351
|
Rigato M, Bittante C, Albiero M, Avogaro A, Fadini GP. Circulating Progenitor Cell Count Predicts Microvascular Outcomes in Type 2 Diabetic Patients. J Clin Endocrinol Metab 2015; 100:2666-72. [PMID: 25942480 DOI: 10.1210/jc.2015-1687] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Diabetes reduces the levels of circulating progenitor cells (CPCs) and endothelial progenitor cells (EPCs), which promote vascular repair and are inversely correlated with cardiovascular risk. OBJECTIVE The objective of the study was to test whether CPC/EPC levels predict onset/progression of microangiopathy in a cohort of type 2 diabetic (T2D) patients. DESIGN This was a pseudoprospective study with a 3.9-year follow-up. SETTING The study was conducted at a tertial referral diabetes outpatient clinic. PATIENTS A total of 187 T2D patients having a baseline determination of CPCs/EPCs participated in the study. INTERVENTION Baseline data on demographics, anthropometrics, concomitant risk factors, diabetic complications, and medications were collected. MAIN OUTCOME MEASURE Onset or progression of microangiopathy was assessed at follow-up compared with baseline. RESULTS New onset or progression of microalbuminuria, chronic kidney disease, retinopathy, and neuropathy occurred in 70 patients (9.5%/y). After controlling the false discovery rate, baseline CD34(+) CPCs and EPCs were significantly lower in patients with onset/progression of microalbuminuria and any microangiopathy. Patients with baseline CD34(+) CPC or CD133(+)/kinase insert domain-containing receptor(+)/EPC levels below the median were more likely to experience worsening microangiopathy than those with high cell levels. Independently from confounders, including age, sex, glycated hemoglobin, and diabetes duration, CD34(+) cells predicted onset/progression of microalbuminuria, retinopathy, and any microangiopathy in false discovery rate-adjusted analyses. A low CD34(+) cell count limited the beneficial effects of renin-angiotensin system blockers on microalbuminuria progression. CONCLUSIONS Levels of circulating (endothelial) progenitor cells predict microvascular outcomes in T2D. Together with previous studies showing an association with cardiovascular events, these data indicate that CPCs/EPCs represent biomarkers of the global complication burden in diabetes.
Collapse
Affiliation(s)
- Mauro Rigato
- Division of Metabolic Diseases, Department of Medicine (M.R., C.B., M.A., A.A., G.P.F.), University of Padova, 35128 Padova, Italy; and Venetian Institute of Molecular Medicine (A.A., G.P.F.), 35128 Padova, Italy
| | - Cristina Bittante
- Division of Metabolic Diseases, Department of Medicine (M.R., C.B., M.A., A.A., G.P.F.), University of Padova, 35128 Padova, Italy; and Venetian Institute of Molecular Medicine (A.A., G.P.F.), 35128 Padova, Italy
| | - Mattia Albiero
- Division of Metabolic Diseases, Department of Medicine (M.R., C.B., M.A., A.A., G.P.F.), University of Padova, 35128 Padova, Italy; and Venetian Institute of Molecular Medicine (A.A., G.P.F.), 35128 Padova, Italy
| | - Angelo Avogaro
- Division of Metabolic Diseases, Department of Medicine (M.R., C.B., M.A., A.A., G.P.F.), University of Padova, 35128 Padova, Italy; and Venetian Institute of Molecular Medicine (A.A., G.P.F.), 35128 Padova, Italy
| | - Gian Paolo Fadini
- Division of Metabolic Diseases, Department of Medicine (M.R., C.B., M.A., A.A., G.P.F.), University of Padova, 35128 Padova, Italy; and Venetian Institute of Molecular Medicine (A.A., G.P.F.), 35128 Padova, Italy
| |
Collapse
|
352
|
Fadini GP, Albiero M, Vigili de Kreutzenberg S, Avogaro A. Hypoglycemia affects the changes in endothelial progenitor cell levels during insulin therapy in type 2 diabetic patients. J Endocrinol Invest 2015; 38:733-8. [PMID: 25722222 DOI: 10.1007/s40618-015-0247-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/20/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE Hypoglycemia is a barrier to the achievement of glycemic targets and limits the beneficial effects of improved glucose control on cardiovascular outcomes in type 2 diabetes (T2D). Circulating endothelial progenitor cells (EPCs) participate in cardiovascular homeostasis and predict future cardiovascular events. Therefore, we herein analyzed the association between occurrence of hypoglycemia and EPC changes in T2D patients after optimization of glucose control with basal insulin therapy. METHODS In the NCT00699686 trial, 42 T2D insulin-naïve patients received a 3 + 3-month cross-over therapy with glargine and detemir. There were 43 minor and 2 severe hypoglycemic episodes in 19 patients (45.2 %, 0.54 episodes/patient/year). Changes in EPCs were analyzed in relation to the occurrence of hypoglycemia during the trial. RESULTS Patients with hypoglycemia had a higher final HbA1c at 6 months than patients without, although absolute HbA1c changes were not significantly different. Though PCs increased at study end, in patients experiencing at least 1 hypoglycemic episode, the changes in CD34(+), CD133(+) progenitor cells and CD34(+)KDR(+) EPCs were significantly lower than the respective changes in patients without incident hypoglycemia, even after correcting for confounders. During treatment with detemir, which induced >twofold less hypoglycemia than glargine, CD34(+)KDR(+) EPCs increased significantly more than during treatment with glargine. CONCLUSIONS In naïve T2D patients initiating basal insulin, hypoglycemia prevents the increase in vasculoprotective PCs. Clinically, these data strengthen the importance of avoiding hypoglycemia to improve cardiovascular outcomes during the treatment of T2D.
Collapse
Affiliation(s)
- G P Fadini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy.
| | - M Albiero
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| | | | - A Avogaro
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padua, Italy
| |
Collapse
|
353
|
Zhang GY, Wu LC, Liao T, Chen G, Chen YH, Meng XC, Wang AY, Chen SY, Lin K, Lin DM, Gao WY, Li QF. Altered circulating endothelial progenitor cells in patients with keloid. Clin Exp Dermatol 2015; 41:152-5. [PMID: 26121920 DOI: 10.1111/ced.12695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2015] [Indexed: 11/30/2022]
Abstract
Evidence has suggested that vascular endothelial growth factor (VEGF), a crucial growth factor in regulating endothelial progenitor cells (EPCs), plays a central role in keloid formation. However, the levels of circulating EPCs in patients with keloid have not yet been explored. The aim of this study was to determine the number of circulating EPCs in patients with keloid. Circulating EPCs (defined as CD45- CD34+CD133+VEGFR2+cells) and VEGF levels from 39 patients with keloid and 22 healthy controls (HCs) were assessed by flow cytometry and ELISA, respectively. EPCs were detectable in the peripheral blood of patients with keloid. The number of circulating EPCs and the levels of plasma VEGF were significantly higher in patients with keloid than in HCs. However, no correlation was found between the number of circulating EPCs and the serum VEGF levels. This study provides the first evidence that EPCs are increased in the peripheral blood of patients with keloid. Understanding the roles of EPCs in keloid fromation may lead to the development of novel therapeutic strategies for keloid.
Collapse
Affiliation(s)
- G-Y Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - L-C Wu
- Department of Dermatology, the Eastern Hospital of the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - T Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - G Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Y-H Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - X-C Meng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - A-Y Wang
- Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - S-Y Chen
- Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - K Lin
- Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - D-M Lin
- Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - W-Y Gao
- Department of Hand and Plastic Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Q-F Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
354
|
Kang HS, Wang KC, Kim SK. Circulating Vascular Progenitor Cells in Moyamoya Disease. J Korean Neurosurg Soc 2015; 57:428-31. [PMID: 26180610 PMCID: PMC4502239 DOI: 10.3340/jkns.2015.57.6.428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 03/30/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022] Open
Abstract
Various approaches have been attempted in translational moyamoya disease research. One promising material for modeling and treating this disease is vascular progenitor cells, which can be acquired and expanded from patient peripheral blood. These cells may provide a novel experimental model and enable us to obtain insights regarding moyamoya disease pathogenesis. We briefly present the recent accomplishments in regard to the studies of vascular progenitor cells in moyamoya disease.
Collapse
Affiliation(s)
- Hyun-Seung Kang
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu-Chang Wang
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea. ; Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-Ki Kim
- Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea. ; Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
355
|
Sulindac metabolites decrease cerebrovascular malformations in CCM3-knockout mice. Proc Natl Acad Sci U S A 2015; 112:8421-6. [PMID: 26109568 DOI: 10.1073/pnas.1501352112] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cerebral cavernous malformation (CCM) is a disease of the central nervous system causing hemorrhage-prone multiple lumen vascular malformations and very severe neurological consequences. At present, the only recommended treatment of CCM is surgical. Because surgery is often not applicable, pharmacological treatment would be highly desirable. We describe here a murine model of the disease that develops after endothelial-cell-selective ablation of the CCM3 gene. We report an early, cell-autonomous, Wnt-receptor-independent stimulation of β-catenin transcription activity in CCM3-deficient endothelial cells both in vitro and in vivo and a triggering of a β-catenin-driven transcription program that leads to endothelial-to-mesenchymal transition. TGF-β/BMP signaling is then required for the progression of the disease. We also found that the anti-inflammatory drugs sulindac sulfide and sulindac sulfone, which attenuate β-catenin transcription activity, reduce vascular malformations in endothelial CCM3-deficient mice. This study opens previously unidentified perspectives for an effective pharmacological therapy of intracranial vascular cavernomas.
Collapse
|
356
|
Lee HJ, Kim W, Kim WS, Woo JS, Kim YG, Moon JY, Lee SH, Ihm CG, Lee TW, Jeong KH. Circulating Endothelial Progenitor Cell Levels Predict Cardiovascular Events in End-Stage Renal Disease Patients on Maintenance Hemodialysis. Nephron Clin Pract 2015; 130:151-8. [PMID: 26089157 DOI: 10.1159/000430471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/13/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The number of circulating endothelial progenitor cells (EPCs) has been identified as a surrogate biologic marker for vascular function and cumulative cardiovascular (CV) risk in the general population. Patients with end-stage renal disease (ESRD) on hemodialysis (HD) have markedly decreased EPC counts and function. We hypothesized that the number of circulating EPCs predicts death from all causes and CV events in patients with ESRD on HD. METHODS We quantified the EPCs in blood samples from 70 patients with ESRD on HD. Circulating EPCs were counted by flow cytometry as the number of CD45(low)CD34(+)VEGFR2(+) cells. Death from all causes and CV events served as outcome variables over a median follow-up period of 20 months. RESULTS It has been postulated that the number of circulating EPCs at baseline ranged from 1 to 350 cells/200 μl, with a mean of ± standard deviation (SD) of 26.0 ± 48.2 cells/200 μl. The median, lowest and highest tertiles of EPC counts were 11.0, 9.0, and 17.0 cells/200 μl, respectively. Patients with the lowest tertile EPC counts had significantly higher rates of CV events, but mortality was similar between the two groups. After adjusting for these risk factors, HbA1c and the lowest tertile EPC count remained as independent predictors of CV events. A cutoff value of 9.5 cells/200 μl maximized the power of the EPC count to predict future CV events as determined by ROC curve analysis. CONCLUSIONS Reduced circulating EPC counts independently predicted CV events in 70 patients with ESRD on maintenance HD. Circulating EPCs may play a role in vascular repair, thereby affecting the clinical course of CV events.
Collapse
Affiliation(s)
- Hong Joo Lee
- Department of Nephrology, Seoul Red Cross Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
357
|
Abstract
The vasculature plays an indispensible role in organ development and maintenance of tissue homeostasis, such that disturbances to it impact greatly on developmental and postnatal health. Although cell turnover in healthy blood vessels is low, it increases considerably under pathological conditions. The principle sources for this phenomenon have long been considered to be the recruitment of cells from the peripheral circulation and the re-entry of mature cells in the vessel wall back into cell cycle. However, recent discoveries have also uncovered the presence of a range of multipotent and lineage-restricted progenitor cells in the mural layers of postnatal blood vessels, possessing high proliferative capacity and potential to generate endothelial, smooth muscle, hematopoietic or mesenchymal cell progeny. In particular, the tunica adventitia has emerged as a progenitor-rich compartment with niche-like characteristics that support and regulate vascular wall progenitor cells. Preliminary data indicate the involvement of some of these vascular wall progenitor cells in vascular disease states, adding weight to the notion that the adventitia is integral to vascular wall pathogenesis, and raising potential implications for clinical therapies. This review discusses the current body of evidence for the existence of vascular wall progenitor cell subpopulations from development to adulthood and addresses the gains made and significant challenges that lie ahead in trying to accurately delineate their identities, origins, regulatory pathways, and relevance to normal vascular structure and function, as well as disease.
