MI mice, the derived ECs significantly increased neovascularization in the infarcted heart, prevented functional worsening, and attenuated expansion of infarct size. The functional integration of the implanted ECs into coronary networks was also enhanced by inhibiting miR-495. miR-495 represents a new target not only for promoting EC generation from hiPSCs but also for enhancing angiogenesis and engraftment of hiPSC-derived ECs in ischemic heart.
Collapse
Affiliation(s)
- Jialiang Liang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Wei Huang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Wenfeng Cai
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Lei Wang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Linlin Guo
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Christian Paul
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Xi-Yong Yu
- b Institute of Molecular and Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yigang Wang
- a Departments of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
256
|
Dai X, Yan X, Zeng J, Chen J, Wang Y, Chen J, Li Y, Barati MT, Wintergerst KA, Pan K, Nystoriak MA, Conklin DJ, Rokosh G, Epstein PN, Li X, Tan Y. Elevating CXCR7 Improves Angiogenic Function of EPCs via Akt/GSK-3β/Fyn-Mediated Nrf2 Activation in Diabetic Limb Ischemia. Circ Res 2017; 120:e7-e23. [PMID: 28137917 DOI: 10.1161/circresaha.117.310619] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Endothelial progenitor cells (EPCs) respond to stromal cell-derived factor 1 (SDF-1) through chemokine receptors CXCR7 and CXCR4. Whether SDF-1 receptors involves in diabetes mellitus-induced EPCs dysfunction remains unknown. OBJECTIVE To determine the role of SDF-1 receptors in diabetic EPCs dysfunction. METHODS AND RESULTS CXCR7 expression, but not CXCR4 was reduced in EPCs from db/db mice, which coincided with impaired tube formation. Knockdown of CXCR7 impaired tube formation of EPCs from normal mice, whereas upregulation of CXCR7 rescued angiogenic function of EPCs from db/db mice. In normal EPCs treated with oxidized low-density lipoprotein or high glucose also reduced CXCR7 expression, impaired tube formation, and increased oxidative stress and apoptosis. The damaging effects of oxidized low-density lipoprotein or high glucose were markedly reduced by SDF-1 pretreatment in EPCs transduced with CXCR7 lentivirus but not in EPCs transduced with control lentivirus. Most importantly, EPCs transduced with CXCR7 lentivirus were superior to EPCs transduced with control lentivirus for therapy of ischemic limbs in db/db mice. Mechanistic studies demonstrated that oxidized low-density lipoprotein or high glucose inhibited protein kinase B and glycogen synthase kinase-3β phosphorylation, nuclear export of Fyn and nuclear localization of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), blunting Nrf2 downstream target genes heme oxygenase-1, NAD(P)H dehydrogenase (quinone 1) and catalase, and inducing an increase in EPC oxidative stress. This destructive cascade was blocked by SDF-1 treatment in EPCs transduced with CXCR7 lentivirus. Furthermore, inhibition of phosphatidylinositol 3-kinase/protein kinase B prevented SDF-1/CXCR7-mediated Nrf2 activation and blocked angiogenic repair. Moreover, Nrf2 knockdown almost completely abolished the protective effects of SDF-1/CXCR7 on EPC function in vitro and in vivo. CONCLUSIONS Elevated expression of CXCR7 enhances EPC resistance to diabetes mellitus-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia. The benefits of CXCR7 are mediated predominantly by a protein kinase B/glycogen synthase kinase-3β/Fyn pathway via increased activity of Nrf2.
Collapse
Affiliation(s)
- Xiaozhen Dai
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Xiaoqing Yan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jun Zeng
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jing Chen
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yuehui Wang
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jun Chen
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yan Li
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Michelle T Barati
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Kupper A Wintergerst
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Kejian Pan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Matthew A Nystoriak
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Daniel J Conklin
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Gregg Rokosh
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Paul N Epstein
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Xiaokun Li
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yi Tan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.).
| |
Collapse
|
257
|
Sandhu K, Mamas M, Butler R. Endothelial progenitor cells: Exploring the pleiotropic effects of statins. World J Cardiol 2017; 9:1-13. [PMID: 28163831 PMCID: PMC5253189 DOI: 10.4330/wjc.v9.i1.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/29/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023] Open
Abstract
Statins have become a cornerstone of risk modification for ischaemic heart disease patients. A number of studies have shown that they are effective and safe. However studies have observed an early benefit in terms of a reduction in recurrent infarct and or death after a myocardial infarction, prior to any significant change in lipid profile. Therefore, pleiotropic mechanisms, other than lowering lipid profile alone, must account for this effect. One such proposed pleiotropic mechanism is the ability of statins to augment both number and function of endothelial progenitor cells. The ability to augment repair and maintenance of a functioning endothelium may have profound beneficial effect on vascular repair and potentially a positive impact on clinical outcomes in patients with cardiovascular disease. The following literature review will discuss issues surrounding endothelial progenitor cell (EPC) identification, role in vascular repair, factors affecting EPC numbers, the role of statins in current medical practice and their effects on EPC number.
Collapse
|
258
|
Gebhard C, Rhéaume E, Berry C, Brand G, Kernaleguen AE, Théberge-Julien G, Alam MA, Lee CYW, Boileau L, Chabot-Blanchet M, Guertin MC, Lavoie MA, Grégoire J, Ibrahim R, L'Allier P, Tardif JC. Beneficial Effects of Reconstituted High-Density Lipoprotein (rHDL) on Circulating CD34+ Cells in Patients after an Acute Coronary Syndrome. PLoS One 2017; 12:e0168448. [PMID: 28060837 PMCID: PMC5218493 DOI: 10.1371/journal.pone.0168448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
Background High-density lipoproteins (HDL) favorably affect endothelial progenitor cells (EPC). Circulating progenitor cell level and function are impaired in patients with acute coronary syndrome (ACS). This study investigates the short-term effects of reconstituted HDL (rHDL) on circulating progenitor cells in patients with ACS. Methods and Findings The study population consisted of 33 patients with recent ACS: 20 patients from the ERASE trial (randomized to receive 4 weekly intravenous infusions of CSL-111 40 mg/kg or placebo) and 13 additional patients recruited as controls using the same enrolment criteria. Blood was collected from 16 rHDL (CSL-111)-treated patients and 17 controls at baseline and at 6–7 weeks (i.e. 2–3 weeks after the fourth infusion of CSL-111 in ERASE). CD34+ and CD34+/kinase insert domain receptor (KDR+) progenitor cell counts were analyzed by flow cytometry. We found preserved CD34+ cell counts in CSL-111-treated subjects at follow-up (change of 1.6%), while the number of CD34+ cells was reduced (-32.9%) in controls (p = 0.017 between groups). The level of circulating SDF-1 (stromal cell-derived factor-1), a chemokine involved in progenitor cell recruitment, increased significantly (change of 21.5%) in controls, while it remained unchanged in CSL-111-treated patients (p = 0.031 between groups). In vitro exposure to CSL-111 of early EPC isolated from healthy volunteers significantly increased CD34+ cells, reduced early EPC apoptosis and enhanced their migration capacity towards SDF-1. Conclusions The relative increase in circulating CD34+ cells and the low SDF-1 levels observed following rHDL infusions in ACS patients point towards a role of rHDL in cardiovascular repair mechanisms.
Collapse
Affiliation(s)
- Catherine Gebhard
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Eric Rhéaume
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Colin Berry
- Montreal Heart Institute, Montreal, Quebec, Canada
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | - Laurianne Boileau
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | | | | | - Marc-André Lavoie
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Jean Grégoire
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Réda Ibrahim
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Philippe L'Allier
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
259
|
Li Q, Xia S, Fang H, Pan J, Jia Y, Deng G. VEGF treatment promotes bone marrow-derived CXCR4 + mesenchymal stromal stem cell differentiation into vessel endothelial cells. Exp Ther Med 2017; 13:449-454. [PMID: 28352314 PMCID: PMC5348687 DOI: 10.3892/etm.2017.4019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/11/2016] [Indexed: 01/08/2023] Open
Abstract
Stem/progenitor cells serve an important role in the process of blood vessel repair. However, the mechanism of vascular repair mediated by C-X-C chemokine receptor type 4-positive (CXCR4+) bone marrow-derived mesenchymal stem cells (BMSCs) following myocardial infarction remains unclear. The aim of the present study was to investigate the effects of vascular endothelial growth factor (VEGF) on vessel endothelial differentiation from BMSCs. CXCR4+ BMSCs were isolated from the femoral bone marrow of 2-month-old mice and the cells were treated with VEGF. Expression of endothelial cell markers and the functional properties were assessed by reverse transcription-quantitative polymerase chain reaction, flow cytometry and vascular formation analyses. The results indicated that the CXCR4+ BMSCs from femoral bone marrow cells expressed putative cell surface markers of mesenchymal stem cells. Treatment with VEGF induced platelet/endothelial cell adhesion molecule-1 (PECAM-1) and von Willebrand factor (vWF) expression at the transcriptional and translational levels, compared with untreated controls. Moreover, VEGF treatment induced CXCR4+ BMSCs to form hollow tube-like structures on Matrigel, suggesting that the differentiated endothelial cells had the functional properties of blood vessels. The results demonstrate that the CXCR4+ BMSCs were able to differentiate into vessel endothelial cells following VEGF treatment. For cell transplantation in vascular disease, it may be concluded that CXCR4+ BMSCs are a novel source of endothelial progenitor cells with high potential for application in vascular repair.
