201
|
Abstract
Endothelial progenitor cells (EPCs) play a critical role in maintenance of the endothelial integrity and vascular homeostasis, as well as in neovascularization. Dysfunctional EPCs are believed to contribute to the endothelial dysfunction and are closely related to the development of various cardiovascular diseases, such as hypertension, hyperlipidemia, and stroke. However, the underlying mechanisms of EPC dysfunction are complicated and remain largely elusive. Recent studies have demonstrated that reactive oxygen species (ROS) are key factors that involve in modulation of stem and progenitor cell function under various physiologic and pathologic conditions. It has been shown that NADPH oxidase (NOX)-derived ROS are the major sources of ROS in cardiovascular system. Accumulating evidence suggests that NOX-mediated oxidative stress can modulate EPC bioactivities, such as mobilization, migration, and neovascularization, and that inhibition of NOX has been shown to improve EPC functions. This review summarized recent progress in the studies on the correlation between NOX-mediated EPC dysfunction and cardiovascular diseases.
Collapse
|
202
|
Isolation of Foreign Material-Free Endothelial Progenitor Cells Using CD31 Aptamer and Therapeutic Application for Ischemic Injury. PLoS One 2015; 10:e0131785. [PMID: 26148001 PMCID: PMC4493074 DOI: 10.1371/journal.pone.0131785] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/05/2015] [Indexed: 01/09/2023] Open
Abstract
Endothelial progenitor cells (EPCs) can be isolated from human bone marrow or peripheral blood and reportedly contribute to neovascularization. Aptamers are 40-120-mer nucleotides that bind to a specific target molecule, as antibodies do. To utilize apatmers for isolation of EPCs, in the present study, we successfully generated aptamers that recognize human CD31, an endothelial cell marker. CD31 aptamers bound to human umbilical cord blood-derived EPCs and showed specific interaction with human CD31, but not with mouse CD31. However, CD31 aptamers showed non-specific interaction with CD31-negative 293FT cells and addition of polyanionic competitor dextran sulfate eliminated non-specific interaction without affecting cell viability. From the mixture of EPCs and 293FT cells, CD31 aptamers successfully isolated EPCs with 97.6% purity and 94.2% yield, comparable to those from antibody isolation. In addition, isolated EPCs were decoupled from CD31 aptamers with a brief treatment of high concentration dextran sulfate. EPCs isolated with CD31 aptamers and subsequently decoupled from CD31 aptamers were functional and enhanced the restoration of blood flow when transplanted into a murine hindlimb ischemia model. In this study, we demonstrated isolation of foreign material-free EPCs, which can be utilized as a universal protocol in preparation of cells for therapeutic transplantation.
Collapse
|
203
|
Salvatore P, Zullo A, Sommese L, Colicchio R, Picascia A, Schiano C, Mancini FP, Napoli C. Infections and cardiovascular disease: is Bartonella henselae contributing to this matter? J Med Microbiol 2015; 64:799-809. [PMID: 26066633 DOI: 10.1099/jmm.0.000099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is still the major cause of death worldwide despite the remarkable progress in its prevention and treatment. Endothelial progenitor cells (EPCs) have recently emerged as key players of vascular repair and regenerative medicine applied to cardiovascular disease. A large amount of effort has been put into discovering the factors that could aid or impair the number and function of EPCs, and also into characterizing these cells at the molecular level in order to facilitate their therapeutic applications in vascular disease. Interestingly, the major cardiovascular risk factors have been associated with reduced number and function of EPCs. The bacterial contribution to cardiovascular disease represents a long-standing controversy. The discovery that Bartonella henselae can infect and damage EPCs revitalizes the enduring debate about the microbiological contribution to atherosclerosis, thus allowing the hypothesis that this infection could impair the cardiovascular regenerative potential and increase the risk for cardiovascular disease. In this review, we summarize the rationale suggesting that Bartonella henselae could favour atherogenesis by infecting and damaging EPCs, thus reducing their vascular repair potential. These mechanisms suggest a novel link between communicable and non-communicable human diseases, and put forward the possibility that Bartonella henselae could enhance the susceptibility and worsen the prognosis in cardiovascular disease.