Collapse
Affiliation(s)
- Peter J Psaltis
- From the Department of Medicine, University of Adelaide and Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia (P.J.P.); Monash Cardiovascular Research Centre, Monash University, Clayton, Victoria, Australia (P.J.P.); and Department of Internal Medicine, University of Kansas School of Medicine (R.D.S.)
| | - Robert D Simari
- From the Department of Medicine, University of Adelaide and Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia (P.J.P.); Monash Cardiovascular Research Centre, Monash University, Clayton, Victoria, Australia (P.J.P.); and Department of Internal Medicine, University of Kansas School of Medicine (R.D.S.).
| |
Collapse
|
358
|
Kachamakova-Trojanowska N, Bukowska-Strakova K, Zukowska M, Dulak J, Jozkowicz A. The real face of endothelial progenitor cells - Circulating angiogenic cells as endothelial prognostic marker? Pharmacol Rep 2015; 67:793-802. [PMID: 26321283 DOI: 10.1016/j.pharep.2015.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 02/08/2023]
Abstract
Endothelial progenitor cells (EPCs) have been extensively studied for almost 19 years now and were considered as a potential marker for endothelial regeneration ability. On the other hand, circulating endothelial cells (CEC) were studied as biomarker for endothelial injury. Yet, in the literature, there is also huge incoherency in regards to terminology and protocols used. This results in misleading conclusions on the role of so called "EPCs", especially in the clinical field. The discrepancies are mainly due to strong phenotypic overlap between EPCs and circulating angiogenic cells (CAC), therefore changes in "EPC" terminology have been suggested. Other factors leading to inconsistent results are varied definitions of the studied populations and the lack of universal data reporting, which could strongly affect data interpretation. The current review is focused on controversies concerning the use of "EPCs"/CAC and CEC as putative endothelial diagnostic markers.
Collapse
Affiliation(s)
- Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Monika Zukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
359
|
Vinci MC, Piacentini L, Chiesa M, Saporiti F, Colombo GI, Pesce M. Inflammatory environment and oxidized LDL convert circulating human proangiogenic cells into functional antigen-presenting cells. J Leukoc Biol 2015; 98:409-21. [PMID: 25990243 DOI: 10.1189/jlb.3a0814-412rr] [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] [Received: 08/27/2014] [Accepted: 04/23/2015] [Indexed: 11/24/2022] Open
Abstract
The function of human circulating PACs has been described extensively. However, little focus has been placed on understanding how these cells differ in their functions in the presence of microenvironments mimicking vascular inflammation. We hypothesized that exposure to proinflammatory cytokines or the oxLDL, an autoantigen abundant in advanced atherosclerotic plaques, converts PACs into immune-modulating/proinflammatory cells. Hence, we examined the effect of oxLDL and inflammatory stimuli on their phenotype by use of a functional genomics model based on secretome and whole genome transcriptome profiling. PACs obtained from culturing a PBMC fraction in angiogenic medium were primed with DC differentiation cytokines and then exposed to proinflammatory cytokines or oxLDL. Under these conditions, PACs converted into APCs, expressed maturation markers CD80 and CD83, and showed an increased up-regulation of CD86. APCcy and APCox induced a robust T cell BrdU incorporation. Despite a similar ability to induce lymphocyte proliferation, APCcy and APCox differed for the secretory pathway and mRNA expression. Analysis of the differentially expressed genes identified 4 gene "clusters," showing reciprocal modulation in APCcy vs. APCox, justifying, according to functional genomics analyses, a different putative function of the cells in antigen processing. Together, these data show that treatment with inflammatory cytokines or oxLDL converts human PAC phenotypes and functions into that of APCs with similar lymphocyte-activating ability but distinct maturation degree and paracrine functions.
Collapse
Affiliation(s)
- Maria Cristina Vinci
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Luca Piacentini
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Mattia Chiesa
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Federica Saporiti
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Gualtiero I Colombo
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Maurizio Pesce
- *Unità di Ingegneria Tissutale Cardiovascolare and Unità di Immunologia e Genomica Funzionale, Centro Cardiologico Monzino, Istituto Di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
360
|
McClung JM, Reinardy JL, Mueller SB, McCord TJ, Kontos CD, Brown DA, Hussain SNA, Schmidt CA, Ryan TE, Green TD. Muscle cell derived angiopoietin-1 contributes to both myogenesis and angiogenesis in the ischemic environment. Front Physiol 2015; 6:161. [PMID: 26042050 PMCID: PMC4436568 DOI: 10.3389/fphys.2015.00161] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/04/2015] [Indexed: 12/13/2022] Open
Abstract
Recent strategies to treat peripheral arterial disease (PAD) have focused on stem cell based therapies, which are believed to result in local secretion of vascular growth factors. Little is known, however, about the role of ischemic endogenous cells in this context. We hypothesized that ischemic muscle cells (MC) are capable of secreting growth factors that act as potent effectors of the local cellular regenerative environment. Both muscle and endothelial cells (ECs) were subjected to experimental ischemia, and conditioned medium (CM) from each was collected and analyzed to assess myogenic and/or angiogenic potential. In muscle progenitors, mRNA expression of VEGF and its cognate receptors (Nrp1, Flt, Flk) was present and decreased during myotube formation in vitro, and EC CM or VEGF increased myoblast proliferation. Angiopoietin-1 (Ang-1), Tie1, and Tie2 mRNA increased during MC differentiation in vitro. Exogenous Ang-1 enhanced myogenic (MyoD and Myogenin) mRNA in differentiating myoblasts and increased myosin heavy chain protein. Myotube formation was enhanced by MC CM and inhibited by EC CM. Ang-1 protein was present in CM from MCs isolated from both the genetically ischemia-susceptible BALB/c and ischemia-resistant C57BL/6 mouse strains, and chimeric Tie2 receptor trapping in situ ablated Ang-1's myogenic effects in vitro. Ang-1 or MC CM enhanced myotube formation in a mixed isolate of muscle progenitors as well as a myoblast co-culture with pluripotent mesenchymal cells (10T1/2) and this effect was abrogated by viral expression of the extracellular domain of Tie2 (AdsTie2). Furthermore, mesh/tube formation by HUVECs was enhanced by Ang-1 or MC CM and abrogated by Tie2 chimeric receptor trapping. Our results demonstrate the ability of muscle and endothelial cell-derived vascular growth factors, particularly Ang-1, to serve as multi-functional stimuli regulating crosstalk between blood vessels and muscle cells during regeneration from ischemic myopathy.
Collapse
Affiliation(s)
- Joseph M McClung
- Department of Physiology, Brody School of Medicine at East Carolina University Greenville, NC, USA ; Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC, USA
| | - Jessica L Reinardy
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine Durham, NC, USA
| | - Sarah B Mueller
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine Durham, NC, USA ; Medical Scientist Training Program, Duke University School of Medicine Durham, NC, USA
| | - Timothy J McCord
- Division of Cardiology, Department of Medicine, Duke University Medical Center Durham, NC, USA
| | - Christopher D Kontos
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine Durham, NC, USA ; Medical Scientist Training Program, Duke University School of Medicine Durham, NC, USA ; Division of Cardiology, Department of Medicine, Duke University Medical Center Durham, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine at East Carolina University Greenville, NC, USA ; Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC, USA
| | - Sabah N A Hussain
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Royal Victoria Hospital, McGill University Montreal, QC, Canada ; Department of Critical Care, McGill University Health Centre, Royal Victoria Hospital, McGill University Montreal, QC, Canada
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine at East Carolina University Greenville, NC, USA ; Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC, USA
| | - Terence E Ryan
- Department of Physiology, Brody School of Medicine at East Carolina University Greenville, NC, USA ; Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine at East Carolina University Greenville, NC, USA ; Diabetes and Obesity Institute, Brody School of Medicine at East Carolina University Greenville, NC, USA
| |
Collapse
|
361
|
Felice F, Di Stefano R, Pini S, Mazzotta G, Bovenzi FM, Bertoli D, Abelli M, Borelli L, Cardini A, Lari L, Gesi C, Michi P, Morrone D, Gnudi L, Balbarini A. Influence of depression and anxiety on circulating endothelial progenitor cells in patients with acute coronary syndromes. Hum Psychopharmacol 2015; 30:183-8. [PMID: 25784019 DOI: 10.1002/hup.2470] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 02/01/2015] [Accepted: 02/02/2015] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Circulating endothelial progenitor cells (EPCs) are related to endothelial function and progression of coronary artery disease. There is evidence of decreased numbers of circulating EPCs in patients with a current episode of major depression. We investigated the relationships between the level of circulating EPCs and depression and anxiety in patients with acute coronary syndrome (ACS). METHODS Patients with ACS admitted to three Cardiology Intensive Care Units were evaluated by the SCID-I to determine the presence of lifetime and/or current mood and anxiety disorders according to DSM-IV criteria. The EPCs were defined as CD133(+) CD34(+) KDR(+) and evaluated by flow cytometry. All patients underwent standardized cardiological and psychopathological evaluations. Parametric and nonparametric statistical tests were performed where appropriate. RESULTS Out of 111 ACS patients, 57 were found to have a DSM-IV lifetime or current mood or anxiety disorder at the time of the inclusion in the study. The ACS group with mood or anxiety disorders showed a significant decrease in circulating EPC number compared with ACS patients without affective disorders. In addition, EPC levels correlated negatively with severity of depression and anxiety at index ACS episode. CONCLUSIONS The current study indicates that EPCs circulate in decreased numbers in ACS patients with depression or anxiety and, therefore, contribute to explore new perspectives in the pathophysiology of the association between cardiovascular disorders and affective disorders.
Collapse
Affiliation(s)
- Francesca Felice
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
362
|
Martini G, Biscaro F, Boscaro E, Calabrese F, Lunardi F, Facco M, Agostini C, Zulian F, Fadini GP. Reduced levels of circulating progenitor cells in juvenile idiopathic arthritis are counteracted by anti TNF-α therapy. BMC Musculoskelet Disord 2015; 16:103. [PMID: 25925313 PMCID: PMC4418050 DOI: 10.1186/s12891-015-0555-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial progenitor cells (EPC) promote angiogenesis and vascular repair. Though reduced EPC levels have been shown in rheumatoid arthritis, no study has so far evaluated EPCs in children with juvenile idiopathic arthritis (JIA). We aimed to study circulating EPCs in children with JIA, their relation to disease activity, and effects of anti TNF-α treatment. Methods Circulating EPCs were quantified by flow cytometry based on CD34, CD133 and KDR expression in peripheral blood of 22 patients with oligoarticular JIA and 29 age-matched controls. EPCs were re-assessed in children with methotrexate-resistant oligo-extended JIA before and up to 12 month after initiation of anti-TNF-alpha therapy. Plasma concentrations of inflammatory and EPC-regulating factors were measured using a multiplex array. Confocal immunofluorescence was used to demonstrate EPCs in synovial tissues. Results Children with active JIA showed a significant reduction of relative and absolute counts of circulating progenitor cells and EPCs compared to age-matched healthy controls. CD34+ cell levels were modestly and inversely correlated to disease activity. A strong inverse correlation was found between serum TNF-α and EPC levels. In 8 patients treated with anti TNF-α agents, the number of EPCs rose to values similar to healthy controls. CD34+KDR+ EPCs were found in the synovial tissue of JIA children, but not in control. Conclusions Children with JIA have reduced levels of the vasculoprotective and proangiogenic EPCs. While EPCs may contribute to synovial tissue remodelling, EPC pauperization may indicate an excess cardiovascular risk if projected later in life.