Collapse
Affiliation(s)
- Qiming Li
- Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Shudong Xia
- Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Hanyun Fang
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Jiansheng Pan
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Yinfeng Jia
- Department of Cardiology, The Second People's Hospital of Yueqing, Yueqing, Zhejiang 325608, P.R. China
| | - Gang Deng
- The Ningbo Central Blood Station, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
260
|
Fadini GP, Rigato M, Cappellari R, Bonora BM, Avogaro A. Long-term Prediction of Cardiovascular Outcomes by Circulating CD34+ and CD34+CD133+ Stem Cells in Patients With Type 2 Diabetes. Diabetes Care 2017; 40:125-131. [PMID: 27815289 DOI: 10.2337/dc16-1755] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 10/19/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Cardiovascular risk varies substantially in the population with diabetes, and biomarkers can improve risk stratification. Circulating stem cells predict future cardiovascular events and death, but data for the population with diabetes are scant. In this study we evaluated the ability of circulating stem cell levels to predict future cardiovascular outcomes and improve risk discrimination in patients with type 2 diabetes. RESEARCH DESIGN AND METHODS A cohort of 187 patients with type 2 diabetes was monitored for a median of 6.1 years. The primary outcome was time to a first cardiovascular event, defined as 3-point major adverse cardiovascular event (cardiovascular death, nonfatal myocardial infarction, or nonfatal stroke) plus hospitalization for cardiovascular causes. At baseline, we measured six stem/progenitor cell phenotypes in peripheral blood based on expression of CD34, CD133, and KDR. RESULTS The primary outcome occurred in 48 patients (4.5/100 patient-years). Patients with incident cardiovascular events had significantly lower CD34+ and CD34+CD133+ cells than those without. Higher rates of cardiovascular events occurred in patients with below median levels of CD34+ and CD34+CD133+. In Cox proportional hazards regression analyses, a reduced CD34+ (hazard ratio 2.21 [95% CI 1.14-4.29]) and CD34+CD133+ (2.98 [1.46-6.08]) cell count independently predicted future events. Addition of the CD34+ cell count to the reference model or the UK Prospective Diabetes Study risk engine improved C statistics, continuous net reclassification improvement, and/or integrated discrimination index. CONCLUSIONS In patients with type 2 diabetes, a reduced baseline level of circulating CD34+ stem cells predicts adverse cardiovascular outcomes up to 6 years later and improves risk stratification.
Collapse
Affiliation(s)
| | - Mauro Rigato
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
261
|
Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee SJ, Oh SY, Tanaka Y, Park IH, An HJ, Shin CM, Sharma S, Yoon YS. Direct Reprogramming of Human Dermal Fibroblasts Into Endothelial Cells Using ER71/ETV2. Circ Res 2016; 120:848-861. [PMID: 28003219 DOI: 10.1161/circresaha.116.309833] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022]
Abstract
RATIONALE Direct conversion or reprogramming of human postnatal cells into endothelial cells (ECs), bypassing stem or progenitor cell status, is crucial for regenerative medicine, cell therapy, and pathophysiological investigation but has remained largely unexplored. OBJECTIVE We sought to directly reprogram human postnatal dermal fibroblasts to ECs with vasculogenic and endothelial transcription factors and determine their vascularizing and therapeutic potential. METHODS AND RESULTS We utilized various combinations of 7 EC transcription factors to transduce human postnatal dermal fibroblasts and found that ER71/ETV2 (ETS variant 2) alone best induced endothelial features. KDR+ (kinase insert domain receptor) cells sorted at day 7 from ER71/ETV2-transduced human postnatal dermal fibroblasts showed less mature but enriched endothelial characteristics and thus were referred to as early reprogrammed ECs (rECs), and did not undergo maturation by further culture. After a period of several weeks' transgene-free culture followed by transient reinduction of ER71/ETV2, early rECs matured during 3 months of culture and showed reduced ETV2 expression, reaching a mature phenotype similar to postnatal human ECs. These were termed late rECs. While early rECs exhibited an immature phenotype, their implantation into ischemic hindlimbs induced enhanced recovery from ischemia. These 2 rECs showed clear capacity for contributing to new vessel formation through direct vascular incorporation in vivo. Paracrine or proangiogenic effects of implanted early rECs played a significant role in repairing hindlimb ischemia. CONCLUSIONS This study for the first time demonstrates that ER71/ETV2 alone can directly reprogram human postnatal cells to functional, mature ECs after an intervening transgene-free period. These rECs could be valuable for cell therapy, personalized disease investigation, and exploration of the reprogramming process.
Collapse
Affiliation(s)
- Sangho Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Changwon Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ji Woong Han
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ju Young Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Kyuwon Cho
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Eun Jae Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Sangsung Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shin-Jeong Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Se Yeong Oh
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Yoshiaki Tanaka
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - In-Hyun Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Hyo Jae An
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Claire Min Shin
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shraya Sharma
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Young-Sup Yoon
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| |
Collapse
|
262
|
Stefanou C, Karatzanos E, Mitsiou G, Psarra K, Angelopoulos E, Dimopoulos S, Gerovasili V, Boviatsis E, Routsi C, Nanas S. Neuromuscular electrical stimulation acutely mobilizes endothelial progenitor cells in critically ill patients with sepsis. Ann Intensive Care 2016; 6:21. [PMID: 26969168 PMCID: PMC4788669 DOI: 10.1186/s13613-016-0123-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/29/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) have been suggested to constitute a restoration index of the disturbed endothelium in ICU patients. Neuromuscular electric stimulation (NMES) is increasingly employed in ICU to prevent comorbidities such as ICU-acquired weakness, which is related to endothelial dysfunction. The role of NMES to mobilize EPCs has not been investigated yet. The purpose of this study was to explore the NMES-induced effects on mobilization of EPCs in septic ICU patients. METHODS Thirty-two septic mechanically ventilated patients (mean ± SD, age 58 ± 14 years) were randomized to one of the two 30-min NMES protocols of different characteristics: a high-frequency (75 Hz, 6 s on-21 s off) or a medium-frequency (45 Hz, 5 s on-12 s off) protocol both applied at maximally tolerated intensity. Blood was sampled before and immediately after the NMES sessions. Different EPCs subpopulations were quantified by cytometry markers CD34(+)/CD133(+)/CD45(-), CD34(+)/CD133(+)/CD45(-)/VEGFR2 (+) and CD34(+)/CD45(-)/VEGFR2 (+). RESULTS Overall, CD34(+)/CD133(+)/CD45(-) EPCs increased from 13.5 ± 10.2 to 20.8 ± 16.9 and CD34(+)/CD133(+)/CD45(-)/VEGFR2 (+) EPCs from 3.8 ± 5.2 to 6.4 ± 8.5 cells/10(6) enucleated cells (mean ± SD, p < 0.05). CD34(+)/CD45(-)/VEGFR2 (+) EPCs also increased from 16.5 ± 14.5 to 23.8 ± 19.2 cells/10(6) enucleated cells (mean ± SD, p < 0.05). EPCs mobilization was not affected by NMES protocol and sepsis severity (p > 0.05), while it was related to corticosteroids administration (p < 0.05). CONCLUSIONS NMES acutely mobilized endothelial progenitor cells, measures of the endothelial restoration potential, in septic ICU patients.
Collapse
Affiliation(s)
- Christos Stefanou
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Eleftherios Karatzanos
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Georgios Mitsiou
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Katerina Psarra
- />Immunology and Histocompatibility Department, Evangelismos General Hospital, 45-47 Ypsilantou Str, 106 75 Athens, Greece
| | - Epameinondas Angelopoulos
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Stavros Dimopoulos
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
- />Critical Care Unit, Guys and St Thomas Hospital, Westminster Bridge Road, London, SE1 7EH UK
| | - Vasiliki Gerovasili
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Efstathios Boviatsis
- />2nd Neurosurgical Department, Attiko University General Hospital, School of Medicine, National and Kapodistrian University of Athens, 1 Rimini Str, 124 62 Athens, Greece
| | - Christina Routsi
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - Serafeim Nanas
- />1st Critical Care Department, Evangelismos General Hospital, School of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| |
Collapse
|
263
|
Malinovskaya NA, Komleva YK, Salmin VV, Morgun AV, Shuvaev AN, Panina YA, Boitsova EB, Salmina AB. Endothelial Progenitor Cells Physiology and Metabolic Plasticity in Brain Angiogenesis and Blood-Brain Barrier Modeling. Front Physiol 2016; 7:599. [PMID: 27990124 PMCID: PMC5130982 DOI: 10.3389/fphys.2016.00599] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022] Open
Abstract
Currently, there is a considerable interest to the assessment of blood-brain barrier (BBB) development as a part of cerebral angiogenesis developmental program. Embryonic and adult angiogenesis in the brain is governed by the coordinated activity of endothelial progenitor cells, brain microvascular endothelial cells, and non-endothelial cells contributing to the establishment of the BBB (pericytes, astrocytes, neurons). Metabolic and functional plasticity of endothelial progenitor cells controls their timely recruitment, precise homing to the brain microvessels, and efficient support of brain angiogenesis. Deciphering endothelial progenitor cells physiology would provide novel engineering approaches to establish adequate microfluidically-supported BBB models and brain microphysiological systems for translational studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alla B. Salmina
- Research Institute of Molecular Medicine & Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| |
Collapse
|
264
|
Abstract
The concept of pericyte has been changing over years. This cell type was believed to possess only a function of trophic support to endothelial cells and to maintain vasculature stabilization. In the last years, the discovery of multipotent ability of perivascular populations led to the concept of vessel/wall niche. Likewise, several perivascular populations have been identified in animal and human bone marrow. In this review, we provide an overview on bone marrow perivascular population, their cross-talk with other niche components, relationship with bone marrow stromal stem cells, and similarities and differences with the perivascular population of the vessel/wall niche. Finally, we focus on the regenerative potential of these cells and the forthcoming challenges related to their use as cell therapy products.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Andrea Cordaro
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
265
|
Zayed SA, Gaafar TM, Samy RM, Sabry D, Nasr AS, Maksoud FA. Production of endothelial progenitor cells obtained from human Wharton's jelly using different culture conditions. Biotech Histochem 2016; 91:532-539. [PMID: 27849398 DOI: 10.1080/10520295.2016.1250284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Endothelial progenitor cells (EPC) participate in revascularization and angiogenesis. EPC can be cultured in vitro from mononuclear cells of peripheral blood, umbilical cord blood or bone marrow; they also can be transdifferentiated from mesenchymal stem cells (MSC). We isolated EPCs from Wharton's jelly (WJ) using two methods. The first method was by obtaining MSC from WJ and characterizing them by flow cytometry and their adipogenic and osteogenic differentiation, then applying endothelial growth differentiating media. The second method was by direct culture of cells derived from WJ into endothelial differentiating media. EPCs were characterized by morphology, Dil-LDL uptake/UEA-1 immunostaining and testing the expression of endothelial markers by flow cytometry and RT-PCR. We found that MSC derived from WJ differentiated into endothelial-like cells using simple culture conditions with endothelium induction agents in the medium.