Collapse
Affiliation(s)
- Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.,CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Alberto Zullo
- CEINGE-Advanced Biotechnologies, Naples, Italy.,Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Linda Sommese
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.,Department of Experimental Medicine, Section of Microbiology, Second University of Naples, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Antonietta Picascia
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.,U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Concetta Schiano
- Foundation SDN, Institute of Diagnostic and Nuclear Development, IRCCS, Naples, Italy
| | | | - Claudio Napoli
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.,Foundation SDN, Institute of Diagnostic and Nuclear Development, IRCCS, Naples, Italy
| |
Collapse
|
204
|
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: 2.1] [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
|
205
|
Abstract
Despite substantial clinical advances over the past 65 years, cardiovascular disease remains the leading cause of death in America. The past 15 years has witnessed major basic and translational interest in the use of stem and precursor cells as a therapeutic agent for chronically injured organs. Among the cell types under investigation, adult mesenchymal stem cells are widely studied, and in early stage, clinical studies show promise for repair and regeneration of cardiac tissues. The ability of mesenchymal stem cells to differentiate into mesoderm- and nonmesoderm-derived tissues, their immunomodulatory effects, their availability, and their key role in maintaining and replenishing endogenous stem cell niches have rendered them one of the most heavily investigated and clinically tested type of stem cell. Accumulating data from preclinical and early phase clinical trials document their safety when delivered as either autologous or allogeneic forms in a range of cardiovascular diseases, but also importantly define parameters of clinical efficacy that justify further investigation in larger clinical trials. Here, we review the biology of mesenchymal stem cells, their interaction with endogenous molecular and cellular pathways, and their modulation of immune responses. Additionally, we discuss factors that enhance their proliferative and regenerative ability and factors that may hinder their effectiveness in the clinical setting.
Collapse
Affiliation(s)
- Vasileios Karantalis
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL
| | - Joshua M Hare
- From the University of Miami Miller School of Medicine, Interdisciplinary Stem Cell Institute, FL.
| |
Collapse
|
206
|
Melanoma cell therapy: Endothelial progenitor cells as shuttle of the MMP12 uPAR-degrading enzyme. Oncotarget 2015; 5:3711-27. [PMID: 25003596 PMCID: PMC4116515 DOI: 10.18632/oncotarget.1987] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The receptor for the urokinase-type plasminogen activator (uPAR) accounts for many features of cancer progression, and is therefore considered a target for anti-tumoral therapy. Only full length uPAR mediates tumor progression. Matrix-metallo-proteinase-12 (MMP12)-dependent uPAR cleavage results into the loss of invasion properties and angiogenesis. MMP12 can be employed in the field of “targeted therapies” as a biological drug to be delivered directly in patient's tumor mass. Endothelial Progenitor Cells (EPCs) are selectively recruited within the tumor and could be used as cellular vehicles for delivering anti-cancer molecules. The aim of our study is to inhibit cancer progression by engeneering ECFCs, a subset of EPC, with a lentivirus encoding the anti-tumor uPAR-degrading enzyme MMP12. Ex vivo manipulated ECFCs lost the capacity to perform capillary morphogenesis and acquired the anti-tumor and anti-angiogenetic activity. In vivo MMP12-engineered ECFCs cleaved uPAR within the tumor mass and strongly inhibited tumor growth, tumor angiogenesis and development of lung metastasis. The possibility to exploit tumor homing and activity of autologous MMP12-engineered ECFCs represents a novel way to combat melanoma by a “personalized therapy”, without rejection risk. The i.v. injection of radiolabelled MMP12-ECFCs can thus provide a new theranostic approach to control melanoma progression and metastasis.