Collapse
Affiliation(s)
- Giorgia Martini
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Francesca Biscaro
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Elisa Boscaro
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Fiorella Calabrese
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesca Lunardi
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Monica Facco
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Carlo Agostini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesco Zulian
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| |
Collapse
|
363
|
Cooke JP, Losordo DW. Modulating the vascular response to limb ischemia: angiogenic and cell therapies. Circ Res 2015; 116:1561-78. [PMID: 25908729 PMCID: PMC4869986 DOI: 10.1161/circresaha.115.303565] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/31/2015] [Indexed: 12/29/2022]
Abstract
The age-adjusted prevalence of peripheral arterial disease in the US population has been estimated to approach 12%. The clinical consequences of occlusive peripheral arterial disease include pain on walking (claudication), pain at rest, and loss of tissue integrity in the distal limbs; the latter may ultimately lead to amputation of a portion of the lower extremity. Surgical bypass techniques and percutaneous catheter-based interventions may successfully reperfuse the limbs of certain patients with peripheral arterial disease. In many patients, however, the anatomic extent and distribution of arterial occlusion is too severe to permit relief of pain and healing of ischemic ulcers. No effective medical therapy is available for the treatment of such patients, for many of whom amputation represents the only hope for alleviation of symptoms. The ultimate failure of medical treatment and procedural revascularization in significant numbers of patients has led to attempts to develop alternative therapies for ischemic disease. These strategies include administration of angiogenic cytokines, either as recombinant protein or as gene therapy, and more recently, to investigations of stem/progenitor cell therapy. The purpose of this review is to provide an outline of the preclinical basis for angiogenic and stem cell therapies, review the clinical research that has been done, summarize the lessons learned, identify gaps in knowledge, and suggest a course toward successfully addressing an unmet medical need in a large and growing patient population.
Collapse
Affiliation(s)
- John P Cooke
- From the Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.); and NeoStem Inc, New York, NY (D.W.L.).
| | - Douglas W Losordo
- From the Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (J.P.C.); and NeoStem Inc, New York, NY (D.W.L.).
| |
Collapse
|
364
|
Lee SH, Lee KB, Lee JH, Kang S, Kim HG, Asahara T, Kwon SM. Selective Interference Targeting of Lnk in Umbilical Cord-Derived Late Endothelial Progenitor Cells Improves Vascular Repair, Following Hind Limb Ischemic Injury, via Regulation of JAK2/STAT3 Signaling. Stem Cells 2015; 33:1490-500. [DOI: 10.1002/stem.1938] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 11/28/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital; Seoul South Korea
- Department of Biochemistry; School of Medicine, Soonchunhyang University; Cheonan South Korea
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
| | - Kyeung Bin Lee
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
| | - Jun Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| | - Songhwa Kang
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| | - Hwi Gon Kim
- Department of Obstetrics and Gynecology; Pusan National University, School of Medicine; Busan South Korea
| | - Takayuki Asahara
- Department of Regenerative Medicine Science; Tokai University School of Medicine; Shimokasuya Isehara Kanagawa Japan
| | - Sang Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| |
Collapse
|
365
|
Dong XH, Sun X, Jiang GJ, Chen AF, Xie HH. Dietary intake of sugar substitutes aggravates cerebral ischemic injury and impairs endothelial progenitor cells in mice. Stroke 2015; 46:1714-8. [PMID: 25908458 DOI: 10.1161/strokeaha.114.007308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/30/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE In our current food supply, sugar substitutes are widely used in beverages and other food products. However, there is limited information about the link between dietary consumption of sugar substitutes and stroke to date. This study sought to determine the effect of various sugar substitutes on the cerebral ischemic injury and endothelial progenitor cells, which have been implicated to play an important role in vascular repair and revascularization in ischemic brain tissues, in mice. METHODS After treatment with sucrose and various sugar substitutes (the doses are in the range of corresponding acceptable daily intake levels) and vehicle for 6 weeks, mice were subjected to permanent left middle cerebral artery occlusion, and the infarct volumes, angiogenesis, and neurobehavioral outcomes were determined. In addition, the number and function of endothelial progenitor cells were also examined. RESULTS After long-term treatment with fructose, erythritol (sugar alcohols), acesulfame K (artificial sweeteners), or rebaudioside A (rare sugars), the cerebral ischemic injury (both infarct volumes and neurobehavioral outcomes) was significantly aggravated, angiogenesis in ischemic brain was reduced, and endothelial progenitor cell function was impaired in mice compared with control. However, the similar impairments were not found in sucrose (with the same dose as fructose's)-treated mice. CONCLUSIONS Long-term consumption of sugar substitutes aggravated cerebral ischemic injury in mice, which might be partly attributed to the impairment of endothelial progenitor cells and the reduction of angiogenesis in ischemic brain. This result implies that dietary intake of sugar substitutes warrants further attention in daily life.
Collapse
Affiliation(s)
- Xiao-Hui Dong
- From the Department of Science for Identifying Chinese Materia Medica, The Second Military Medical University, Shanghai, China (X.-H.D., X.S., A.F.C., H.-H.X.); Department of Cardiology and Center of Clinical Pharmacology, Third Xiangya Hospital and the Institute of Vascular Disease and Translational Medicine, Central South University, Changsha, Hunan, China (A.F.C., H.-H.X.); and Department of Pharmacy, Xiaoshan Hospital, Hangzhou, China (G.-J.J.)
| | - Xin Sun
- From the Department of Science for Identifying Chinese Materia Medica, The Second Military Medical University, Shanghai, China (X.-H.D., X.S., A.F.C., H.-H.X.); Department of Cardiology and Center of Clinical Pharmacology, Third Xiangya Hospital and the Institute of Vascular Disease and Translational Medicine, Central South University, Changsha, Hunan, China (A.F.C., H.-H.X.); and Department of Pharmacy, Xiaoshan Hospital, Hangzhou, China (G.-J.J.)
| | - Guo-Jun Jiang
- From the Department of Science for Identifying Chinese Materia Medica, The Second Military Medical University, Shanghai, China (X.-H.D., X.S., A.F.C., H.-H.X.); Department of Cardiology and Center of Clinical Pharmacology, Third Xiangya Hospital and the Institute of Vascular Disease and Translational Medicine, Central South University, Changsha, Hunan, China (A.F.C., H.-H.X.); and Department of Pharmacy, Xiaoshan Hospital, Hangzhou, China (G.-J.J.)
| | - Alex F Chen
- From the Department of Science for Identifying Chinese Materia Medica, The Second Military Medical University, Shanghai, China (X.-H.D., X.S., A.F.C., H.-H.X.); Department of Cardiology and Center of Clinical Pharmacology, Third Xiangya Hospital and the Institute of Vascular Disease and Translational Medicine, Central South University, Changsha, Hunan, China (A.F.C., H.-H.X.); and Department of Pharmacy, Xiaoshan Hospital, Hangzhou, China (G.-J.J.).
| | - He-Hui Xie
- From the Department of Science for Identifying Chinese Materia Medica, The Second Military Medical University, Shanghai, China (X.-H.D., X.S., A.F.C., H.-H.X.); Department of Cardiology and Center of Clinical Pharmacology, Third Xiangya Hospital and the Institute of Vascular Disease and Translational Medicine, Central South University, Changsha, Hunan, China (A.F.C., H.-H.X.); and Department of Pharmacy, Xiaoshan Hospital, Hangzhou, China (G.-J.J.).
| |
Collapse
|
366
|
Barsotti MC, Al Kayal T, Tedeschi L, Dinucci D, Losi P, Sbrana S, Briganti E, Giorgi R, Chiellini F, Di Stefano R, Soldani G. Oligonucleotide biofunctionalization enhances endothelial progenitor cell adhesion on cobalt/chromium stents. J Biomed Mater Res A 2015; 103:3284-92. [PMID: 25809157 DOI: 10.1002/jbm.a.35461] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/13/2015] [Accepted: 03/20/2015] [Indexed: 11/11/2022]
Abstract
As the endothelium still represents the ideal surface for cardiovascular devices, different endothelialization strategies have been attempted for biocompatibility and nonthrombogenicity enhancement. Since endothelial progenitor cells (EPCs) could accelerate endothelialization, preventing thrombosis and restenosis, the aim of this study was to use oligonucleotides (ONs) to biofunctionalize stents for EPC binding. In order to optimize the functionalization procedure before its application to cobalt-chromium (Co/Cr) stents, discs of the same material were preliminarily used. Surface aminosilanization was assessed by infrared spectroscopy and scanning electron microscopy. A fluorescent endothelial-specific ON was immobilized on aminosilanized surfaces and its presence was visualized by confocal microscopy. Fluorescent ON binding to porcine blood EPCs was assessed by flow cytometry. Viability assay was performed on EPCs cultured on unmodified, nontargeting ON or specific ON-coated discs; fluorescent staining of nuclei and F-actin was then performed on EPCs cultured on unmodified or specific ON-coated discs and stents. Disc biofunctionalization significantly increased EPC viability as compared to both unmodified and nontargeting ON-coated surfaces; cell adhesion was also significantly increased. Stents were successfully functionalized with the specific ON, and EPC binding was confirmed by confocal microscopy. In conclusion, stent biofunctionalization for EPC binding was successfully achieved in vitro, suggesting its use to obtain in vivo endothelialization, exploiting the natural regenerative potential of the human body.
Collapse
Affiliation(s)
| | - Tamer Al Kayal
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| | - Lorena Tedeschi
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| | - Dinuccio Dinucci
- BioLab-UdR-INSTM, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, 56122, Italy
| | - Paola Losi
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| | - Silverio Sbrana
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| | - Enrica Briganti
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| | - Rodorico Giorgi
- Department of Chemistry and CSGI, University of Florence, Sesto Fiorentino (Florence), 50019, Italy
| | - Federica Chiellini
- BioLab-UdR-INSTM, Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, 56122, Italy
| | - Rossella Di Stefano
- Cardiovascular Research Laboratory, Department of Surgery, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, 56124, Italy
| | - Giorgio Soldani
- Institute of Clinical Physiology, National Research Council, Massa, 54100, Italy
| |
Collapse
|
367
|
Of mice and men: the best laid scheme? J Am Coll Cardiol 2015; 65:1435-7. [PMID: 25857909 DOI: 10.1016/j.jacc.2015.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 02/22/2015] [Indexed: 11/21/2022]
|
368
|
Bonello L, Frere C, Cointe S, Laine M, Mancini J, Thuny F, Kerbaul F, Lemesle G, Paganelli F, Guieu R, Arnaud L, Dignat-George F, Sabatier F. Ticagrelor increases endothelial progenitor cell level compared to clopidogrel in acute coronary syndromes: A prospective randomized study. Int J Cardiol 2015; 187:502-7. [PMID: 25846661 DOI: 10.1016/j.ijcard.2015.03.414] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/27/2015] [Accepted: 03/28/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND The clinical benefit of ticagrelor compared to clopidogrel in ACS patients suggested off-target property. Such pleiotropic effect could be mediated by circulating endothelial progenitor cells (EPC) which are critical for vascular healing. We aimed to investigate the impact of ticagrelor on EPC in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI). METHODS We prospectively randomized 106 ACS patients to ticagrelor or clopidogrel. Sub-populations of CD34+ circulating progenitor cells (PC) were analyzed by flow cytometry allowing one to determine the levels of CD34+ PC, CD34+CD45+ Hematopoietic PC, CD34+133+ immature PC and CD34+KDR+ EPC on admission and at 1 month. Changes in PC level were calculated as the difference between 1 month and baseline value. RESULTS The 2 groups were similar regarding baseline characteristics including PC numbers on admission. The 2 groups had similar change in overall CD34+ PC and hematopoietic CD34+45+ PC level (p=0.2). On the contrary, when considering CD34+133+ PC and CD34+KDR+ EPC, we observed that patients treated by ticagrelor had a significantly higher increase in levels of these PC subtypes compared to those treated by clopidogrel (0.23 (-0.33; 0.79) vs 0.00 (-0.5; 0.34); p=0.04 and 0.01 (-0.04; 0.05) vs -0.01 (-0.06; 0.03); p=0.02). Changes in the level of CD34+CD133+ PC correlated with platelet activity measured by the VASP index (r=-0.30; p=0.008). By contrast the increase in the level of CD34+KDR+ EPC in the ticagrelor group was independent of platelet activity. CONCLUSIONS Ticagrelor increases the number of EPC in ACS patients suggesting a benefit on endothelial regeneration that may participate in the pleiotropic property of the drug.