Collapse
Affiliation(s)
- S A Zayed
- a Department of Clinical and Chemical Pathology , Cairo , Egypt
| | - T M Gaafar
- a Department of Clinical and Chemical Pathology , Cairo , Egypt
| | - R M Samy
- a Department of Clinical and Chemical Pathology , Cairo , Egypt
| | - D Sabry
- b Department of Biochemistry , Cairo , Egypt
| | - A S Nasr
- c Department of Obstetrics and Gynecology , Cairo University , Cairo , Egypt
| | | |
Collapse
|
266
|
Puca AA, Spinetti G, Vono R, Vecchione C, Madeddu P. The genetics of exceptional longevity identifies new druggable targets for vascular protection and repair. Pharmacol Res 2016; 114:169-174. [PMID: 27818232 DOI: 10.1016/j.phrs.2016.10.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/15/2016] [Accepted: 10/31/2016] [Indexed: 12/23/2022]
Abstract
Therapeutic angiogenesis is a relatively new medical strategy in the field of cardiovascular diseases. The underpinning concept is that angiogenic growth factors or proangiogenic cells could be exploited therapeutically in cardiovascular patients to enhance native revascularization responses to an ischemic insult, thereby accelerating tissue healing. The initial enthusiasm generated by preclinical studies has been tempered by the modest success of clinical trials assessing therapeutic angiogenesis. Similarly, proangiogenic cell therapy has so far not maintained the original promises. Intriguingly, the current trend is to consider regeneration as a prerogative of the youngest organism. Consequentially, the embryonic and foetal models are attracting much attention for clinical translation into corrective modalities in the adulthood. Scientists seem to undervalue the lesson from Mother Nature, e.g. all humans are born young but very few achieve the goal of an exceptional healthy longevity. Either natural experimentation is driven by a supreme intelligence or stochastic phenomena, one has to accept the evidence that healthy longevity is the fruit of an evolutionary process lasting million years. It is therefore extremely likely that results of this natural experimentation are more reliable and translatable than the intensive, but very short human investigation on mechanisms governing repair and regeneration. With this preamble in mind, here we propose to shift the focus from the very beginning to the very end of human life and thus capture the secret of prolonged health span to improve well-being in the adulthood.
Collapse
Affiliation(s)
- Annibale A Puca
- IRCCS MultiMedica, Milan, Italy; University of Salerno, Salerno, Italy
| | | | | | - Carmine Vecchione
- University of Salerno, Salerno, Italy; IRCCS Neuromed, Pozzilli, Italy
| | | |
Collapse
|
267
|
Landers-Ramos RQ, Sapp RM, VandeWater E, Macko J, Robinson S, Wang Y, Chin ER, Spangenburg EE, Prior SJ, Hagberg JM. Investigating the extremes of the continuum of paracrine functions in CD34-/CD31+ CACs across diverse populations. Am J Physiol Heart Circ Physiol 2016; 312:H162-H172. [PMID: 27793853 DOI: 10.1152/ajpheart.00342.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/30/2016] [Accepted: 10/14/2016] [Indexed: 11/22/2022]
Abstract
Paracrine function of circulating angiogenic cells (CACs) is thought to contribute to vascular maintenance. We previously identified S100A8 and S100A9 secreted from physically inactive individuals' CD34-/CD31+ CACs as negative regulators of capillary-like network formation. The purpose of this study was to investigate further the extremes of the continuum of CAC paracrine actions using two distinctly different groups representing "healthy" and "impaired" CAC function. We aimed to determine how capillary-like network formation in human umbilical vein endothelial cells (HUVECs) is affected by S100A8 and S100A9 in concentrations secreted by CACs from different ends of the health spectrum. CD34-/CD31+ CACs were isolated and cultured from 10 impaired function individuals defined as older (50-89 yr), non-ST-elevation myocardial infarction patients and 10 healthy individuals defined as younger (18-35 yr), healthy individuals, and conditioned media (CM) was generated. CM from the impaired function group's CACs significantly diminished network formation compared with CM from the healthy group (P < 0.05). We identified elevations in S100A8, S100A9, and S100A8/A9 in the CM from the impaired function group (P < 0.05). Pretreatment of HUVECs with inhibitors to a known S100A8 and S100A9 receptor, Toll-like receptor 4 (TLR4), but not receptor for advanced glycation end products, improved HUVEC network formation (P < 0.05) compared with CM alone in the impaired function conditions. Exposure of HUVECs to the TLR4 signaling inhibitor also blocked recombinant S100A8- and S100A9-mediated reductions in network formation. Collectively, the results suggest that the mechanisms behind impaired CAC CD34-/CD31+ CM-mediated reductions in capillary-like network formation involve secretion of S100A8 and S100A9 and binding of these proteins to TLR4 receptors on HUVECs. NEW & NOTEWORTHY S100A8 and S100A9 proteins in concentrations secreted by CD34-/CD31+ circulating angiogenic cells (CACs) with impaired function reduce endothelial cell capillary-like network formation. These effects appear to be mediated by Toll-like receptor 4 and are absent with S100A8 and S100A9 in concentrations secreted by healthy CD34-/CD31+ CACs.
Collapse
Affiliation(s)
- Rian Q Landers-Ramos
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Ryan M Sapp
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Emily VandeWater
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Jennifer Macko
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Shawn Robinson
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Yan Wang
- Proteomics Core Facility, College of Computer, Mathematics, and Natural Sciences, University of Maryland, College Park, Maryland; and
| | - Eva R Chin
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Espen E Spangenburg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland
| | - Steven J Prior
- University of Maryland School of Medicine and Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, Maryland
| | - James M Hagberg
- Department of Kinesiology, School of Public Health, University of Maryland, College Park, Maryland;
| |
Collapse
|
268
|
Wils J, Favre J, Bellien J. Modulating putative endothelial progenitor cells for the treatment of endothelial dysfunction and cardiovascular complications in diabetes. Pharmacol Ther 2016; 170:98-115. [PMID: 27773788 DOI: 10.1016/j.pharmthera.2016.10.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetes induces a decrease in the number and function of different pro-angiogenic cell types generically designated as putative endothelial progenitor cells (EPC), which encompasses cells from myeloid origin that act in a paracrine fashion to promote angiogenesis and putative "true" EPC that contribute to endothelial replacement. This not only compromises neovasculogenesis in ischemic tissues but also impairs, at an early stage, the reendotheliziation process at sites of injury, contributing to the development of endothelial dysfunction and cardiovascular complications. Hyperglycemia, insulin resistance and dyslipidemia promote putative EPC dysregulation by affecting the SDF-1/CXCR-4 and NO pathways and the p53/SIRT1/p66Shc axis that contribute to their mobilization, migration, homing and vasculogenic properties. To optimize the clinical management of patients with hypoglycemic agents, statins and renin-angiotensin system inhibitors, which display pleiotropic effects on putative EPC, is a first step to improve their number and angiogenic potential but specific strategies are needed. Among them, mobilizing therapies based on G-CSF, erythropoietin or CXCR-4 antagonism have been developed to increase putative EPC number to treat ischemic diseases with or without prior cell isolation and transplantation. Growth factors, genetic and pharmacological strategies are also evaluated to improve ex vivo cultured EPC function before transplantation. Moreover, pharmacological agents increasing in vivo the bioavailability of NO and other endothelial factors demonstrated beneficial effects on neovascularization in diabetic ischemic models but their effects on endothelial dysfunction remain poorly evaluated. More experiments are warranted to develop orally available drugs and specific agents targeting p66Shc to reverse putative EPC dysfunction in the expected goal of preventing endothelial dysfunction and diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Julien Wils
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Julie Favre
- MITOVASC Institute, Angers, France; Centre National de la Recherche Scientifique (CNRS) UMR 6214, Angers, France; INSERM U1083, Angers, France; University of Angers, Angers, France
| | - Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
269
|
Qu Q, Liu L, Chen G, Xu Y, Wu X, Wu D. Endothelial progenitor cells promote efficient ex vivo expansion of cord blood-derived hematopoietic stem/progenitor cells. Cytotherapy 2016; 18:452-64. [PMID: 26857234 DOI: 10.1016/j.jcyt.2015.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 12/27/2015] [Accepted: 12/30/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Cord blood (CB) hematopoietic stem cell transplantation has often been limited by the scarcity of stem cells. Therefore, the number of CB hematopoietic stem/progenitor cells (HSPCs) should be increased while maintaining the stem cell characteristics. METHODS We designed an ex vivo culture system using endothelial progenitor cells (EPCs) as stroma to determine the capacity of expanding CB-HSPCs in a defined medium, the effect on engraftment of the expanded cells in a mouse model and the underlying mechanism. RESULTS After 7 days of culture, compared with those cultured with cytokines alone (3.25 ± 0.59), CD34+ cells under contact and non-contact co-culture with EPCs were expanded by 5.38 ± 0.61 (P = 0.003) and 4.06 ± 0.43 (P = 0.025)-fold, respectively. Direct cell-to-cell contact co-culture with EPCs resulted in more primitive CD34+ CD38- cells than stroma-free culture (156.17 ± 21.32 versus 79.12 ± 19.77-fold; P = 0.010). Comparable engraftment of day 7 co-cultured HSPCs with respect to HSPCs at day 0 in nonobese diabetic-severe combined immunodeficiency disease (NOD/SCID) mice was measured as a percentage of chimerism (13.3% ± 11.0% versus 16.0% ± 14.3%; P = 0.750). EPCs highly expressed interleukin 6 (IL6) and angiopoietin 1 (ANGPT1), the hematopoietic- related cytokines. A higher transcriptional level of WNT5A genes in EPCs and co-cultured HSPCs suggests that the activation of Wnt signaling pathway may play a role in HSPCs' expansion ex vivo. DISCUSSION These data demonstrated that EPCs improve the CD34+ population but do not compromise the repopulating efficacy of the amplified HSPCs, possibly via cytokine secretion and Wnt signaling pathway activation.