Collapse
|
207
|
Nishimura R, Nishiwaki T, Kawasaki T, Sekine A, Suda R, Urushibara T, Suzuki T, Takayanagi S, Terada J, Sakao S, Tatsumi K. Hypoxia-induced proliferation of tissue-resident endothelial progenitor cells in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 308:L746-58. [PMID: 25502500 DOI: 10.1152/ajplung.00243.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/05/2014] [Indexed: 11/22/2022] Open
Abstract
Exposure to hypoxia induces changes in the structure and functional phenotypes of the cells composing the pulmonary vascular wall from larger to most peripheral vessels. Endothelial progenitor cells (EPCs) may be involved in vascular endothelial repair. Resident EPCs with a high proliferative potential are found in the pulmonary microcirculation. However, their potential location, identification, and functional role have not been clearly established. We investigated whether resident EPCs or bone marrow (BM)-derived EPCs play a major role in hypoxic response of pulmonary vascular endothelial cells (PVECs). Mice were exposed to hypoxia. The number of PVECs transiently decreased followed by an increase in hypoxic animals. Under hypoxic conditions for 1 wk, prominent bromodeoxyuridine incorporation was detected in PVECs. Some Ki67-positive cells were detected among PVECs after 1 wk under hypoxic conditions, especially in the capillaries. To clarify the origin of proliferating endothelial cells, we used BM chimeric mice expressing green fluorescent protein (GFP). The percentage of GFP-positive PVECs was low and constant during hypoxia in BM-transplanted mice, suggesting little engraftment of BM-derived cells in lungs under hypoxia. Proliferating PVECs in hypoxic animals showed increased expression of CD34, suggesting hypoxia-induced gene expression and cell surface antigen of EPC or stem/progenitor cells markers. Isolated PVECs from hypoxic mice showed colony- and tube-forming capacity. The present study indicated that hypoxia could induce proliferation of PVECs, and the origin of these cells might be tissue-resident EPCs.
Collapse
Affiliation(s)
- Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsu Nishiwaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Urushibara
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin Takayanagi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
208
|
Martí-Fàbregas J, Delgado-Mederos R, Crespo J, Peña E, Marín R, Jiménez-Xarrié E, Fernández-Arcos A, Pérez-Pérez J, Martínez-Domeño A, Camps-Renom P, Prats-Sánchez L, Casoni F, Badimon L. Circulating endothelial progenitor cells and the risk of vascular events after ischemic stroke. PLoS One 2015; 10:e0124895. [PMID: 25874380 PMCID: PMC4395144 DOI: 10.1371/journal.pone.0124895] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 03/07/2015] [Indexed: 11/18/2022] Open
Abstract
Background and Purpose We evaluated the hypothesis that the number of circulating EPC could be associated with the risk of stroke recurrence (SR) or vascular events (VE) after an ischemic stroke. Methods We studied prospectively consecutive patients with cerebral infarction within the first 48 hours after the onset. We recorded demographic factors, vascular risk factors, previous Rankin scale (RS) score, and etiology. We analyzed EPC counts by flow cytometry in blood collected at day 7 and defined EPC as CD34+/CD133+/KDR+ cells. Mean follow-up was 29.3 ± 16 months. We evaluated SR as well as VE. Patients were classified as to the presence or absence of EPC in the circulation (either EPC+ or EPC-). Bivariate analyses, Kaplan-Meier survival curves and Cox regression models were used. Results We included 121 patients (mean age 70.1±12.6 years; 65% were men). The percentage of EPC+ patients was 47.1%. SR occurred in 12 (9.9%) and VE in 18 (14.9%) patients. SR was associated significantly with a worse prior RS score, previous stroke and etiology, but not with EPC count. VE were associated significantly with EPC-, worse prior RS score, previous stroke, high age, peripheral artery disease and etiology. Cox regression model showed that EPC- (HR 7.07, p=0.003), age (HR 1.08, p=0.004) and a worse prior RS score (HR 5.8, p=0.004) were associated significantly with an increased risk of VE. Conclusions The absence of circulating EPC is not associated with the risk of stroke recurrence, but is associated with an increased risk of future vascular events.