Collapse
Affiliation(s)
- Laurent Bonello
- Service de Cardiologie, Centre Hospitalier Universitaire de Marseille, Hôpital NORD, Aix-Marseille Université, France; Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France.
| | - Corinne Frere
- Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France
| | - Sylvie Cointe
- Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France
| | - Marc Laine
- Service de Cardiologie, Centre Hospitalier Universitaire de Marseille, Hôpital NORD, Aix-Marseille Université, France
| | - Julien Mancini
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Timone, BiosTIC, France; Aix-Marseille Université, Inserm, IRD, UMR_S912, SESSTIM, Faculté de Médecine de Marseille, France
| | - Franck Thuny
- Service de Cardiologie, Centre Hospitalier Universitaire de Marseille, Hôpital NORD, Aix-Marseille Université, France
| | - François Kerbaul
- Research Unit of Physiology and Pathophysiology in Extreme Oxygenation Conditions (UMR MD2), Faculty of Medicine, Aix-Marseille University, Marseille, France; Pole RUSH, Assistance-Publique Hôpitaux de Marseille, Marseille, France
| | | | - Franck Paganelli
- Service de Cardiologie, Centre Hospitalier Universitaire de Marseille, Hôpital NORD, Aix-Marseille Université, France
| | - Regis Guieu
- Research Unit of Physiology and Pathophysiology in Extreme Oxygenation Conditions (UMR MD2), Faculty of Medicine, Aix-Marseille University, Marseille, France
| | - Laurent Arnaud
- Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France; Laboratoire d'Hématologie et de Biologie Vasculaire, Assistance-Publique Hôpitaux de Marseille, Centre hospitalo-universitaire de la Conception, Marseille, France
| | - Françoise Dignat-George
- Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France; Laboratoire d'Hématologie et de Biologie Vasculaire, Assistance-Publique Hôpitaux de Marseille, Centre hospitalo-universitaire de la Conception, Marseille, France
| | - Florence Sabatier
- Vascular Research Center of Marseille, INSERM UMR-S 1076, Aix-Marseille Université, Marseille, France; Laboratoire d'Hématologie et de Biologie Vasculaire, Assistance-Publique Hôpitaux de Marseille, Centre hospitalo-universitaire de la Conception, Marseille, France
| |
Collapse
|
369
|
Spigoni V, Lombardi C, Cito M, Picconi A, Ridolfi V, Andreoli R, Anelli N, Gnudi L, Goldoni M, Zavaroni I, Raddino R, Dei Cas A. N-3 PUFA increase bioavailability and function of endothelial progenitor cells. Food Funct 2015; 5:1881-90. [PMID: 24942553 DOI: 10.1039/c3fo60641d] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Recent data suggest that n-3 PUFA exert beneficial effects on endothelial progenitor cell (EPC) biology. We sought to investigate whether these effects might be mediated by enhanced EPC in vitro function and/or in vivo bioavailability. METHODS AND RESULTS CACs and late-outgrowth EPCs were isolated from peripheral blood mononuclear cells obtained from 12 donor buffy-coats. The effect of n-3 PUFA (EPA:DHA = 0.9:1.5; 9 μM EPA plus 15 μM DHA) was tested on CAC/EPC viability, function (tube-formation) and pro-inflammatory molecule expression. Circulating EPC (cells positive for CD34, CD133 and kinase insert domain receptor - KDR cell-surface antigens by flow cytometry) number was evaluated in 20 healthy subjects (10 F/10 M, 32 ± 5 years), randomized to receive 4 mackerel or sardine portions per week for 6 weeks followed by a 6 week free-diet period. N-3 PUFA improved CAC and late-outgrowth EPC viability (p < 0.05) and the capacity to form tube-like structures in CACs (+38%; p < 0.05) and late-outgrowth EPCs (+15%; p < 0.05). ICAM-1 expression was reduced in both CACs (p < 0.05) and late-outgrowth EPCs (p < 0.05) and VCAM-1 in late-outgrowth EPCs (p < 0.005). N-3 PUFA significantly decreased TNF-α and MCP-1 expression in CACs and IL-8, TNF-α and MCP-1 in late-outgrowth EPCs (p < 0.05). Circulating EPC number significantly improved after 6 weeks of a fish-enriched diet (p < 0.01) and returned to baseline levels after a 6 week free-diet period (p < 0.01). Plasma EPA levels were independently and positively associated with EPC levels (p < 0.005). CONCLUSION Our findings support the case of a beneficiary role played by n-3 PUFA in EPC function and bioavailability.
Collapse
Affiliation(s)
- Valentina Spigoni
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health. University of Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
370
|
Atkins GB, Orasanu G, Jain MK. Endothelial Cells. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
371
|
Spigoni V, Cito M, Alinovi R, Pinelli S, Passeri G, Zavaroni I, Goldoni M, Campanini M, Aliatis I, Mutti A, Bonadonna RC, Dei Cas A. Effects of TiO₂ and Co₃O₄ nanoparticles on circulating angiogenic cells. PLoS One 2015; 10:e0119310. [PMID: 25803285 PMCID: PMC4372399 DOI: 10.1371/journal.pone.0119310] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/12/2015] [Indexed: 12/03/2022] Open
Abstract
Background and Aim Sparse evidence suggests a possible link between exposure to airborne nanoparticles (NPs) and cardiovascular (CV) risk, perhaps through mechanisms involving oxidative stress and inflammation. We assessed the effects of TiO2 and Co3O4 NPs in human circulating angiogenic cells (CACs), which take part in vascular endothelium repair/replacement. Methods CACs were isolated from healthy donors’ buffy coats after culturing lymphomonocytes on fibronectin-coated dishes in endothelial medium for 7 days. CACs were pre-incubated with increasing concentration of TiO2 and Co3O4 (from 1 to 100 μg/ml) to test the effects of NP – characterized by Transmission Electron Microscopy – on CAC viability, apoptosis (caspase 3/7 activation), function (fibronectin adhesion assay), oxidative stress and inflammatory cytokine gene expression. Results Neither oxidative stress nor cell death were associated with exposure to TiO2 NP (except at the highest concentration tested), which, however, induced a higher pro-inflammatory effect compared to Co3O4 NPs (p<0.01). Exposure to Co3O4 NPs significantly reduced cell viability (p<0.01) and increased caspase activity (p<0.01), lipid peroxidation end-products (p<0.05) and pro-inflammatory cytokine gene expression (p<0.05 or lower). Notably, CAC functional activity was impaired after exposure to both TiO2 (p<0.05 or lower) and Co3O4 (p<0.01) NPs. Conclusions In vitro exposure to TiO2 and Co3O4 NPs exerts detrimental effects on CAC viability and function, possibly mediated by accelerated apoptosis, increased oxidant stress (Co3O4 NPs only) and enhancement of inflammatory pathways (both TiO2 and Co3O4 NPs). Such adverse effects may be relevant for a potential role of exposure to TiO2 and Co3O4 NPs in enhancing CV risk in humans.
Collapse
Affiliation(s)
- Valentina Spigoni
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Monia Cito
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Rossella Alinovi
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Silvana Pinelli
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Giovanni Passeri
- Department of Clinical and Experimental Medicine. Unit of Andrology, Metabolic Bone Diseases and Endocrinology. University of Parma, Parma, Italy
| | - Ivana Zavaroni
- Department of Clinical and Experimental Medicine. Unit of Diabetes and prevention of associated diseases. University of Parma, Parma, Italy
| | - Matteo Goldoni
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Marco Campanini
- IMEM-CNR (Istituto Materiale per l’Elettronica ed il Magnetismo – Consiglio Nazionale delle Ricerche) Institute, Parma, Italy
| | - Irene Aliatis
- Department of Physics and Earth Sciences. University of Parma, Parma, Italy
| | - Antonio Mutti
- Department of Clinical and Experimental Medicine. Unit of Occupational and Environmental Medicine. University of Parma, Parma, Italy
| | - Riccardo C. Bonadonna
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
| | - Alessandra Dei Cas
- Department of Clinical and Experimental Medicine. Division of Endocrinology. University of Parma and Azienda Ospedaliero-Universitaria of Parma, Parma, Italy
- * E-mail:
| |
Collapse
|
372
|
Steinmetz M, Lucanus E, Zimmer S, Nickenig G, Werner N. Mobilization of sca1/flk-1 positive endothelial progenitor cells declines in apolipoprotein E-deficient mice with a high-fat diet. J Cardiol 2015; 66:532-8. [PMID: 25818640 DOI: 10.1016/j.jjcc.2015.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/13/2015] [Accepted: 02/05/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Atherosclerosis features a deterioration of the endothelial layer in all stages. Restoration of the endothelium is associated with circulating stem cell antigen 1 (sca1) and vascular endothelial growth factor receptor type 2 (flk-1) positive endothelial progenitor cells (EPCs). We investigated whether EPC production and/or a mobilization from bone marrow are reduced in severe atherosclerosis. METHODS AND RESULTS EPCs in peripheral blood were diminished in ApoE-/- mice with high-fat diet (HFD) whereas bone marrow levels of these cells were not significantly altered compared to controls. In situ perfusion of the hind limbs demonstrated that EPC mobilization was reduced compared to ApoE-/- mice with normal chow, although increased plasma stromal cell-derived factor (SDF) 1α and responsivity suggested a mobilizing stimulus. The proliferation of sca1/flk-1 positive cells showed no functional impairment. EPCs could not only be significantly mobilized from the bone marrow through the application of granulocyte colony stimulating factor (GCSF), but also led by trend to a depletion of the bone marrow pool. GCSF levels in plasma were equal in ApoE-/- mice with normal chow or HFD, which excluded a decline in GCSF production. CONCLUSION The capability of the bone marrow pool to adapt the proliferation and mobilization of sca1/flk-1 positive EPCs seems overstrained in ApoE-/- mice with a HFD.
Collapse
Affiliation(s)
- Martin Steinmetz
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany; Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.
| | - Eva Lucanus
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Nikos Werner
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
373
|
Schmidt-Lucke C, Zobel T, Schrepfer S, Kuhl U, Wang D, Klingel K, Becher PM, Fechner H, Pozzuto T, Van Linthout S, Lassner D, Spillmann F, Escher F, Holinski S, Volk HD, Schultheiss HP, Tschope C. Impaired Endothelial Regeneration Through Human Parvovirus B19-Infected Circulating Angiogenic Cells in Patients With Cardiomyopathy. J Infect Dis 2015; 212:1070-81. [PMID: 25805750 DOI: 10.1093/infdis/jiv178] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/26/2015] [Indexed: 11/13/2022] Open
Abstract
Human parvovirus B19 (B19V) is a common pathogen in microvascular disease and cardiomyopathy, owing to infection of endothelial cells. B19V replication, however, is almost restricted to erythroid progenitor cells (ErPCs). Endothelial regeneration attributable to bone marrow-derived circulating angiogenic cells (CACs) is a prerequisite for organ function. Because of many similarities of ErPCs and CACs, we hypothesized that B19V is a perpetrator of impaired endogenous endothelial regeneration. B19V DNA and messenger RNA from endomyocardial biopsy specimens, bone marrow specimens, and circulating progenitor cells were quantified by polymerase chain reaction analysis. The highest B19V DNA concentrations were found in CD34(+)KDR(+) cells from 17 patients with chronic B19V-associated cardiomyopathy. B19V replication intermediates could be detected in nearly half of the patients. Furthermore, chronic B19V infection was associated with impaired endothelial regenerative capacity. B19V infection of CACs in vitro resulted in expression of transcripts encoding B19V proteins. The capsid protein VP1 was identified as a novel inducer of apoptosis, as were nonstructural proteins. Inhibition studies identified so-called death receptor signaling with activation of caspase-8 and caspase-10 to be responsible for apoptosis induction. B19V causally impaired endothelial regeneration with spreading of B19V in CACs in an animal model in vivo. We thus conclude that B19V infection and damage to CACs result in dysfunctional endogenous vascular repair, supporting the emergence of primary bone marrow disease with secondary end-organ damage.