Collapse
Affiliation(s)
- Qi Qu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Limin Liu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanghua Chen
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojin Wu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- Jiangsu Institute of Hematology, Suzhou Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
270
|
Wu Y, He MY, Ye JK, Ma SY, Huang W, Wei YY, Kong H, Wang H, Zeng XN, Xie WP. Activation of ATP-sensitive potassium channels facilitates the function of human endothelial colony-forming cells via Ca 2+ /Akt/eNOS pathway. J Cell Mol Med 2016; 21:609-620. [PMID: 27709781 PMCID: PMC5323860 DOI: 10.1111/jcmm.13006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Accumulating data, including those from our laboratory, have shown that the opening of ATP‐sensitive potassium channels (KATP) plays a protective role in pulmonary vascular diseases (PVD). As maintainers of the endothelial framework, endothelial colony‐forming cells (ECFCs) are considered excellent candidates for vascular regeneration in cases of PVD. Although KATP openers (KCOs) have been demonstrated to have beneficial effects on endothelial cells, the impact of KATP on ECFC function remains unclear. Herein, this study investigated whether there is a distribution of KATP in ECFCs and what role KATP play in ECFC modulation. By human ECFCs isolated from adult peripheral blood, KATP were confirmed for the first time to express in ECFCs, comprised subunits of Kir (Kir6.1, Kir6.2) and SUR2b. KCOs such as the classical agent nicorandil (Nico) and the novel agent iptakalim (Ipt) notably improved the function of ECFCs, promoting cell proliferation, migration and angiogenesis, which were abolished by a non‐selective KATP blocker glibenclamide (Gli). To determine the underlying mechanisms, we investigated the impacts of KCOs on CaMKII, Akt and endothelial nitric oxide synthase pathways. Enhanced levels were detected by western blotting, which were abrogated by Gli. This suggested an involvement of Ca2+ signalling in the regulation of ECFCs by KATP. Our findings demonstrated for the first time that there is a distribution of KATP in ECFCs and KATP play a vital role in ECFC function. The present work highlighted a novel profile of KATP as a potential target for ECFC modulation, which may hold the key to the treatment of PVD.
Collapse
Affiliation(s)
- Yan Wu
- Department of Respiratory Medicine, WuXi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Meng-Yu He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian-Kui Ye
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shu-Ying Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong-Yue Wei
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao-Ning Zeng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei-Ping Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
271
|
Recchioni R, Marcheselli F, Antonicelli R, Lazzarini R, Mensà E, Testa R, Procopio AD, Olivieri F. Physical activity and progenitor cell-mediated endothelial repair in chronic heart failure: Is there a role for epigenetics? Mech Ageing Dev 2016; 159:71-80. [DOI: 10.1016/j.mad.2016.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 02/09/2023]
|
272
|
Cañete A, Comaills V, Prados I, Castro AM, Hammad S, Ybot-Gonzalez P, Bockamp E, Hengstler JG, Gottgens B, Sánchez MJ. Characterization of a Fetal Liver Cell Population Endowed with Long-Term Multiorgan Endothelial Reconstitution Potential. Stem Cells 2016; 35:507-521. [PMID: 27615355 PMCID: PMC5298023 DOI: 10.1002/stem.2494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/27/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022]
Abstract
Stable reconstitution of vascular endothelial beds upon transplantation of progenitor cells represents an important challenge due to the paucity and generally limited integration/expansion potential of most identified vascular related cell subsets. We previously showed that mouse fetal liver (FL) hemato/vascular cells from day 12 of gestation (E12), expressing the Stem Cell Leukaemia (SCL) gene enhancer transgene (SCL‐PLAP+ cells), had robust endothelial engraftment potential when transferred to the blood stream of newborns or adult conditioned recipients, compared to the scarce vascular contribution of adult bone marrow cells. However, the specific SCL‐PLAP+ hematopoietic or endothelial cell subset responsible for the long‐term reconstituting endothelial cell (LTR‐EC) activity and its confinement to FL developmental stages remained unknown. Using a busulfan‐treated newborn transplantation model, we show that LTR‐EC activity is restricted to the SCL‐PLAP+VE‐cadherin+CD45− cell population, devoid of hematopoietic reconstitution activity and largely composed by Lyve1+ endothelial‐committed cells. SCL‐PLAP+ Ve‐cadherin+CD45− cells contributed to the liver sinusoidal endothelium and also to the heart, kidney and lung microvasculature. LTR‐EC activity was detected at different stages of FL development, yet marginal activity was identified in the adult liver, revealing unknown functional differences between fetal and adult liver endothelial/endothelial progenitors. Importantly, the observations that expanding donor‐derived vascular grafts colocalize with proliferating hepatocyte‐like cells and participate in the systemic circulation, support their functional integration into young livers. These findings offer new insights into the engraftment, phonotypical, and developmental characterization of a novel endothelial/endothelial progenitor cell subtype with multiorgan LTR‐EC activity, potentially instrumental for the treatment/genetic correction of vascular diseases. Stem Cells2017;35:507–521
Collapse
Affiliation(s)
- Ana Cañete
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Valentine Comaills
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Isabel Prados
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Ana María Castro
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Seddik Hammad
- Faculty of Veterinary Medicine, Department of Forensic Medicine and Veterinary Toxicology, South Valley University, Qena, Egypt.,Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Patricia Ybot-Gonzalez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jan G Hengstler
- Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Bertie Gottgens
- Cambridge Institute for Medical Research & Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge University, United Kingdom
| | - María José Sánchez
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| |
Collapse
|
273
|
Hassanpour M, Cheraghi O, Siavashi V, Rahbarghazi R, Nouri M. A reversal of age-dependent proliferative capacity of endothelial progenitor cells from different species origin in in vitro condition. J Cardiovasc Thorac Res 2016; 8:102-106. [PMID: 27777694 PMCID: PMC5075357 DOI: 10.15171/jcvtr.2016.22] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/11/2016] [Indexed: 01/09/2023] Open
Abstract
Introduction: A large number of cardiovascular disorders and abnormalities, notably accelerated vascular deficiencies could be related to aging changes and increased length of life. During the past decades, the discovery of different stem cells facilitates ongoing attempts for attenuating many disorders, especially in vascular beds. Endothelial progenitor cells (EPCs) are a subtype of stem cells that have potent capacity to differentiate into mature endothelial cells (ECs). However, some documented studies reported an age-related decline in proliferation and function of many stem cells. There is no data on aging effect upon proliferation and morphological feature of EPCs. Methods: To show aging effect on EPCs proliferation and multipotentiality, bone marrow samples were provided from old and young cases in three different species; human, mouse and dog. After 7 days of culture, the cell morphology and clonogenic capacity were evaluated. We also calculated the mean number of colonies both in bone marrow samples from old and young subjects. To confirm the cell phenotype, isolated cells were immune-phenotyped by a panel of antibodies against Tie-2, CD133 and CD309 markers. Results: Our results showed that EPCs exhibited prominent spindle form in all bone marrow samples from young cases while the cell shape became more round by aging. Notably, the number of colonies was reduced in aged samples as compared to parallel young subject samples (P < 0.05). We also detected that the expression of endothelial related markers diminished by aging. Conclusion: The results of this study suggest that the age-related vascular abnormalities could be presumably related to the decline in stemness capacity of EPCs.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Cheraghi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran ; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Siavashi
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
274
|
Lake JE, Seang S, Kelesidis T, Currier JS, Yang OO. Telmisartan increases vascular reparative capacity in older HIV-infected adults: a pilot study. HIV CLINICAL TRIALS 2016; 17:225-232. [PMID: 27658740 DOI: 10.1080/15284336.2016.1234222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are bone marrow-derived cells that contribute to vascular repair. EPCs may be reduced in HIV-infected (HIV+) persons, contributing to cardiovascular disease (CVD). Telmisartan is an angiotensin receptor blocker that increases EPCs in HIV-uninfected adults. OBJECTIVE To assess telmisartan's effects on EPC number and immunophenotype in older HIV + adults at risk for CVD. METHODS HIV + persons ≥50 years old with HIV-1 RNA < 50 copies/mL on suppressive antiretroviral therapy and ≥1 CVD risk factor participated in a prospective, open-label, pilot study of oral telmisartan 80 mg daily for 12 weeks. Using CD34 and CD133 as markers of early maturity and KDR as a marker of endothelial lineage commitment, EPCs were quantified via flow cytometry and defined as viable CD3-/CD33-/CD19-/glycophorin- cells of four immunophenotypes: CD133+/KDR+, CD34+/KDR+, CD34+/CD133+, or CD34+/KDR+/CD133+. The primary endpoint was a 12-week change in EPC subsets (NCT01578772). RESULTS Seventeen participants (88% men, median age 60 years and peripheral CD4+ T lymphocyte count 625 cells/mm3) enrolled and completed the study. After 6 and 12 weeks of telmisartan, frequencies of all EPC immunophenotypes were higher than baseline (all p < 0.10 except week 12 CD133+/KDR+ EPC, p = 0.13). Participants with lower baseline EPC levels had the largest gains. Additionally, the percentage of CD34+ cells with endothelial commitment (KDR+) increased. CONCLUSIONS Our data suggest that telmisartan use is associated with an increase in circulating EPCs in older HIV + individuals with CVD risk factors. Further controlled studies are needed to assess whether EPC increases translate to a reduction in CVD risk in this population.