Collapse
Affiliation(s)
- Joan Martí-Fàbregas
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- * E-mail:
| | - Raquel Delgado-Mederos
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Rebeca Marín
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Elena Jiménez-Xarrié
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Ana Fernández-Arcos
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Jesús Pérez-Pérez
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | | | - Pol Camps-Renom
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Luís Prats-Sánchez
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Francesca Casoni
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| |
Collapse
|
209
|
Basile DP, Yoder MC. Renal endothelial dysfunction in acute kidney ischemia reperfusion injury. Cardiovasc Hematol Disord Drug Targets 2015; 14:3-14. [PMID: 25088124 DOI: 10.2174/1871529x1401140724093505] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/15/2014] [Accepted: 05/30/2014] [Indexed: 01/11/2023]
Abstract
Acute kidney injury is associated with alterations in vascular tone that contribute to an overall reduction in GFR. Studies in animal models indicate that ischemia triggers alterations in endothelial function that contribute significantly to the overall degree and severity of a kidney injury. Putative mediators of vasoconstriction that may contribute to the initial loss of renal blood flow and GFR are highlighted. In addition, there is discussion of how intrinsic damage to the endothelium impairs homeostatic responses in vascular tone as well as promotes leukocyte adhesion and exacerbating the reduction in renal blood flow. The timing of potential therapies in animal models as they relate to the evolution of AKI, as well as the limitations of such approaches in the clinical setting are discussed. Finally, we discuss how acute kidney injury induces permanent alterations in renal vascular structure. We posit that the cause of the sustained impairment in kidney capillary density results from impaired endothelial growth responses and suggest that this limitation is a primary contributing feature underlying progression of chronic kidney disease.
Collapse
Affiliation(s)
| | - Mervin C Yoder
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Med Sci 334, Indianapolis, IN 46202, USA.
| |
Collapse
|
210
|
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: 3.1] [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
|
211
|
Minami Y, Nakajima T, Ikutomi M, Morita T, Komuro I, Sata M, Sahara M. Angiogenic potential of early and late outgrowth endothelial progenitor cells is dependent on the time of emergence. Int J Cardiol 2015; 186:305-14. [PMID: 25838182 DOI: 10.1016/j.ijcard.2015.03.166] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 02/18/2015] [Accepted: 03/16/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Recent studies have suggested that late-outgrowth endothelial progenitor cells (EPCs) derived from human peripheral blood mononuclear cells (hPBMNCs) might have higher angiogenic potential than classically-defined early-outgrowth EPCs (EOCs). However, it still remains unclear which of "so-called" EPC subpopulations defined in a variety of ways has the highest angiogenic potential. METHODS AND RESULTS We classified hPBMNC-derived EPC subpopulations by the time of their emergence in culture. EOCs were defined as attached cells on culture days 3-7. Late-outgrowth EPCs, defined as the cell forming colonies with cobblestone appearance since day 10, were further classified as follows: "moderate"-outgrowth EPCs (MOCs) emerging on days 10-16, "late"-outgrowth EPCs (LOCs) on days 17-23, and "very late"-outgrowth EPCs (VOCs) on days 24-30. Flow cytometry analyses showed the clear differences of hematopoietic/endothelial markers between EOC (CD31(+)VE-cadherin(-)CD34(-)CD14(+)CD45(+)) and LOC (CD31(+)VE-cadherin(+)CD34(+)CD14(-)CD45(-)). We found that LOCs had the highest proliferation and tube formation capabilities in vitro along with the highest expression of angiogenic genes including KDR and eNOS. To investigate the in vivo therapeutic efficacies, each EPC subpopulation was intravenously transplanted into immunocompromised mice (total 4 × 10(5) cells) after unilateral hindlimb ischemia surgery. The LOC-treated mice exhibited significantly-enhanced blood flow recovery (flow ratios of ischemic/non-ischemic leg: 0.99±0.02 [LOC group] versus 0.67 ± 0.07 to 0.78 ± 0.09 [other groups]; P < 0.05) and augmented capillary collateral formation in ischemic leg, which were attributable to their direct engraftment into host angiogenic vessels (approximately 10%) and paracrine effects. CONCLUSION hPBMNC-derived late-outgrowth EPCs emerging on culture days 17-23 are superior to other EPC subpopulations with regard to therapeutic angiogenic potential.