Collapse
Affiliation(s)
- Caroline Schmidt-Lucke
- Department of Cardiology and Pneumology Berlin-Brandenburg Center for Regenerative Therapies Medico-Academic Consultings
| | | | - Sonja Schrepfer
- Transplant and Stem Cell Immunobiology Laboratory, University Heart Center Hamburg
| | - Uwe Kuhl
- Department of Cardiology and Pneumology
| | - Dong Wang
- Transplant and Stem Cell Immunobiology Laboratory, University Heart Center Hamburg
| | - Karin Klingel
- Department of Molecular Pathology, Institute of Pathology, Tübingen, Germany
| | | | - Henry Fechner
- Department of Cardiology and Pneumology Institute for Biotechnology, University of Technology
| | | | | | | | | | | | | | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin-Brandenburg Center for Regenerative Therapies
| | | | - Carsten Tschope
- Department of Cardiology and Pneumology Berlin-Brandenburg Center for Regenerative Therapies Deutsches Zentrum für Herz-Kreislauf-Forschung, Berlin
| |
Collapse
|
374
|
Zhang Y, Kuraitis D, Burgon PG, Crowe S, Vulesevic B, Beanlands RS, deKemp RA, DaSilva JN, Ruel M, Suuronen EJ. Development of reporter gene imaging techniques for long-term assessment of human circulating angiogenic cells. ACTA ACUST UNITED AC 2015; 10:034104. [PMID: 25782444 DOI: 10.1088/1748-6041/10/3/034104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The use of biomaterials and tracking the long-term fate of the transplanted cells is expected to help improve the clinical translation of cell therapies for cardiac regeneration. To this end, reporter gene strategies are promising for monitoring the fate of cells transplanted with or without a delivery biomaterial; however, their application with primary adult progenitor cells (such as human circulating angiogenic cells (CACs)) has not been extensively evaluated. In this study, human CACs were transduced with reporter genes via one of two lentiviral (LV) vectors: LV-GFP-iresTK or LV-Fluc-RFP-tTK. The mean transduction efficiency was 15% (LV-GFP-iresTK) and 13% (LV-Fluc-RFP-tTK) at multiplicities of infection (MOI) of 10 and 50, respectively. Western blot analysis confirmed HSV1-tk protein expression in transduced CACs. There was no significant difference in viability between the transduced CACs and the untreated controls at a MOI of 50 or below. However, a reduction was observed in cell viability of CACs transduced with LV-Fluc-RFP-tTK at an MOI of 100. Cell migration and angiogenic potential were not affected by transduction protocol. After 4 weeks, 80.3 ± 8.4% of the labeled cells continued to express the reporters and could be visualized when embedded within a collagen matrix scaffold. Therefore, quiescent human CACs can be stably transduced to express reporter genes without affecting their function. This reporter gene technique is a promising approach to be further tested for tracking transplanted CACs (±delivery matrix) non-invasively and longitudinally.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, K1Y 4W7, Canada. Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada. Division of Cardiology (Department of Medicine), Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
375
|
Sukmawati D, Tanaka R. Introduction to next generation of endothelial progenitor cell therapy: a promise in vascular medicine. Am J Transl Res 2015; 7:411-421. [PMID: 26045883 PMCID: PMC4448183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/08/2015] [Indexed: 06/04/2023]
Abstract
The concept of Endothelial Progenitor Cells (EPCs) therapy for adult neovascularization has continuously received attention. They are believed to participate in endothelial repair and post natal angiogenesis due to their abilities in differentiating into endothelial cells and producing protective cytokines and growth factors. Abundant evidence supports the involvement of EPCs in capillary growth and in participating in the formation of collateral vessels, which lead to improved vascular perfusion and functional recovery in target tissue. Autologous EPC now is becoming a novel treatment option for therapeutic revascularization and vascular repair in ischemic diseases. However, various diseases such as diabetes, heart disease and ischemic diseases are related to EPC dysfunction and give rise to additional challenges of autologous EPC therapy. A novel strategy to enhance the number and function of EPCs is needed to be established to provide successful autologous EPCs therapy. Currently, clinical trials for the new generation of EPC therapy in treating peripheral ischemic diseases are underway. In this review we provide an overview and the limitations of current EPCs therapy with an introduction to the new strategies of next generation EPC therapy for more promising vascular and tissue regeneration therapy.
Collapse
Affiliation(s)
- Dewi Sukmawati
- Department of Plastic and Reconstructive Surgery, Juntendo University School of MedicineTokyo, Japan
- Department of Cardiovascular Medicine, Juntendo University School of MedicineTokyo, Japan
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of MedicineTokyo, Japan
| |
Collapse
|
376
|
González-Pecchi V, Valdés S, Pons V, Honorato P, Martinez LO, Lamperti L, Aguayo C, Radojkovic C. Apolipoprotein A-I enhances proliferation of human endothelial progenitor cells and promotes angiogenesis through the cell surface ATP synthase. Microvasc Res 2015; 98:9-15. [PMID: 25445031 DOI: 10.1016/j.mvr.2014.11.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/28/2014] [Accepted: 11/02/2014] [Indexed: 12/14/2022]
|
377
|
Zhou L, Xia J, Qiu X, Wang P, Jia R, Chen Y, Yang B, Dai Y. In vitro evaluation of endothelial progenitor cells from adipose tissue as potential angiogenic cell sources for bladder angiogenesis. PLoS One 2015; 10:e0117644. [PMID: 25706311 PMCID: PMC4338275 DOI: 10.1371/journal.pone.0117644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/29/2014] [Indexed: 01/22/2023] Open
Abstract
Autologous endothelial progenitor cells (EPCs) might be alternative angiogenic cell sources for vascularization of tissue-engineered bladder, while isolation and culture of EPCs from peripheral blood in adult are usually time-consuming and highly inefficient. Recent evidence has shown that EPCs also exist in the adipose tissue. As adipose tissue is plentiful in the human body and can be easily harvested through a minimally invasive method, the aim of this study was to culture and characterize EPCs from adipose tissue (ADEPCs) and investigate their potential for the neovascularization of tissue-engineered bladder. Adipose stromal vascular fraction (SVF) was isolated and used for the culture of ADEPCs and adipose derived stem cells (ADSCs). After SVF was cultured for one week, ADEPCs with typical cobblestone morphology emerged and could be isolated from ADSCs according to their different responses to trypsinization. Rat bladder smooth muscle cells (RBSMCs) were isolated and cultured from rat bladder. RBSMCs exhibited typical spindle-shaped morphology. ADEPCs had higher proliferative potential than ADSCs and RBSMCs. ADEPCs stained positive for CD34, Stro-1, VEGFR-2, eNOS and CD31 but negative for α-SMA, CD14 and CD45. ADSCs stained positive for CD34, Stro-1 and α-SMA but negative for VEGFR-2, eNOS, CD31, CD14 and CD45. RBSMCs stained only positive for α-SMA. ADEPCs could be expanded from a single cell at an early passage to a cell cluster containing more than 10,000 cells. ADEPCs were able to uptake DiI-Ac-LDL, bind UEA-1 and form capillary-like structures in three-dimensional scaffolds (Matrigel and bladder acellular matrix). ADEPCs were also able to enhance the human umbilical vein endothelial cells’ capability of capillary-like tube formation on Matrigel. Additionally, significantly higher levels of mRNA and protein of vascular endothelial growth factor were found in ADEPCs than in RBSMCs. These results suggest the potential use of ADEPCs as angiogenic cell sources for engineering bladder tissue.
Collapse
Affiliation(s)
- Liuhua Zhou
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiadong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Qiu
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Pengji Wang
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Chen
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Bin Yang
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
- Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- * E-mail: (BY); (YTD)
| | - Yutian Dai
- Department of Urology and Andrology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
- * E-mail: (BY); (YTD)
| |
Collapse
|
378
|
van Ierssel SH, Conraads VM, Van Craenenbroeck EM, Liu Y, Maas AIR, Parizel PM, Hoymans VY, Vrints CJ, Jorens PG. Endothelial dysfunction in acute brain injury and the development of cerebral ischemia. J Neurosci Res 2015; 93:866-72. [PMID: 25677574 DOI: 10.1002/jnr.23566] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 11/22/2014] [Accepted: 01/05/2015] [Indexed: 01/13/2023]
Abstract
Cerebral ischemia (CeI) is a major complicating event after acute brain injury (ABI) in which endothelial dysfunction is a key player. This study evaluates cellular markers of endothelial function and in vivo reactive hyperemia in patients with ABI and their relationship to the development of cerebral ischemia. We studied cellular markers of endothelial dysfunction and the peripheral reactive hyperemia index (RHI) in 26 patients with ABI at admission and after 6 and 12 days, and compared these with those of healthy volunteers (n = 15). CeI was determined clinically or by computer tomography. In patients with ABI, RHI at admission was significantly reduced compared with healthy subjects (P = 0.003), coinciding with a decrease in circulating endothelial progenitor cells (EPC; P = 0.002). The RHI recovered in eight patients without development of CeI, but failed to fully recover by day 12 in three of four patients who developed CeI. Despite recovery of the RHI within 12 days in these patients (P = 0.003), EPC count remained significantly lower after 12 days in patients with ABI (P = 0.022). CD31(+) T cells and endothelial microparticles were not different between controls and patients. No differences were noted in cellular markers of endothelial dysfunction in patients developing CeI and those not. In conclusion, patients with ABI exhibit impaired microvascular endothelial function measured as RHI and a decreased circulating level of EPC.
Collapse
Affiliation(s)
- Sabrina H van Ierssel
- Department of Critical Care Medicine, Antwerp University Hospital, University of Antwerp, Edegem, Belgium; Department of Internal Medicine, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
379
|
Desposito D, Potier L, Chollet C, Gobeil F, Roussel R, Alhenc-Gelas F, Bouby N, Waeckel L. Kinin receptor agonism restores hindlimb postischemic neovascularization capacity in diabetic mice. J Pharmacol Exp Ther 2015; 352:218-26. [PMID: 25398240 DOI: 10.1124/jpet.114.219196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Limb ischemia is a major complication of thromboembolic diseases. Diabetes worsens prognosis by impairing neovascularization. Genetic or pharmacological inactivation of the kallikrein-kinin system aggravates limb ischemia in nondiabetic animals, whereas angiotensin I-converting enzyme/kininase II inhibition improves outcome. The role of kinins in limb ischemia in the setting of diabetes is not documented. We assessed whether selective activation of kinin receptors by pharmacological agonists can influence neovascularization in diabetic mice with limb ischemia and have a therapeutic effect. Selective pseudopeptide kinin B1 or B2 receptor agonists resistant to peptidase action were administered by osmotic minipumps at a nonhypotensive dosage for 14 days after unilateral femoral artery ligation in mice previously rendered diabetic by streptozotocin. Comparison was made with ligatured, nonagonist-treated nondiabetic and diabetic mice. Diabetes reduced neovascularization, assessed by microangiography and histologic capillary density analysis, by roughly 40%. B1 receptor agonist or B2 receptor agonist similarly restored neovascularization in diabetic mice. Neovascularization in agonist-treated diabetic mice was indistinguishable from nondiabetic mice. Both treatments restored blood flow in the ischemic hindfoot, measured by laser-Doppler perfusion imaging. Macrophage infiltration increased 3-fold in the ischemic gastrocnemius muscle during B1 receptor agonist or B2 receptor agonist treatment, and vascular endothelial growth factor (VEGF) level increased 2-fold. Both treatments increased, by 50-100%, circulating CD45/CD11b-positive monocytes and CD34(+)/VEGFR2(+) progenitor cells. Thus, selective pharmacological activation of B1 or B2 kinin receptor overcomes the effect of diabetes on postischemic neovascularization and restores tissue perfusion through monocyte/macrophage mobilization. Kinin receptors are potential therapeutic targets in limb ischemia in diabetes.