Collapse
Affiliation(s)
- Jordan E Lake
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Sophie Seang
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Theodoros Kelesidis
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Judith S Currier
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Otto O Yang
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA.,b Department of Microbiology, Immunology, and Molecular Genetics , University of California , Los Angeles , CA , USA.,c AIDS Healthcare Foundation , Los Angeles , CA , USA
| |
Collapse
|
275
|
Nadlacki B, Suuronen EJ. Biomaterial strategies to improve the efficacy of bone marrow cell therapy for myocardial infarction. Expert Opin Biol Ther 2016; 16:1501-1516. [DOI: 10.1080/14712598.2016.1235149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
276
|
Miao X, Zhang W, Huang Z, Li N. Unaltered Angiogenesis-Regulating Activities of Platelets in Mild Type 2 Diabetes Mellitus despite a Marked Platelet Hyperreactivity. PLoS One 2016; 11:e0162405. [PMID: 27612088 PMCID: PMC5017716 DOI: 10.1371/journal.pone.0162405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/22/2016] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with platelet dysfunction and impaired angiogenesis. Aim of the study is to investigate if platelet dysfunction might hamper platelet angiogenic activities in T2DM patients. Sixteen T2DM patients and gender/age-matched non-diabetic controls were studied. Flow cytometry and endothelial colony forming cell (ECFC) tube formation on matrigel were used to assess platelet reactivity and angiogenic activity, respectively. Thrombin receptor PAR1-activating peptide (PAR1-AP) induced higher platelet P-selectin expression, and evoked more rapid and intense platelet annexin V binding in T2DM patients, seen as a more rapid increase of annexin V+ platelets (24.3±6.4% vs 12.6±3.8% in control at 2 min) and a higher elevation (30.9±5.1% vs 24.3±3.0% at 8 min). However, PAR1-AP and PAR4-AP induced similar releases of angiogenic regulators from platelets, and both stimuli evoked platelet release of platelet angiogenic regulators to similar extents in T2DM and control subjects. Thus, PAR1-stimulated platelet releasate (PAR1-PR) and PAR4-PR similarly enhanced capillary-like network/tube formation of ECFCs, and the enhancements did not differ between T2DM and control subjects. Direct supplementation of platelets to ECFCs at the ratio of 1:200 enhanced ECFC tube formation even more markedly, leading to approximately 100% increases of the total branch points of ECFC tube formation, for which the enhancements were also similar between patients and controls. In conclusion, platelets from T2DM subjects are hyperreactive. Platelet activation induced by high doses of PAR1-AP, however, results in similar releases of angiogenic regulators in mild T2DM and control subjects. Platelets from T2DM and control subjects also demonstrate similar enhancements on ECFC angiogenic activities.
Collapse
Affiliation(s)
- Xinyan Miao
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Wei Zhang
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Zhangsen Huang
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Nailin Li
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
277
|
Bhatnagar A, Bolli R, Johnstone BH, Traverse JH, Henry TD, Pepine CJ, Willerson JT, Perin EC, Ellis SG, Zhao DXM, Yang PC, Cooke JP, Schutt RC, Trachtenberg BH, Orozco A, Resende M, Ebert RF, Sayre SL, Simari RD, Moyé L, Cogle CR, Taylor DA. Bone marrow cell characteristics associated with patient profile and cardiac performance outcomes in the LateTIME-Cardiovascular Cell Therapy Research Network (CCTRN) trial. Am Heart J 2016; 179:142-50. [PMID: 27595689 PMCID: PMC5014395 DOI: 10.1016/j.ahj.2016.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 06/25/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Although several preclinical studies have shown that bone marrow cell (BMC) transplantation promotes cardiac recovery after myocardial infarction, clinical trials with unfractionated bone marrow have shown variable improvements in cardiac function. METHODS To determine whether in a population of post-myocardial infarction patients, functional recovery after BM transplant is associated with specific BMC subpopulation, we examined the association between BMCs with left ventricular (LV) function in the LateTIME-CCTRN trial. RESULTS In this population, we found that older individuals had higher numbers of BM CD133(+) and CD3(+) cells. Bone marrow from individuals with high body mass index had lower CD45(dim)/CD11b(dim) levels, whereas those with hypertension and higher C-reactive protein levels had higher numbers of CD133(+) cells. Smoking was associated with higher levels of CD133(+)/CD34(+)/VEGFR2(+) cells and lower levels of CD3(+) cells. Adjusted multivariate analysis indicated that CD11b(dim) cells were negatively associated with changes in LV ejection fraction and wall motion in both the infarct and border zones. Change in LV ejection fraction was positively associated with CD133(+), CD34(+), and CD45(+)/CXCR4(dim) cells as well as faster BMC growth rates in endothelial colony forming assays. CONCLUSIONS In the LateTIME population, BM composition varied with patient characteristics and treatment. Irrespective of cell therapy, recovery of LV function was greater in patients with greater BM abundance of CD133(+) and CD34(+) cells and worse in those with higher levels of CD11b(dim) cells. Bone marrow phenotype might predict clinical response before BMC therapy and administration of selected BM constituents could potentially improve outcomes of other future clinical trials.
Collapse
Affiliation(s)
| | | | | | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN
| | | | - Carl J Pepine
- University of Florida College of Medicine, Gainesville, FL
| | - James T Willerson
- Texas Heart Institute, CHI St. Luke's Health Baylor College of Medicine Medical Center, Houston, TX
| | - Emerson C Perin
- Texas Heart Institute, CHI St. Luke's Health Baylor College of Medicine Medical Center, Houston, TX
| | | | | | | | - John P Cooke
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| | - Robert C Schutt
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| | | | - Aaron Orozco
- Texas Heart Institute, CHI St. Luke's Health Baylor College of Medicine Medical Center, Houston, TX
| | - Micheline Resende
- Texas Heart Institute, CHI St. Luke's Health Baylor College of Medicine Medical Center, Houston, TX
| | - Ray F Ebert
- National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Shelly L Sayre
- University of Texas School of Public Health, Houston, TX
| | | | - Lem Moyé
- University of Texas School of Public Health, Houston, TX.
| | | | - Doris A Taylor
- Texas Heart Institute, CHI St. Luke's Health Baylor College of Medicine Medical Center, Houston, TX
| |
Collapse
|
278
|
Boraldi F, Bartolomeo A, De Biasi S, Orlando S, Costa S, Cossarizza A, Quaglino D. Innovative Flow Cytometry Allows Accurate Identification of Rare Circulating Cells Involved in Endothelial Dysfunction. PLoS One 2016; 11:e0160153. [PMID: 27560136 PMCID: PMC5004589 DOI: 10.1371/journal.pone.0160153] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/14/2016] [Indexed: 01/20/2023] Open
Abstract
Introduction Although rare, circulating endothelial and progenitor cells could be
considered as markers of endothelial damage and repair potential, possibly
predicting the severity of cardiovascular manifestations. A number of
studies highlighted the role of these cells in age-related diseases,
including those characterized by ectopic calcification. Nevertheless, their
use in clinical practice is still controversial, mainly due to difficulties
in finding reproducible and accurate methods for their determination. Methods Circulating mature cells (CMC, CD45-, CD34+,
CD133-) and circulating progenitor cells (CPC,
CD45dim, CD34bright, CD133+) were
investigated by polychromatic high-speed flow cytometry to detect the
expression of endothelial (CD309+) or osteogenic
(BAP+) differentiation markers in healthy subjects and in
patients affected by peripheral vascular manifestations associated with
ectopic calcification. Results This study shows that: 1) polychromatic flow cytometry represents a valuable
tool to accurately identify rare cells; 2) the balance of CD309+
on CMC/CD309+ on CPC is altered in patients affected by
peripheral vascular manifestations, suggesting the occurrence of vascular
damage and low repair potential; 3) the increase of circulating cells
exhibiting a shift towards an osteoblast-like phenotype (BAP+) is
observed in the presence of ectopic calcification. Conclusion Differences between healthy subjects and patients with ectopic calcification
indicate that this approach may be useful to better evaluate endothelial
dysfunction in a clinical context.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via
Campi 287, Modena, Italy
| | - Angelica Bartolomeo
- Department of Life Sciences, University of Modena and Reggio Emilia, Via
Campi 287, Modena, Italy
| | - Sara De Biasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences,
University of Modena and Reggio Emilia, Via Campi 287, Modena,
Italy
| | - Stefania Orlando
- Department of Life Sciences, University of Modena and Reggio Emilia, Via
Campi 287, Modena, Italy
| | - Sonia Costa
- Department of Life Sciences, University of Modena and Reggio Emilia, Via
Campi 287, Modena, Italy
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences,
University of Modena and Reggio Emilia, Via Campi 287, Modena,
Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Via
Campi 287, Modena, Italy
- * E-mail:
| |
Collapse
|
279
|
Samuel R, Duda DG, Fukumura D, Jain RK. Vascular diseases await translation of blood vessels engineered from stem cells. Sci Transl Med 2016; 7:309rv6. [PMID: 26468328 DOI: 10.1126/scitranslmed.aaa1805] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of human induced pluripotent stem cells (hiPSCs) might pave the way toward a long-sought solution for obtaining sufficient numbers of autologous cells for tissue engineering. Several methods exist for generating endothelial cells or perivascular cells from hiPSCs in vitro for use in the building of vascular tissue. We discuss current developments in the generation of vascular progenitor cells from hiPSCs and the assessment of their functional capacity in vivo, opportunities and challenges for the clinical translation of engineered vascular tissue, and modeling of vascular diseases using hiPSC-derived vascular progenitor cells.
Collapse
Affiliation(s)
- Rekha Samuel
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA. Centre for Stem Cell Research, Christian Medical College, Bagayam, Vellore 632002, Tamil Nadu, India
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
280
|
Patschan S, Tampe D, Müller C, Seitz C, Herink C, Müller GA, Zeisberg E, Zeisberg M, Henze E, Patschan D. Early Endothelial Progenitor Cells (eEPCs) in systemic sclerosis (SSc) - dynamics of cellular regeneration and mesenchymal transdifferentiation. BMC Musculoskelet Disord 2016; 17:339. [PMID: 27519706 PMCID: PMC4983068 DOI: 10.1186/s12891-016-1197-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/30/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Patients with systemic sclerosis (SSc) are endagered by tissue fibrosis and by microvasculopathy, with the latter caused by endothelial cell expansion/proliferation. SSc-associated fibrosis potentially results from mesenchymal transdifferentiation of endothelial cells. Early Endothelial Progenitor Cells (eEPCs) act proangiogenic under diverse conditions. Aim of the study was to analyze eEPC regeneration and mesenchymal transdifferentiation in patients with limited and diffuse SSs (lSSc and dSSc). METHODS Patients with both, lSSc and dSSc were included into the study. The following parameters were evaluated: eEPC numbers and regeneration, concentrations of vasomodulatory mediators, mesenchymal properties of blood-derived eEPC. Serum samples of healthy subjects and SS patients were used for stimulation of cultured human eEPC, subsequently followed by analysis of mesenchymal cell characteristics and mobility. RESULTS Twenty-nine patients were included into the study. Regenerative activity of blood-derived eEPCs did not differ between Controls and patients. Circulating eEPC were significantly lower in all patients with SSc, and in limited and diffuse SSc (lSSc/dSSc). Serum concentrations of promesenchymal TGF-b was elevated in all patients with SSc. Cultured mononuclear cells from SS patients displayed higher abundances of CD31 and of CD31 and aSMA combined. Finally, serum from SSc patients inhibited migration of cultured eEPCs and the cells showed lower sensitivity towards the endothelin antagonist Bosentan. CONCLUSIONS The eEPC system, which represents an essential element of the endogenous vascular repair machinery is affected in SSc. The increased appearance of mesenchymal properties in eEPC may indicate that alterations of the cells potentially contribute to the accumulation of connective tissue and to vascular malfunction.