Collapse
Affiliation(s)
- Dorinne Desposito
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Louis Potier
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Catherine Chollet
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Fernand Gobeil
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ronan Roussel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Francois Alhenc-Gelas
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Nadine Bouby
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| | - Ludovic Waeckel
- Institut National de la Sante et de la Recherche Medicale U1138, Université Paris Descartes, and Université Pierre et Marie Curie, Paris, France (D.D., L.P., C.C., R.R., F.A.-G., N.B., L.W.); Université Paris Diderot, and Diabétologie-Endocrinologie-Nutrition, DHU FIRE, Hôpital Bichat, AP-HP, Paris, France (L.P., R.R.); and Department of Pharmacology, University of Sherbrooke, Sherbrooke, Quebec, Canada (F.G.)
| |
Collapse
|
380
|
Anderson EM, Kwee BJ, Lewin SA, Raimondo T, Mehta M, Mooney DJ. Local delivery of VEGF and SDF enhances endothelial progenitor cell recruitment and resultant recovery from ischemia. Tissue Eng Part A 2015; 21:1217-27. [PMID: 25434326 DOI: 10.1089/ten.tea.2014.0508] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Biomaterials may improve outcomes of endothelial progenitor-based therapies for the treatment of ischemic cardiovascular disease, due to their ability to direct cell behavior. We hypothesized that local, sustained delivery of exogenous vascular endothelial growth factor (VEGF) and stromal cell-derived factor (SDF) from alginate hydrogels could increase recruitment of systemically infused endothelial progenitors to ischemic tissue, and subsequent neovascularization. VEGF and SDF were found to enhance in vitro adhesion and migration of outgrowth endothelial cells (OECs) and circulating angiogenic cells (CACs), two populations of endothelial progenitors, by twofold to sixfold, and nearly doubled recruitment to both ischemic and nonischemic muscle tissue in vivo. Local delivery of VEGF and SDF to ischemic hind-limbs in combination with systemic CAC delivery significantly improved functional perfusion recovery over OEC delivery, or either treatment alone. Compared with OECs, CACs were more responsive to VEGF and SDF treatment, promoted in vitro endothelial sprout formation in a paracrine manner more potently, and demonstrated greater influence on infiltrating inflammatory cells in vivo. These studies demonstrate that accumulation of infused endothelial progenitors can be enriched using biomaterial-based delivery of VEGF and SDF, and emphasize the therapeutic benefit of using CACs for the treatment of ischemia.
Collapse
Affiliation(s)
- Erin M Anderson
- 1 School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts
| | | | | | | | | | | |
Collapse
|
381
|
Guerin CL, Loyer X, Vilar J, Cras A, Mirault T, Gaussem P, Silvestre JS, Smadja DM. Bone-marrow-derived very small embryonic-like stem cells in patients with critical leg ischaemia: evidence of vasculogenic potential. Thromb Haemost 2015; 113:1084-94. [PMID: 25608764 DOI: 10.1160/th14-09-0748] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/07/2014] [Indexed: 12/21/2022]
Abstract
Very small embryonic-like stem cells (VSELs) are multipotent stem cells localised in adult bone marrow (BM) that may be mobilised into peripheral blood (PB) in response to tissue injury. We aimed to quantify VSELs in BM and PB of patients with critical limb ischaemia (CLI) and to test their angiogenic potential in vitro as well as their therapeutic capacity in mouse model of CLI. We isolated BM VSELs from patients with CLI and studied their potential to differentiate into vascular lineages. Flow and imaging cytometry showed that VSEL counts were lower in BM (p< 0.001) and higher (p< 0.001) in PB from CLI patients compared to healthy controls, suggesting that ischaemia may trigger VSELs mobilisation in this patient population. Sorted BM-VSELs cultured in angiogenic media acquired a mesenchymal phenotype (CD90+, Thy-1 gene positive expression). VSEL-derived cells had a pattern of secretion similar to that of endothelial progenitor cells, as they released low levels of VEGF-A and inflammatory cytokines. Noteworthy, VSELs triggered post-ischaemic revascularisation in immunodeficient mice (p< 0.05 vs PBS treatment), and acquired an endothelial phenotype either in vitro when cultured in the presence of VEGF-B (Cdh-5 gene positive expression), or in vivo in Matrigel implants (human CD31+ staining in neo-vessels from plug sections). In conclusion, VSELs are a potential new source of therapeutic cells that may give rise to cells of the endothelial lineage in humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David M Smadja
- Prof. David Smadja, European Georges Pompidou Hospital, Hematology Department, 20 rue Leblanc, 75015 Paris, France, Tel.: +31 56093933, Fax: +31 56093393, E-mail:
| |
Collapse
|
382
|
Abstract
In ageing populations many patients have multiple diseases characterised by acceleration of the normal ageing process. Better understanding of the signalling pathways and cellular events involved in ageing shows that these are characteristic of many chronic degenerative diseases, such as chronic obstructive pulmonary disease (COPD), chronic cardiovascular and metabolic diseases, and neurodegeneration. Common mechanisms have now been identified in these diseases, which show evidence of cellular senescence with telomere shortening, activation of PI3K–AKT–mTOR signalling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence and low grade chronic inflammation (“inflammaging”). Many of these pathways are driven by chronic oxidative stress. There is also a reduction in anti-ageing molecules, such as sirtuins and Klotho, which further accelerates the ageing process. Understanding these molecular mechanisms has identified several novel therapeutic targets and several drugs have already been developed that may slow the ageing process, as well as lifestyle interventions, such as diet and physical activity. This indicates that in the future new treatment approaches may target the common pathways involved in multimorbidity and this area of research should be given high priority. Thus, COPD should be considered as a component of multimorbidity and common disease pathways, particularly accelerated ageing, should be targeted.
Collapse
|
383
|
Affiliation(s)
- Gian Paolo Fadini
- From the Department of Medicine, University of Padova, Padova, Italy; and Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
384
|
Fraineau S, Palii CG, Allan DS, Brand M. Epigenetic regulation of endothelial-cell-mediated vascular repair. FEBS J 2015; 282:1605-29. [PMID: 25546332 DOI: 10.1111/febs.13183] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/16/2023]
Abstract
Maintenance of vascular integrity is essential for the prevention of vascular disease and for recovery following cardiovascular, cerebrovascular and peripheral vascular events including limb ischemia, heart attack and stroke. Endothelial stem/progenitor cells have recently gained considerable interest due to their potential use in stem cell therapies to mediate revascularization after ischemic injury. Therefore, there is an urgent need to understand fundamental mechanisms regulating vascular repair in specific cell types to develop new beneficial therapeutic interventions. In this review, we highlight recent studies demonstrating that epigenetic mechanisms (including post-translational modifications of DNA and histones as well as non-coding RNA-mediated processes) play essential roles in the regulation of endothelial stem/progenitor cell functions through modifying chromatin structure. Furthermore, we discuss the potential of using small molecules that modulate the activities of epigenetic enzymes to enhance the vascular repair function of endothelial cells and offer insight on potential strategies that may accelerate clinical applications.
Collapse
Affiliation(s)
- Sylvain Fraineau
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Canada; Ottawa Institute of Systems Biology, Canada
| | | | | | | |
Collapse
|
385
|
Ferreras C, Cole CL, Urban K, Jayson GC, Avizienyte E. Segregation of late outgrowth endothelial cells into functional endothelial CD34- and progenitor-like CD34+ cell populations. Angiogenesis 2015; 18:47-68. [PMID: 25269667 DOI: 10.1007/s10456-014-9446-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 09/19/2014] [Indexed: 01/16/2023]
Abstract
Late outgrowth endothelial cells (OECs) that originate from peripheral blood mononuclear cells ex vivo have phenotypic and functional properties of mature endothelial cells. Given the potential therapeutic applications of OECs, understanding their biology is crucial. We have identified two distinct OEC populations based on differential expression of the cell surface marker CD34. OEC colonies lacked CD34 expression (CD34-), expressed CD34 in the majority of cells (CD34+), or showed a mixed expression pattern within a colony (CD34+/-). CD34+ and CD34- OECs were negative for hematopoietic cell marker CD45 and expressed the endothelial cell surface markers CD31, CD146, CD105, and VEGFR-2. Functionally CD34- and CD34+ OECs exhibited strikingly distinct behaviors. CD34- OECs, unlike CD34+ OECs, were capable of sprouting, formed tubes, and responded to angiogenic growth factors in vitro. In vivo, CD34- OECs formed endothelial tubes, while CD34+ OECs, despite being unable to form tubes, promoted infiltration of murine vasculature. Global gene expression profiling in CD34- and CD34+ OECs identified functional importance of the MMP-1/PAR-1 pathway in CD34- OECs. MMP-1 stimulated the expression of VEGFR-2, neuropilin-1, neuropilin-2, and CXCR4 and activated ERK1/2, whereas down-regulation of PAR-1 in CD34- OECs resulted in impaired angiogenic responses in vitro and reduced VEGFR-2 levels. In contrast, the CD34+ OEC colonies expressed high levels of the progenitor cell marker ALDH, which was absent in CD34- OECs. In summary, we show that OECs can be classified into functionally mature endothelial cells (CD34- OECs) that depend on the MMP-1/PAR-1 pathway and progenitor-like angiogenesis-promoting cells (CD34+ OECs).
Collapse
Affiliation(s)
- Cristina Ferreras
- Institute of Cancer Sciences, Faculty of Medical and Human Sciences, The University of Manchester, Paterson Building, Wilmslow Road, Manchester, M20 4BX, UK
| | | | | | | | | |
Collapse
|
386
|
Nathan AA, Tej MAC, Chitiprolu M, Rangan S, Mohan V, Harish R, Anand SB, Dixit M. Impaired glucose tolerance alters functional ability of peripheral blood-derived mononuclear cells in Asian Indian men. Diab Vasc Dis Res 2015; 12:13-22. [PMID: 25303939 DOI: 10.1177/1479164114551745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
AIM To compare the adhesion, migration and endothelial differentiation potential of peripheral blood-derived mononuclear cells (PBMCs) obtained from drug-naive normal glucose tolerance (NGT) and impaired glucose tolerance (IGT) Asian Indian men. METHODS Based on the 75-g oral glucose tolerance test, 30 NGT and 31 IGT subjects were recruited into the study. PBMCs were isolated from fasting blood using histopaque density gradient centrifugation. Isolated PBMCs were analysed for their ability to adhere to extracellular matrices, incorporation into tubular structures formed by matured endothelial cells and differentiation into endothelial cells upon 7-day culture in endothelial-specific growth medium. RESULTS PBMCs obtained from IGT subjects exhibit poor adherence to fibronectin and reduced incorporation into tubular structures. Migration towards stromal cell-derived factor-1α (SDF-1α) in a trans-well filter assembly was also reduced for these cells. Semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) analysis revealed decreased expression of CXCR4 and β2 integrin and increased expression of arginase II in IGT subjects. No differences were observed with regard to endothelial differentiation; however, cultured PBMCs of IGT subjects had decreased intracellular nitric oxide (NO) production. CONCLUSION In pre-diabetic, Asian Indian men, PBMCs exhibit defective migration and homing potential.
Collapse
Affiliation(s)
- Abel Arul Nathan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - Mallu Abhiram Charan Tej
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - Maneka Chitiprolu
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - Shreyas Rangan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| | - Viswanathan Mohan
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF) and Dr Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, India
| | - Ranjani Harish
- Department of Diabetology, Madras Diabetes Research Foundation (MDRF) and Dr Mohan's Diabetes Specialties Centre, Gopalapuram, Chennai, India
| | - Setty B Anand
- Department of Genetic Engineering, Madurai Kamaraj University, Madurai, India
| | - Madhulika Dixit
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences and Bioengineering, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
387
|
Hamburg NM, Leeper NJ. Therapeutic Potential of Modulating MicroRNA in Peripheral Artery Disease. Curr Vasc Pharmacol 2015; 13:316-23. [PMID: 23713861 PMCID: PMC4886469 DOI: 10.2174/15701611113119990014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 12/16/2022]
Abstract
Peripheral artery disease (PAD) produces significant disability attributable to lower extremity ischemia. Limited treatment modalities exist to ameliorate clinical symptoms in patients with PAD. Growing evidence links microRNAs to key processes that govern disease expression in PAD including angiogenesis, endothelial function, inflammation, vascular regeneration, vascular smooth muscle cell function, restenosis, and mitochondrial function. MicroRNAs have been identified in circulation and may serve as novel biomarkers in PAD. This article reviews the potential contribution of microRNA to key pathways of disease development in PAD that may lead to microRNA-based diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Naomi M Hamburg
- Section of Cardiology, Boston Medical Center, 88 East Newton St., Boston, MA, 02118.
| | | |
Collapse
|
388
|
Goh ET, Wong E, Farhatnia Y, Tan A, Seifalian AM. Accelerating in situ endothelialisation of cardiovascular bypass grafts. Int J Mol Sci 2014; 16:597-627. [PMID: 25551605 PMCID: PMC4307264 DOI: 10.3390/ijms16010597] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 12/19/2014] [Indexed: 12/18/2022] Open
Abstract
The patency of synthetic cardiovascular grafts in the long run is synonymous with their ability to inhibit the processes of intimal hyperplasia, thrombosis and calcification. In the human body, the endothelium of blood vessels exhibits characteristics that inhibit such processes. As such it is not surprising that research in tissue engineering is directed towards replicating the functionality of the natural endothelium in cardiovascular grafts. This can be done either by seeding the endothelium within the lumen of the grafts prior to implantation or by designing the graft such that in situ endothelialisation takes place after implantation. Due to certain difficulties identified with in vitro endothelialisation, in situ endothelialisation, which will be the focus of this article, has garnered interest in the last years. To promote in situ endothelialisation, the following aspects can be taken into account: (1) Endothelial progenital cell mobilization, adhesion and proliferation; (2) Regulating differentiation of progenitor cells to mature endothelium; (3) Preventing thrombogenesis and inflammation during endothelialisation. This article aims to review and compile recent developments to promote the in situ endothelialisation of cardiovascular grafts and subsequently improve their patency, which can also have widespread implications in the field of tissue engineering.