Collapse
Affiliation(s)
- S. Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - D. Tampe
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - C. Müller
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - C. Seitz
- Clinic of Dermatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - C. Herink
- Clinic of Dermatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - G. A. Müller
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - E. Zeisberg
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - M. Zeisberg
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - E. Henze
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - D. Patschan
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| |
Collapse
|
281
|
Development of a Modular Assay for Detailed Immunophenotyping of Peripheral Human Whole Blood Samples by Multicolor Flow Cytometry. Int J Mol Sci 2016; 17:ijms17081316. [PMID: 27529227 PMCID: PMC5000713 DOI: 10.3390/ijms17081316] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/18/2016] [Accepted: 07/28/2016] [Indexed: 12/19/2022] Open
Abstract
The monitoring of immune cells gained great significance in prognosis and prediction of therapy responses. For analyzing blood samples, the multicolor flow cytometry has become the method of choice as it combines high specificity on single cell level with multiple parameters and high throughput. Here, we present a modular assay for the detailed immunophenotyping of blood (DIoB) that was optimized for an easy and direct application in whole blood samples. The DIoB assay characterizes 34 immune cell subsets that circulate the peripheral blood including all major immune cells such as T cells, B cells, natural killer (NK) cells, monocytes, dendritic cells (DCs), neutrophils, eosinophils, and basophils. In addition, it evaluates their functional state and a few non-leukocytes that also have been associated with the outcome of cancer therapy. This DIoB assay allows a longitudinal and close-meshed monitoring of a detailed immune status in patients requiring only 2.0 mL of peripheral blood and it is not restricted to peripheral blood mononuclear cells. It is currently applied for the immune monitoring of patients with glioblastoma multiforme (IMMO-GLIO-01 trial, NCT02022384), pancreatic cancer (CONKO-007 trial, NCT01827553), and head and neck cancer (DIREKHT trial, NCT02528955) and might pave the way for immune biomarker identification for prediction and prognosis of therapy outcome.
Collapse
|
282
|
Dual Anti-Inflammatory and Anti-Angiogenic Action of miR-15a in Diabetic Retinopathy. EBioMedicine 2016; 11:138-150. [PMID: 27531575 PMCID: PMC5049929 DOI: 10.1016/j.ebiom.2016.08.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/26/2016] [Accepted: 08/06/2016] [Indexed: 11/22/2022] Open
Abstract
Activation of pro-inflammatory and pro-angiogenic pathways in the retina and the bone marrow contributes to pathogenesis of diabetic retinopathy. We identified miR-15a as key regulator of both pro-inflammatory and pro-angiogenic pathways through direct binding and inhibition of the central enzyme in the sphingolipid metabolism, ASM, and the pro-angiogenic growth factor, VEGF-A. miR-15a was downregulated in diabetic retina and bone marrow cells. Over-expression of miR-15a downregulated, and inhibition of miR-15a upregulated ASM and VEGF-A expression in retinal cells. In addition to retinal effects, migration and retinal vascular repair function was impaired in miR-15a inhibitor-treated circulating angiogenic cells (CAC). Diabetic mice overexpressing miR-15a under Tie-2 promoter had normalized retinal permeability compared to wild type littermates. Importantly, miR-15a overexpression led to modulation toward nondiabetic levels, rather than complete inhibition of ASM and VEGF-A providing therapeutic effect without detrimental consequences of ASM and VEGF-A deficiencies.
Collapse
|
283
|
Chantzichristos VG, Agouridis AP, Moutzouri E, Stellos K, Elisaf MS, Tselepis AD. Effect of rosuvastatin or its combination with omega-3 fatty acids on circulating CD34 + progenitor cells and on endothelial colony formation in patients with mixed dyslipidaemia. Atherosclerosis 2016; 251:240-247. [DOI: 10.1016/j.atherosclerosis.2016.06.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 06/24/2016] [Accepted: 06/29/2016] [Indexed: 11/29/2022]
|
284
|
Endothelial progenitor cells accelerate the resolution of deep vein thrombosis. Vascul Pharmacol 2016; 83:10-6. [DOI: 10.1016/j.vph.2015.07.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/27/2015] [Accepted: 07/11/2015] [Indexed: 11/23/2022]
|
285
|
Al Mheid I, Hayek SS, Ko YA, Akbik F, Li Q, Ghasemzadeh N, Martin GS, Long Q, Hammadah M, Maziar Zafari A, Vaccarino V, Waller EK, Quyyumi AA. Age and Human Regenerative Capacity Impact of Cardiovascular Risk Factors. Circ Res 2016; 119:801-9. [PMID: 27436845 DOI: 10.1161/circresaha.116.308461] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/19/2016] [Indexed: 01/11/2023]
Abstract
RATIONALE We investigated aging of human endogenous reparative capacity and aimed to clarify whether it is affected by presence of cardiovascular disease or its risk factors (RFs). OBJECTIVE Circulating progenitor cell (PC) levels reflect endogenous regenerative potential. The effect on PC of healthy aging compared with aging with RFs or cardiovascular disease (CVD) is unknown. We examined whether exposure to RF and CVD leads to an accelerated decline in circulating PC with increasing age. METHODS AND RESULTS In 2792 adult subjects, 498 were free of RFs (smoking, diabetes mellitus, hypertension, or hyperlipidemia), 1036 subjects had 1 to 2 RF, and 1253 had ≥3 RFs or CVD. PC were enumerated by flow cytometry as CD45(med+) mononuclear cells expressing CD34 and subsets coexpressing CD133, CXCR4, and vascular endothelial growth factor receptor-2 epitopes. Younger age, male sex, and larger body size correlated with higher PC counts (P<0.01). After multivariable adjustment, both age and RF categories were independently associated with PC counts (P<0.05), with lower PC counts in older subjects and those with higher RF burden or CVD. PC counts remained unchanged with increasing age in healthy individuals. There were significant interactions between age and RF categories (P≤0.005), such that for younger subjects (<40 years), RFs were associated with increased PC counts, whereas for older subjects (>60 years), RFs and CVD were associated with lower PC counts. CONCLUSIONS Circulating PC levels do not decline with healthy aging; RF exposure at a younger age stimulates PC mobilization, whereas continued exposure is associated with lower PC levels in later life. Over the lifespan, exposure to RFs and CVD is associated with an initial stimulation and subsequent decline in circulating PC levels, which reflect endogenous regenerative capacity.
Collapse
Affiliation(s)
- Ibhar Al Mheid
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Salim S Hayek
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Yi-An Ko
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Faysal Akbik
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Qunna Li
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Nima Ghasemzadeh
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Greg S Martin
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Qi Long
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Muhammad Hammadah
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - A Maziar Zafari
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Viola Vaccarino
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Edmund K Waller
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Arshed A Quyyumi
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA.
| |
Collapse
|
286
|
Zahran AM, Aly SS, Altayeb HA, Ali AM. Circulating endothelial cells and their progenitors in acute myeloid leukemia. Oncol Lett 2016; 12:1965-1970. [PMID: 27602121 DOI: 10.3892/ol.2016.4859] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 06/16/2016] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy characterized by the accumulation of immature myeloid progenitor cells in the bone marrow. Studies are required to investigate the prognostic and predictive value of surrogate biomarkers. Given the importance of angiogenesis in oncology in terms of pathogenesis as well as being a target for treatment, circulating endothelial cells (CECs) and endothelial progenitor cells (EPCs) are promising candidates to serve as such markers. The aim of the present study was to quantify CECs and EPCs in patients with AML at initial diagnosis and following induction chemotherapy, and to correlate these findings with the response to treatment in AML patients. The present study included 40 patients with de novo AML and 20 age- and gender-matched healthy controls. CECs and EPCs were evaluated by flow cytometry at initial diagnosis and after induction chemotherapy (3+7 protocol for AML other than M3 and all-trans-retinoic acid plus anthracycline for M3 disease). CECs and EPCs were significantly higher in AML patients at diagnosis and after induction chemotherapy than in controls. After induction chemotherapy, CECs and EPCs were significantly decreased compared with the levels at initial diagnosis. Patients who achieved complete response (n=28) had lower initial CEC and EPC levels compared with patients who did not respond to treatment. These results suggest that CEC levels are higher in AML patients and may correlate with disease status and treatment response. Further investigations are required to better determine the predictive value and implication of these cells in AML management.
Collapse
Affiliation(s)
- Asmaa Mohammed Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut 71111, Egypt
| | - Sanaa Shaker Aly
- Department of Clinical Pathology, Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Hanan Ahmed Altayeb
- Department of Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut 71111, Egypt
| | - Arwa Mohammed Ali
- Department of Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut 71111, Egypt
| |
Collapse
|
287
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
288
|
Blix ES, Husebekk A. Raiders of the lost mark - endothelial cells and their role in transplantation for hematologic malignancies. Leuk Lymphoma 2016; 57:2752-2762. [PMID: 27396981 DOI: 10.1080/10428194.2016.1201566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Endothelial cells (EC) are crucial for normal angiogenesis and important for patients with leukemia, myeloma, and lymphoma during and after hematopoietic stem cell transplantation (HSCT). Knowledge of endothelial dysfunction in hematologic malignancies is provided by translational studies analyzing soluble endothelial markers, morphologic and functional changes of EC cultured in patients' sera or enumeration of circulating EC or endothelial progenitor cells (EPC). EC are important for stem cell homing and maintenance. Endothelial activation or damage is a central component in the pathogenesis of several complications after HSCT, like acute and chronic graft-versus-host disease, sinusoidal obstruction syndrome, capillary leak syndrome, engraftment syndrome, diffuse alveolar syndrome, idiopathic pneumonia syndrome, and transplant-associated microangiopathy. Finally, EC or EPC may facilitate tumor cell survival thus representing potential factors for both disease progression and relapse in hematologic malignancies.
Collapse
Affiliation(s)
- Egil S Blix
- a Immunology Research Group, Institute of Medical Biology , UiT the Arctic University of Norway , Tromsø , Norway.,b Department of Oncology , University Hospital of North Norway , Tromsø , Norway
| | - Anne Husebekk
- a Immunology Research Group, Institute of Medical Biology , UiT the Arctic University of Norway , Tromsø , Norway
| |
Collapse
|
289
|
Dong Y, Sun Q, Liu T, Wang H, Jiao K, Xu J, Liu X, Liu H, Wang W. Nitrative Stress Participates in Endothelial Progenitor Cell Injury in Hyperhomocysteinemia. PLoS One 2016; 11:e0158672. [PMID: 27391949 PMCID: PMC4938535 DOI: 10.1371/journal.pone.0158672] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/20/2016] [Indexed: 01/01/2023] Open
Abstract
In order to investigate the role of nitrative stress in vascular endothelial injury in hyperhomocysteinemia (HHcy), thirty healthy adult female Wistar rats were randomly divided into three groups: control, hyperhomocysteinemia model, and hyperhomocysteinemia with FeTMPyP (peroxynitrite scavenger) treatment. The endothelium-dependent dilatation of thoracic aorta in vitro was determined by response to acetylcholine (ACh). The histological changes in endothelium were assessed by HE staining and scanning electron microscopy (SEM). The expression of 3-nitrotyrosine (NT) in thoracic aorta was demonstrated by immunohistochemistry and immunofluorescence, and the number of circulating endothelial progenitor cells (EPCs) was quantified by flow cytometry. Hyperhomocysteinemia caused significant endothelial injury and dysfunction including vasodilative and histologic changes, associated with higher expression of NT in thoracic aorta. FeTMPyP treatment reversed these injuries significantly. Further, the effect of nitrative stress on cultured EPCs in vitro was investigated by administering peroxynitrite donor (3-morpholino-sydnonimine, SIN-1) and peroxynitrite scavenger (FeTMPyP). The roles of nitrative stress on cell viability, necrosis and apoptosis were evaluated with 3-(4,5-dimethylthiazol)-2,5-diphenyl tetrazolium (MTT) assay, lactate dehydrogenase (LDH) release assay and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay, respectively. Also, the phospho-eNOS expression and tube formation in Matrigel of cultured EPCs was detected. Our data showed that the survival of EPCs was much lower in SIN-1 group than in vehicle group, both the apoptosis and necrosis of EPCs were much more severe, and the p-eNOS expression and tube formation in Matrigel were obviously declined. Subsequent pretreatment with FeTMPyP reversed these changes. Further, pretreatment with FeTMPyP reversed homocysteine-induced EPC injury. In conclusion, this study indicates that nitrative stress plays a role in vascular endothelial injury in hyperhomocysteinemia, as well as induces endothelial progenitor cell injury directly.
Collapse
Affiliation(s)
- Yu Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Qi Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Teng Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Huanyuan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Kun Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Jiahui Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Xin Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
290
|
Matrix metalloproteinase-13 participates in neuroprotection and neurorepair after cerebral ischemia in mice. Neurobiol Dis 2016; 91:236-46. [DOI: 10.1016/j.nbd.2016.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 03/09/2016] [Accepted: 03/17/2016] [Indexed: 12/22/2022] Open
|
291
|
Prisco AR, Hoffmann BR, Kaczorowski CC, McDermott-Roe C, Stodola TJ, Exner EC, Greene AS. Tumor Necrosis Factor α Regulates Endothelial Progenitor Cell Migration via CADM1 and NF-kB. Stem Cells 2016; 34:1922-33. [PMID: 26867147 PMCID: PMC4931961 DOI: 10.1002/stem.2339] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
Shortly after the discovery of endothelial progenitor cells (EPCs) in 1997, many clinical trials were conducted using EPCs as a cellular based therapy with the goal of restoring damaged organ function by inducing growth of new blood vessels (angiogenesis). Results were disappointing, largely because the cellular and molecular mechanisms of EPC-induced angiogenesis were not clearly understood. Following injection, EPCs must migrate to the target tissue and engraft prior to induction of angiogenesis. In this study EPC migration was investigated in response to tumor necrosis factor α (TNFα), a pro-inflammatory cytokine, to test the hypothesis that organ damage observed in ischemic diseases induces an inflammatory signal that is important for EPC homing. In this study, EPC migration and incorporation were modeled in vitro using a coculture assay where TNFα treated EPCs were tracked while migrating toward vessel-like structures. It was found that TNFα treatment of EPCs increased migration and incorporation into vessel-like structures. Using a combination of genomic and proteomic approaches, NF-kB mediated upregulation of CADM1 was identified as a mechanism of TNFα induced migration. Inhibition of NF-kB or CADM1 significantly decreased migration of EPCs in vitro suggesting a role for TNFα signaling in EPC homing during tissue repair. Stem Cells 2016;34:1922-1933.
Collapse
Affiliation(s)
- Anthony R. Prisco
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Brian R. Hoffmann
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
- Medical College of Wisconsin, Department of Medicine, Division of Cardiology, Cardiovascular Center, Milwaukee, WI
| | - Catherine C. Kaczorowski
- University of Tennessee Health Science Center, Department of Anatomy and Neurobiology, Memphis, TN
| | - Chris McDermott-Roe
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Human and Molecular Genetics Center, Milwaukee, WI
| | - Timothy J. Stodola
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Eric C. Exner
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Andrew S. Greene
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| |
Collapse
|
292
|
Hajjar I, Goldstein FC, Waller EK, Moss LD, Quyyumi A. Circulating Progenitor Cells is Linked to Cognitive Decline in Healthy Adults. Am J Med Sci 2016; 351:147-52. [PMID: 26897269 DOI: 10.1016/j.amjms.2015.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/14/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cognitive and cardiovascular disorders share many risk factors. Higher bone-marrow derived progenitor cells (PC) in blood are associated with lower rates of cardiovascular events but the association of PC with cognitive function is unclear. The objective of this study was to assess the association between PC and cognition in a sample of healthy adults enrolled in a cohort study. MATERIALS AND METHODS A random sample of employees at Emory University and Georgia Institute of Technology were followed for 4 years and underwent yearly vascular and cognitive assessment (N = 430, mean age = 49.2 years, 70% women, and 27% African-American). Cognition was assessed using computerized versions of 15 cognitive tests and principal component analysis was used for deriving cognitive scores: executive function, memory and working memory. PC were defined as mononuclear cells with specific surface markers (7 phenotypes). Decreased cognition in a domain was defined as performing below the lowest quartile for the corresponding domain at baseline. Generalized estimating equations were used to investigate associations between PC and cognition. RESULTS Higher PC levels at baseline were associated with lower risk of cognitive decline in the executive and working memory domains during the follow-up period (P < 0.002 for all PC phenotypes). Further, the degree of decline in PC over the follow-up period was correlated with a corresponding decline in performances in all 3 cognitive domains over the same period (All P < 0.002). CONCLUSION Lower PC and greater yearly declines in PC are associated with greater cognitive decline. These findings suggest the role for PC in neurocognitive aging.
Collapse
Affiliation(s)
- Ihab Hajjar
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Emory University, Atlanta, Georgia.
| | | | - Edmund K Waller
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Lauren D Moss
- Program Management Associate, Sarah Cannon Research Institute, Nashville, Tennessee
| | - Arshed Quyyumi
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
293
|
Aman J, Weijers EM, van Nieuw Amerongen GP, Malik AB, van Hinsbergh VWM. Using cultured endothelial cells to study endothelial barrier dysfunction: Challenges and opportunities. Am J Physiol Lung Cell Mol Physiol 2016; 311:L453-66. [PMID: 27343194 DOI: 10.1152/ajplung.00393.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/20/2016] [Indexed: 12/24/2022] Open
Abstract
Despite considerable progress in the understanding of endothelial barrier regulation and the identification of approaches that have the potential to improve endothelial barrier function, no drug- or stem cell-based therapy is presently available to reverse the widespread vascular leak that is observed in acute respiratory distress syndrome (ARDS) and sepsis. The translational gap suggests a need to develop experimental approaches and tools that better mimic the complex environment of the microcirculation in which the vascular leak develops. Recent studies have identified several elements of this microenvironment. Among these are composition and stiffness of the extracellular matrix, fluid shear stress, interaction of endothelial cells (ECs) with pericytes, oxygen tension, and the combination of toxic and mechanic injurious stimuli. Development of novel cell culture techniques that integrate these elements would allow in-depth analysis of EC biology that closely approaches the (patho)physiological conditions in situ. In parallel, techniques to isolate organ-specific ECs, to define EC heterogeneity in its full complexity, and to culture patient-derived ECs from inducible pluripotent stem cells or endothelial progenitor cells are likely to advance the understanding of ARDS and lead to development of therapeutics. This review 1) summarizes the advantages and pitfalls of EC cultures to study vascular leak in ARDS, 2) provides an overview of elements of the microvascular environment that can directly affect endothelial barrier function, and 3) discusses alternative methods to bridge the gap between basic research and clinical application with the intent of improving the translational value of present EC culture approaches.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Department of Pulmonary Diseases, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands;
| | - Ester M Weijers
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Geerten P van Nieuw Amerongen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
294
|
Endothelial progenitor cells promote tumor growth and progression by enhancing new vessel formation. Oncol Lett 2016; 12:793-799. [PMID: 27446353 PMCID: PMC4950911 DOI: 10.3892/ol.2016.4733] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/02/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor growth and progression require new blood vessel formation to deliver nutrients and oxygen for further cell proliferation and to create a neovascular network exit for tumor cell metastasis. Endothelial progenitor cells (EPCs) are a bone marrow (BM)-derived stem cell population that circulates in the peripheral circulation and homes to the tumor bed to participate in new blood vessel formation. In addition to structural support to nascent vessels, these cells can also regulate the angiogenic process by paracrine secretion of a number of proangiogenic growth factors and cytokines, thus playing a crucial role in tumor neovascularization and development. Inhibition of EPC-mediated new vessel formation may be a promising therapeutic strategy in tumor treatment. EPC-mediated neovascularization is a complex process that includes multiple steps and requires a series of cytokines and modulators, thus understanding the underlying mechanisms may provide anti-neovasculogenesis targets that may be blocked for the prevention of tumor development. The present review stresses the process and contribution of EPCs to the formation of new blood vessels in solid tumors, in an attempt to gain an improved understanding of the underlying cellular and molecular mechanisms involved, and to provide a potential effective therapeutic target for cancer treatment.