Collapse
Affiliation(s)
- Ee Teng Goh
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Eleanor Wong
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Yasmin Farhatnia
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Aaron Tan
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| | - Alexander M Seifalian
- Centre for Nanotechnology & Regenerative Medicine, Research Department of Nanotechnology, UCL Division of Surgery & Interventional Science, University College London (UCL), London NW3 2QG, UK.
| |
Collapse
|
389
|
Di Stefano R, Felice F, Pini S, Mazzotta G, Bovenzi FM, Bertoli D, Abelli M, Borelli L, Cardini A, Lari L, Gesi C, Muccignat A, Oligeri C, Michi P, Balbarini A. Impact of depression on circulating endothelial progenitor cells in patients with acute coronary syndromes: a pilot study. J Cardiovasc Med (Hagerstown) 2014; 15:353-9. [PMID: 24685963 DOI: 10.2459/jcm.0b013e328365c195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Depression has been identified as a risk factor for an adverse prognosis and reduced survival in patients with acute coronary syndrome (ACS). The number of endothelial progenitor cells (EPCs) is an independent predictor of clinical outcomes in patients with ACS. The aim of this study was to evaluate the impact of depression on EPC levels in patients with ACS. METHODS Out of 74 ACS patients [23 non-ST-segment elevation myocardial infarction (NSTEMI), 48 STEMI], 36 had a diagnosis of major depressive episode (MDE) according to the Diagnostic and Statistical Manual of Mental Disorders 4th edition (DSM-IV) criteria at the time of the inclusion in the study. Control groups were as follows: 15 healthy individuals and 18 patients with current MDE without a history of cardiovascular diseases. EPCs were defined as CD34CD133KDR and evaluated by flow cytometry. All patients underwent standardized cardiological and psychopathological evaluations. Parametric and nonparametric statistical tests were performed wherever appropriate. RESULTS ACS patients with MDE showed a significant decrease in circulating EPC number compared with ACS patients without MDE (P < 0.001). The ACS study population was then subdivided into STEMI and NSTEMI groups, and within each group patients with MDE again showed a significant decrease in circulating CD34CD133KDR EPCs compared with others (P <0.001). CONCLUSION We showed that ACS patients with MDE have a reduced number of circulating CD34CD133KDR cells compared with ACS patients without MDE, suggesting that the presence of MDE reduces the response of bone marrow to acute ischemic events. Considering the reparative role of EPCs in ACS patients, we propose that patients with MDE might be protected less than patients without MDE.
Collapse
Affiliation(s)
- Rossella Di Stefano
- aDepartment of Surgical, Medical and Molecular Pathology and Critic Area bDepartment of Clinical and Experimental Medicine, Psychiatry Sector, University of Pisa, Pisa cUnit of Cardiology, Ospedale S. Andrea, La Spezia dCardio-Respiratory Department, Ospedale Campo di Marte, Lucca eUnit of Cardiology, USL5 Sarzana (SP), Italy *Rossella Di Stefano and Francesca Felice contributed equally to the writing of this article
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
390
|
Werner CM, Schirmer SH, Gensch C, Pavlickova V, Pöss J, Wright MB, Böhm M, Laufs U. The dual PPARα/γ agonist aleglitazar increases the number and function of endothelial progenitor cells: implications for vascular function and atherogenesis. Br J Pharmacol 2014; 171:2685-703. [PMID: 24467636 DOI: 10.1111/bph.12608] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/30/2013] [Accepted: 01/16/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Aleglitazar is a dual PPARα/γ agonist but little is known about its effects on vascular function and atherogenesis. Hence, we characterized its effects on circulating angiogenic cells (CAC), neoangiogenesis, endothelial function, arteriogenesis and atherosclerosis in mice. EXPERIMENTAL APPROACH C57Bl/6 wild-type (WT, normal chow), endothelial NOS (eNOS)(-/-) (normal chow) and ApoE(-/-) (Western-type diet) mice were treated with aleglitazar (10 mg·kg(-1) ·day(-1) , i.p.) or vehicle. KEY RESULTS Aleglitazar enhanced expression of PPARα and PPARγ target genes, normalized glucose tolerance and potently reduced hepatic fat in ApoE(-/-) mice. In WT mice, but not in eNOS(-/-) , aleglitazar up-regulated Sca-1/VEGFR2-positive CAC in the blood and bone marrow and up-regulated diLDL/lectin-positive CAC. Aleglitazar augmented CAC migration and enhanced neoangiogenesis. In ApoE(-/-) mice, aleglitazar up-regulated CAC number and function, reduced markers of vascular inflammation and potently improved perfusion restoration after hindlimb ischaemia and aortic endothelium-dependent vasodilatation. This was associated with markedly reduced formation of atherosclerotic plaques. In human cultured CAC from healthy donors and patients with coronary artery disease with or without diabetes mellitus, aleglitazar increased migration and colony-forming units in a concentration-dependent manner. Furthermore, oxidative stress-induced CAC apoptosis and expression of p53 were reduced, while telomerase activity and expression of phospho-eNOS and phospho-Akt were elevated. Comparative agonist and inhibitor experiments revealed that aleglitazar's effects on CAC migration and colony-forming units were mediated by both PPARα and PPARγ signalling and required Akt. CONCLUSIONS AND IMPLICATIONS Aleglitazar augments the number, function and survival of CAC, which correlates with improved vascular function, enhanced arteriogenesis and prevention of atherosclerosis in mice.
Collapse
Affiliation(s)
- C M Werner
- Klinik für Innere Medizin III (Kardiologie, Angiologie und Internistische Intensivmedizin), Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | | | | | | | | | | | | | | |
Collapse
|
391
|
Fadini GP, Rigato M, Boscari F, Cappellari R, Menegazzo L, Pilutti C, Iori E, Marescotti M, Plebani M, Albiero M, Avogaro A. Short-term statin discontinuation increases endothelial progenitor cells without inflammatory rebound in type 2 diabetic patients. Vascul Pharmacol 2014; 67-69:21-9. [PMID: 25483979 DOI: 10.1016/j.vph.2014.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/24/2014] [Accepted: 11/15/2014] [Indexed: 11/24/2022]
Abstract
Type 2 diabetes (T2D) is characterized by impaired vascular regeneration owing to reduced endothelial progenitor cells (EPCs). While statins are known to increase EPCs, the effects of statin withdrawal on EPCs are unknown. Herein, we evaluated the effects of statin discontinuation on EPCs, inflammation and in vivo angiogenesis. Thirty-four T2D patients were randomized to 5-day discontinuation or continuation of statin treatment. At baseline and at day 5, we determined lipid profile, EPC levels, monocyte-macrophage polarization, and concentrations of hsCRP, VEGF, SDF-1α, and G-CSF. Angiogenesis by human circulating cells was assessed in vivo. At day 5, patients who stopped statins showed raised total and LDL cholesterol and EPCs compared to baseline, while no changes were observed in patients who continued statins. No changes were observed in hsCRP, VEGF, SDF-1α, G-CSF, M1 and M2 macrophages and classical, intermediate and nonclassical monocytes in both groups. In vivo angiogenesis by circulating cells was increased in patients who stopped statin treatment. In vitro, cholesterol supplementation stimulated mobilizing signals in human bone marrow mesenchymal stem cells. In conclusion, a brief statin withdrawal increases circulating EPCs and functional proangiogenic cells in T2D. These findings identify statin-sensitive pathways as reverse target mechanisms to stimulate vascular repair in diabetes.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Mauro Rigato
- Department of Medicine, University of Padova, Italy
| | | | | | - Lisa Menegazzo
- Department of Medicine, University of Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy
| | | | | | | | | | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
392
|
Madonna R, Ferdinandy P, De Caterina R, Willerson JT, Marian AJ. Recent developments in cardiovascular stem cells. Circ Res 2014; 115:e71-8. [PMID: 25477490 DOI: 10.1161/circresaha.114.305567] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Rosalinda Madonna
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Peter Ferdinandy
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Raffaele De Caterina
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - James T Willerson
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.)
| | - Ali J Marian
- From the Center of Excellence on Aging, Institute of Cardiology, Department of Neuroscience and Imaging, "G. d'Annunzio" University, Chieti, Italy (R.M., R.D.C.); Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Texas Heart Institute, Houston (R.M., J.T.W.); Division of Cardiology, Department of Internal Medicine (R.M., J.T.W., A.J.M.), and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (A.J.M.).
| |
Collapse
|
393
|
McNeill B, Vulesevic B, Ostojic A, Ruel M, Suuronen EJ. Collagen matrix-induced expression of integrin αVβ3 in circulating angiogenic cells can be targeted by matricellular protein CCN1 to enhance their function. FASEB J 2014; 29:1198-207. [PMID: 25466895 DOI: 10.1096/fj.14-261586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/03/2014] [Indexed: 01/03/2023]
Abstract
Circulating angiogenic cells (CACs) play an important role in vascular homeostasis and hold therapeutic promise for treating a variety of cardiovascular diseases. However, further improvements are needed because the effects of CAC therapy remain minimal or transient. The regenerative potential of these cells can be improved by culture on a collagen-based matrix through the up-regulation of key integrin proteins. We found that human CAC function was enhanced by using the matricellular protein CCN1 (CYR61/CTGF/NOV family member 1) to target integrin αV and β3, which are up-regulated on matrix. Compared to matrix-cultured CACs, CCN1-matrix CACs exhibited a 2.2-fold increase in cell proliferation, 1.8-fold greater migration toward VEGF, and 1.7-fold more incorporation into capillary-like structures in an angiogenesis assay. In vivo, intramuscular injection of CCN1-matrix-cultured CACs into ischemic hind limbs of CD-1 nude mice resulted in blood flow recovery to 80% of baseline, which was greater than matrix-cultured CACs (66%) and PBS (35%) treatment groups. Furthermore, transplanted CCN1-matrix CACs exhibited greater engraftment (11-fold) and stimulated the up-regulation of survival and angiogenic genes (>3-fold). These findings reveal the importance of cell-matrix interactions in regulating CAC function and also reveal a mechanism by which these may be exploited to enhance cell therapies for ischemic disease.
Collapse
Affiliation(s)
- Brian McNeill
- *Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Branka Vulesevic
- *Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Aleksandra Ostojic
- *Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc Ruel
- *Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Erik J Suuronen
- *Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada; and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
394
|
Circulating endothelial progenitor cell: a promising biomarker in clinical oncology. Med Oncol 2014; 32:332. [PMID: 25428376 DOI: 10.1007/s12032-014-0332-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 12/11/2022]
Abstract
Human cancers are endowed with sustained vascularization capability, and their growth, invasion, and metastasis are vascularization dependent. Recently, accumulated body of evidence suggests that endothelial progenitor cells (EPCs) can support vasculogenesis and induce angiogenesis through paracrine mechanisms. In addition, numerous clinical studies have revealed the increase in the number of EPCs in the peripheral blood of cancer patients and demonstrated the correlation of circulating EPCs (CEPCs) with the clinical outcomes. This review highlights current enrichment procedures and methods for the detection of CEPCs and different biomarkers to identify CEPCs as well as the functions of EPCs in tumor vascularization. Furthermore, we systematically review available studies on the clinical relevance of CEPCs in cancer patients to explore the potential diagnostic and prognostic values of CEPCs. Although several contrasting results exist, CEPCs can conceivably serve as a promising biomarker for the early diagnosis, prognosis prediction, and treatment response indication in the future. Additionally, further well-designed clinical studies with larger sample size and unique, specific enumeration procedures are warranted to achieve further insight into the clinical implications of CEPCs.