Collapse
|
295
|
Rigato M, Avogaro A, Fadini GP. Levels of Circulating Progenitor Cells, Cardiovascular Outcomes and Death. Circ Res 2016; 118:1930-9. [DOI: 10.1161/circresaha.116.308366] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/12/2016] [Indexed: 11/16/2022]
Abstract
Rationale:
Circulating progenitor cells (CPCs), including endothelial progenitor cells (EPCs) are biologically related to many aspects of cardiovascular disease, as they promote angiogenesis and vascular repair.
Objective:
We herein aimed to meta-analyze studies reporting the prognostic role of the CPC/EPC measure on cardiovascular outcomes and death.
Methods and Results:
We screened the English-language literature for longitudinal studies reporting the association between baseline CPC/EPC levels, future cardiovascular events, and death. We retrieved 28 studies, 21 of which contained poolable data and entered the meta-analysis, for a total of 4155 patients, mostly with a high baseline cardiovascular risk. Sixty percent of the studies met at least 11 of 16 items of quality assessment. Overall, reduced CPC/EPC levels were associated with a ≈2-fold increased risk of future cardiovascular events and cardiovascular death. The most predictive phenotype was CD34
+
CD133
+
: low versus high levels predicted cardiovascular events, restenosis after endovascular intervention, cardiovascular death, and all-cause mortality. Heterogeneity among studies and according to the CPC/EPC phenotype was generally high. Excluding studies for which the risk estimate had to be extrapolated or limiting the analyses to higher quality studies still indicated a significant risk for future cardiovascular events and death in patients with low versus high progenitor cell counts.
Conclusions:
This meta-analysis shows that a reduction in the levels of circulating cells putatively provided with vasculoregenerative properties represents a risk factor for adverse cardiovascular outcomes and death.
Collapse
Affiliation(s)
- Mauro Rigato
- From the Department of Medicine, University of Padova, Padova, Italy
| | - Angelo Avogaro
- From the Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- From the Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
296
|
Ming GF, Tang YJ, Hu K, Chen Y, Huang WH, Xiao J. Visfatin attenuates the ox-LDL-induced senescence of endothelial progenitor cells by upregulating SIRT1 expression through the PI3K/Akt/ERK pathway. Int J Mol Med 2016; 38:643-9. [DOI: 10.3892/ijmm.2016.2633] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 05/26/2016] [Indexed: 11/05/2022] Open
|
297
|
Hayek SS, MacNamara J, Tahhan AS, Awad M, Yadalam A, Ko YA, Healy S, Hesaroieh I, Ahmed H, Gray B, Sher SS, Ghasemzadeh N, Patel R, Kim J, Waller EK, Quyyumi AA. Circulating Progenitor Cells Identify Peripheral Arterial Disease in Patients With Coronary Artery Disease. Circ Res 2016; 119:564-71. [PMID: 27267067 DOI: 10.1161/circresaha.116.308802] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/03/2016] [Indexed: 01/23/2023]
Abstract
RATIONALE Peripheral arterial disease (PAD) is a clinical manifestation of extracoronary atherosclerosis. Despite sharing the same risk factors, only 20% to 30% of patients with coronary artery disease (CAD) develop PAD. Decline in the number of bone marrow-derived circulating progenitor cells (PCs) is thought to contribute to the pathogenesis of atherosclerosis. Whether specific changes in PCs differentiate patients with both PAD and CAD from those with CAD alone is unknown. OBJECTIVE Determine whether differences exist in PCs counts of CAD patients with and without known PAD. METHODS AND RESULTS 1497 patients (mean age: 65 years; 62% men) with known CAD were identified in the Emory Cardiovascular Biobank. Presence of PAD (n=308) was determined by history, review of medical records, or imaging and was classified as carotid (53%), lower extremity (41%), upper extremity (3%), and aortic disease (33%). Circulating PCs were enumerated by flow cytometry. Patients with CAD and PAD had significantly lower PC counts compared with those with only CAD. In multivariable analysis, a 50% decrease in cluster of differentiation 34 (CD34+) or CD34+/vascular endothelial growth factor receptor-2 (VEGFR2+) counts was associated with a 31% (P=0.032) and 183% (P=0.002) increase in the odds of having PAD, respectively. CD34+ and CD34+/VEGFR2+ counts significantly improved risk prediction metrics for prevalent PAD. Low CD34+/VEGFR2+ counts were associated with a 1.40-fold (95% confidence interval, 1.03-1.91) and a 1.64-fold (95% confidence interval, 1.07-2.50) increases in the risk of mortality and PAD-related events, respectively. CONCLUSIONS PAD is associated with low CD34+ and CD34+/VEGFR2+ PC counts. Whether low PC counts are useful in screening for PAD needs to be investigated.
Collapse
Affiliation(s)
- Salim S Hayek
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - James MacNamara
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Ayman S Tahhan
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Mosaab Awad
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Adithya Yadalam
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Yi-An Ko
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Sean Healy
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Iraj Hesaroieh
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Hina Ahmed
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Brandon Gray
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Salman S Sher
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Nima Ghasemzadeh
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Riyaz Patel
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Jinhee Kim
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Edmund K Waller
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA
| | - Arshed A Quyyumi
- From the Division of Cardiology (S.S.H., M.A., A.Y., S.H., I.H., H.A., B.G., S.S.S., N.G., R.P., A.A.Q.) and Department of Internal Medicine, Emory University School of Medicine, Atlanta, GA (J.M., A.S.T.); and Department of Biostatistics and Bioinformatics (Y.-A.K.) and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA.
| |
Collapse
|
298
|
Koller L, Hohensinner P, Sulzgruber P, Blum S, Maurer G, Wojta J, Hülsmann M, Niessner A. Prognostic relevance of circulating endothelial progenitor cells in patients with chronic heart failure. Thromb Haemost 2016; 116:309-16. [PMID: 27412580 DOI: 10.1160/th16-01-0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022]
Abstract
Novel strategies for a tailored risk prediction in chronic heart failure (CHF) are crucial to identify patients at very high risk for an improved patient management and to specify treatment regimens. Endothelial progenitor cells (EPCs) are an important endogenous repair mechanism with the ability to counteract endothelial injury and the possibility of new vessel formation. We hypothesised that exhaustion of circulating EPCs may be a suitable prognostic biomarker in patients with CHF. EPCs, defined as CD34+CD45dimKDR+ cells, were analysed using fluorescence-activated cell sorting. EPCs were measured in 185 patients with CHF including 87 (47 %) patients with ischaemic aetiology and 98 (53 %) patients with non-ischaemic CHF and followed for a median time of 2.7 years. During this period, 34.7 % of patients experienced the primary study endpoint all-cause mortality. EPC count was a significant and independent inverse predictor of mortality with an hazard ratio hazard ratio (HR) per increase of one standard deviation (1-SD) of 0.47 (95 % confidence interval [CI]: 0.35-0.61; p<0.001) and remained significant after multivariable adjustment for a comprehensive set of cardiovascular risk factors and potential confounders with a HR per 1-SD of 0.54 (95 % CI: 0.4-0.73; p<0.001). EPCs further demonstrated additional prognostic information indicated by improvements in C-statistic, net reclassification index and integrated discrimination increment. In conclusion, in our study circulating EPCs turned out as strong and independent inverse predictors of mortality underlining the importance of an impaired endothelial repair mechanism in the pathophysiology and progression of CHF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alexander Niessner
- Alexander Niessner, MD, MSc, Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria, Tel.: +43 1 404004614, Fax: +43 1 404004216, E-mail:
| |
Collapse
|
299
|
|
300
|
Cappellari R, D'Anna M, Avogaro A, Fadini GP. Plerixafor improves the endothelial health balance. The effect of diabetes analysed by polychromatic flow cytometry. Atherosclerosis 2016; 251:373-380. [PMID: 27255499 DOI: 10.1016/j.atherosclerosis.2016.05.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/03/2016] [Accepted: 05/18/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Diabetes damages the endothelium and reduces the availability of bone marrow (BM)-derived endothelial progenitor cells (EPCs). The mobilization of hematopoietic stem cells (HSCs) and EPCs in response to G-CSF is impaired by diabetes, owing to CXCL12 dysregulation. We have previously shown that the CXCR4/CXCL12 disruptor plerixafor rescues HSC and EPC mobilization in diabetes. We herein explored the effects of plerixafor on HSCs, EPCs, and circulating endothelial cells (CECs) in patients with and without diabetes. METHODS We re-analysed data gathered in the NCT02056210 trial, wherein patients with (n = 10) and without diabetes (n = 10) received plerixafor to test stem/progenitor cell mobilization. We applied a novel and very specific polychromatic flow cytometry (PFC) approach to identify and quantify HSCs, EPCs, and CECs. RESULTS We found that 7-AAD(-)Syto16(+)CD34(+)CD45(dim) HSC levels determined by PFC strongly correlated to the traditional enumeration of CD34(+) cells, whereas 7-AAD(-)Syto16(+)CD34(+)CD45(neg)KDR(+) EPCs were unrelated to the traditional enumeration of CD34(+)KDR(+) cells. Using PFC, we confirmed that plerixafor induces rapid mobilization of HSCs and EPCs in both groups, with a marginally significant defect in patients with diabetes. Plerixafor reduced live (7-AAD(-)) and dead (7-AAD(+)) Syto16(+)CD34(bright)CD45(neg)CD146(+) CECs more in patients without than in those with diabetes. The EPC/CEC ratio, a measure of the vascular health balance, was increased by plerixafor, but less prominently in patients with that in those without diabetes. CONCLUSIONS In addition to rescuing defective mobilization associated with diabetes, plerixafor improves the balance between EPCs and CECs, but the latter effect is blunted in patients with diabetes.
Collapse
Affiliation(s)
- Roberta Cappellari
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Marianna D'Anna
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine, 35128 Padova, Italy.
| |
Collapse
|