Collapse
|
395
|
Li J, Zhang Q, Li D, An Y, Kutryk MBJ. Hydroxybutyl Chitosan Polymer-Mediated CD133 Antibody Coating of Metallic Stents to Reduce Restenosis in a Porcine Model of Atherosclerosis. J Cardiovasc Pharmacol Ther 2014; 20:322-9. [PMID: 25412893 DOI: 10.1177/1074248414558518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/07/2014] [Indexed: 12/11/2022]
Abstract
Antibody-coated stents to capture circulating endothelial progenitor cells (EPCs) for re-endothelialization appear to be a novel therapeutic option for the treatment of atherosclerotic disease. Hydroxybutyl chitosan (HBC), a linear polysaccharide made from shrimps and other crustacean shells, is biocompatible, nontoxic, and hydrophilic, making it ideal for biomedical applications. In this study, HBC was explored for the immobilization of anti-CD133 antibodies. We demonstrated that CD133 antibodies mediated by HBC were successfully coated on cobalt-chromium alloy discs and metal stents. The coating was homogeneous and smooth as shown by electronic microscopy analysis. Balloon expansion of coated stents did not cause cracking or peeling. The HBC discs promoted CD133+ EPCs and human umbilical vein endothelial cell growth in vitro. The CD133 antibody-coated but not bare discs bound CD133+ EPCs in vitro. Implantation of CD133 antibody-coated stents significantly inhibited intimal hyperplasia and reduced restenosis compared with implantation of bare stents in a porcine model of atherosclerosis. These findings suggest HBC is a valuable anchoring agent that can be applied for bioactive coating of stents and that CD133 antibody-coated stents might be a potential therapeutic alternative for the treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Jian Li
- Department of Cardiology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Dan Li
- Department of Cardiology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Yi An
- Department of Cardiology, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China
| | - Michael B J Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Canada
| |
Collapse
|
396
|
Lucchesi D, Russo R, Gabriele M, Longo V, Del Prato S, Penno G, Pucci L. Grain and bean lysates improve function of endothelial progenitor cells from human peripheral blood: involvement of the endogenous antioxidant defenses. PLoS One 2014; 9:e109298. [PMID: 25329912 PMCID: PMC4201454 DOI: 10.1371/journal.pone.0109298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/29/2014] [Indexed: 12/14/2022] Open
Abstract
Increased oxidative stress contributes to the functional impairment of endothelial progenitor cells (EPCs), the pivotal players in the servicing of the endothelial cell lining. Several evidences suggest that decreasing oxidative stress by natural compounds with antioxidant properties may improve EPCs bioactivity. Here, we investigated the effects of Lisosan G (LG), a Triticum Sativum grain powder, and Lady Joy (LJ), a bean lysate, on function of EPCs exposed to oxidative stress. Peripheral blood mononuclear cells were isolated and plated on fibronectin-coated culture dishes; adherent cells, identified as early EPCs, were pre-treated with different concentrations of LG and LJ and incubated with hydrogen peroxide (H2O2). Viability, senescence, adhesion, ROS production and antioxidant enzymes gene expression were evaluated. Lysate-mediated Nrf-2 (nuclear factor (erythroid-derived 2)-like 2)/ARE (antioxidant response element) activation, a modulator of oxidative stress, was assessed by immunocytochemistry. Lady Joy 0.35-0.7 mg/ml increases EPCs viability; pre-treatment with either LG 0.7 mg/ml and LJ 0.35-0.7 mg/ml protect EPCs viability against H2O2-induced injury. LG 0.7 and LJ 0.35-0.7 mg/ml improve EPCs adhesion; pre-treatment with either LG 0.35 and 0.7 mg/ml or LJ 0.35, 0.7 and 1.4 mg/ml preserve adhesiveness of EPCs exposed to H2O2. Senescence is attenuated in EPCs incubated with lysates 0.35 mg/ml. After exposure to H2O2, LG pre-treated cells show a lower senescence than untreated EPCs. Lysates significantly decrease H2O2-induced ROS generation. Both lysates increase glutathione peroxidase-1 and superoxide dismutase-2 (SOD-2) expression; upon H2O2 exposure, pre-treatment with LJ allows higher SOD-2 expression. Heme oxigenase-1 increases in EPCs pre-treated with LG even upon H2O2 exposure. Finally, incubation with LG 0.7 mg/ml results in Nrf-2 translocation into the nucleus both at baseline and after the oxidative challenge. Our data suggest a protective effect of lysates on EPCs exposed to oxidative stress through the involvement of antioxidant systems. Lisosan G seems to activate the Nrf-2/ARE pathways.
Collapse
Affiliation(s)
- Daniela Lucchesi
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Rossella Russo
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Morena Gabriele
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Stefano Del Prato
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Giuseppe Penno
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| |
Collapse
|
397
|
Difference in mobilization of progenitor cells after myocardial infarction in smoking versus non-smoking patients: insights from the BONAMI trial. Stem Cell Res Ther 2014; 4:152. [PMID: 24423369 PMCID: PMC4054959 DOI: 10.1186/scrt382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/14/2013] [Accepted: 12/10/2013] [Indexed: 01/15/2023] Open
Abstract
Introduction Although autologous bone marrow cell (BMC) therapy has emerged as a promising treatment for acute myocardial infarction (AMI), trials reported mixed results. In the BONAMI trial, active smoking reduced cardiac function recovery after reperfused AMI. Therefore, we hypothesized that variability in the functionality of BMCs retrieved from patients with cardiovascular risk factors may partly explain these mixed results. We investigated the characteristics of progenitor cells in active smokers and non-smokers with AMI and their potential impact on BMC therapy efficacy. Methods Bone marrow and blood samples from 54 smoking and 47 non-smoking patients enrolled in the BONAMI cell therapy trial were analyzed. Results The white BMC and CD45dimCD34+ cell numbers were higher in active smokers (P = 0.001, P = 0.03, respectively). In marked contrast, either bone marrow or blood endothelial progenitor CD45dimCD34 + KDR + cells (EPCs) were decreased in active smokers (P = 0.005, P = 0.04, respectively). Importantly, a multivariate analysis including cardiovascular risk factors confirmed the association between active smoking and lower EPC number in bone marrow (P = 0.04) and blood (P = 0.04). Furthermore, baseline circulating EPC count predicted infarct size decrease at three months post-AMI in non-smokers (P = 0.01) but not in active smokers. Interestingly, baseline circulating EPCs were no longer predictive of cardiac function improvement in the BMC therapy group. Conclusions These data suggest that circulating EPCs play an important role in cardiac repair post-AMI only in non-smokers and that active smoking-associated EPC alterations may participate in the impairment of cardiac function recovery observed in smokers after AMI, an effect that was overridden by BMC therapy.
Collapse
|
398
|
Patel RS, Li Q, Ghasemzadeh N, Eapen DJ, Moss LD, Janjua AU, Manocha P, Kassem HA, Veledar E, Samady H, Taylor WR, Zafari AM, Sperling L, Vaccarino V, Waller EK, Quyyumi AA. Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease. Circ Res 2014; 116:289-297. [PMID: 25323857 DOI: 10.1161/circresaha.116.304187] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Low circulating progenitor cell numbers and activity may reflect impaired intrinsic regenerative/reparative potential, but it remains uncertain whether this translates into a worse prognosis. OBJECTIVES To investigate whether low numbers of progenitor cells associate with a greater risk of mortality in a population at high cardiovascular risk. METHODS AND RESULTS Patients undergoing coronary angiography were recruited into 2 cohorts (1, n=502 and 2, n=403) over separate time periods. Progenitor cells were enumerated by flow cytometry as CD45(med+) blood mononuclear cells expressing CD34, with additional quantification of subsets coexpressing CD133, vascular endothelial growth factor receptor 2, and chemokine (C-X-C motif) receptor 4. Coefficient of variation for CD34 cells was 2.9% and 4.8%, 21.6% and 6.5% for the respective subsets. Each cohort was followed for a mean of 2.7 and 1.2 years, respectively, for the primary end point of all-cause death. There was an inverse association between CD34(+) and CD34(+)/CD133(+) cell counts and risk of death in cohort 1 (β=-0.92, P=0.043 and β=-1.64, P=0.019, respectively) that was confirmed in cohort 2 (β=-1.25, P=0.020 and β=-1.81, P=0.015, respectively). Covariate-adjusted hazard ratios in the pooled cohort (n=905) were 3.54 (1.67-7.50) and 2.46 (1.18-5.13), respectively. CD34(+)/CD133(+) cell counts improved risk prediction metrics beyond standard risk factors. CONCLUSIONS Reduced circulating progenitor cell counts, identified primarily as CD34(+) mononuclear cells or its subset expressing CD133, are associated with risk of death in individuals with coronary artery disease, suggesting that impaired endogenous regenerative capacity is associated with increased mortality. These findings have implications for biological understanding, risk prediction, and cell selection for cell-based therapies.
Collapse
Affiliation(s)
- Riyaz S Patel
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Institute of Cardiovascular Sciences, University College London, London, UK
| | - Qunna Li
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nima Ghasemzadeh
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Danny J Eapen
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lauren D Moss
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - A Umair Janjua
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Pankaj Manocha
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hatem Al Kassem
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Emir Veledar
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Dept of Medicine, Baptist Health South Florida, Florida, USA
| | - Habib Samady
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - W Robert Taylor
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - A Maziar Zafari
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Dept of Medicine, Baptist Health South Florida, Florida, USA.,Dept. of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Laurence Sperling
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Dept. of Medicine, Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Edmund K Waller
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Dept. of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
399
|
Bongiovanni D, Bassetti B, Gambini E, Gaipa G, Frati G, Achilli F, Scacciatella P, Carbucicchio C, Pompilio G. The CD133+Cell as Advanced Medicinal Product for Myocardial and Limb Ischemia. Stem Cells Dev 2014; 23:2403-21. [DOI: 10.1089/scd.2014.0111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Dario Bongiovanni
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
- Cardiovascular and Thoracic Diseases Department, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Beatrice Bassetti
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Elisa Gambini
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giuseppe Gaipa
- Laboratorio Interdipartimentale di Terapia Cellulare Stefano Verri, Azienda Ospedaliera San Gerardo, Monza, Milan, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Department of AngioCardioNeurology, IRCCS NeuroMed, Pozzilli, Italy
| | - Felice Achilli
- Department of Cardiology, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Paolo Scacciatella
- Cardiovascular and Thoracic Diseases Department, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Corrado Carbucicchio
- Cardiac Arrhythmia Research Centre, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Giulio Pompilio
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
- Department of Clinical and Community Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
400
|
Mangialardi G, Spinetti G, Reni C, Madeddu P. Reactive oxygen species adversely impacts bone marrow microenvironment in diabetes. Antioxid Redox Signal 2014; 21:1620-33. [PMID: 25089632 PMCID: PMC4175424 DOI: 10.1089/ars.2014.5944] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Significance: Patients with diabetes mellitus suffer an excess of cardiovascular complications and recover worse from them as compared with their nondiabetic peers. It is well known that microangiopathy is the cause of renal damage, blindness, and heart attacks in patients with diabetes. This review highlights molecular deficits in stem cells and a supporting microenvironment, which can be traced back to oxidative stress and ultimately reduce stem cells therapeutic potential in diabetic patients. RECENT ADVANCES New research has shown that increased oxidative stress contributes to inducing microangiopathy in bone marrow (BM), the tissue contained inside the bones and the main source of stem cells. These precious cells not only replace old blood cells but also exert an important reparative function after acute injuries and heart attacks. CRITICAL ISSUES The starvation of BM as a consequence of microangiopathy can lead to a less efficient healing in diabetic patients with ischemic complications. Furthermore, stem cells from a patient's BM are the most used in regenerative medicine trials to mend hearts damaged by heart attacks. FUTURE DIRECTIONS A deeper understanding of redox signaling in BM stem cells will lead to new modalities for preserving local and systemic homeostasis and to more effective treatments of diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- 1 Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol , Bristol, United Kingdom
| | | | | | | |
Collapse
|