Collapse
Affiliation(s)
- Yoshiyasu Minami
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Toshiaki Nakajima
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masayasu Ikutomi
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Cardiovascular Medicine, Teikyo University Chiba Medical Center, 3426-3 Anegasaki, Ichihara 299-0111, Japan
| | - Toshihiro Morita
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School of Medicine, 2-10-1 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Makoto Sahara
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
212
|
Herlea-Pana O, Yao L, Heuser-Baker J, Wang Q, Wang Q, Georgescu C, Zou MH, Barlic-Dicen J. Chemokine receptors CXCR2 and CX3CR1 differentially regulate functional responses of bone-marrow endothelial progenitors during atherosclerotic plaque regression. Cardiovasc Res 2015; 106:324-37. [PMID: 25765938 DOI: 10.1093/cvr/cvv111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/07/2015] [Indexed: 01/03/2023] Open
Abstract
AIMS Atherosclerosis manifests itself as arterial plaques, which lead to heart attacks or stroke. Treatments supporting plaque regression are therefore aggressively pursued. Studies conducted in models in which hypercholesterolaemia is reversible, such as the Reversa mouse model we have employed in the current studies, will be instrumental for the development of such interventions. Using this model, we have shown that advanced atherosclerosis regression occurs when lipid lowering is used in combination with bone-marrow endothelial progenitor cell (EPC) treatment. However, it remains unclear how EPCs home to regressing plaques and how they augment atherosclerosis reversal. Here we identify molecules that support functional responses of EPCs during plaque resolution. METHODS AND RESULTS Chemokines CXCL1 and CX3CL1 were detected in the vascular wall of atheroregressing Reversa mice, and their cognate receptors CXCR2 and CX3CR1 were observed on adoptively transferred EPCs in circulation. We tested whether CXCL1-CXCR2 and CX3CL1-CX3CR1 axes regulate functional responses of EPCs during plaque reversal. We show that pharmacological inhibition of CXCR2 or CX3CR1, or genetic inactivation of these two chemokine receptors interfered with EPC-mediated advanced atherosclerosis regression. We also demonstrate that CXCR2 directs EPCs to regressing plaques while CX3CR1 controls a paracrine function(s) of these cells. CONCLUSION CXCR2 and CX3CR1 differentially regulate EPC functional responses during atheroregression. Our study improves understanding of how chemokines and chemokine receptors regulate plaque resolution, which could determine the effectiveness of interventions reducing complications of atherosclerosis.
Collapse
Affiliation(s)
- Oana Herlea-Pana
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Longbiao Yao
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Janet Heuser-Baker
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Qiongxin Wang
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Qilong Wang
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Constantin Georgescu
- Arthritis and Clinical Immunology Programs, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ming-Hui Zou
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jana Barlic-Dicen
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
213
|
Li S, Plouffe BD, Belov AM, Ray S, Wang X, Murthy SK, Karger BL, Ivanov AR. An Integrated Platform for Isolation, Processing, and Mass Spectrometry-based Proteomic Profiling of Rare Cells in Whole Blood. Mol Cell Proteomics 2015; 14:1672-83. [PMID: 25755294 DOI: 10.1074/mcp.m114.045724] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Indexed: 12/29/2022] Open
Abstract
Isolation and molecular characterization of rare cells (e.g. circulating tumor and stem cells) within biological fluids and tissues has significant potential in clinical diagnostics and personalized medicine. The present work describes an integrated platform of sample procurement, preparation, and analysis for deep proteomic profiling of rare cells in blood. Microfluidic magnetophoretic isolation of target cells spiked into 1 ml of blood at the level of 1000-2000 cells/ml, followed by focused acoustics-assisted sample preparation has been coupled with one-dimensional PLOT-LC-MS methodology. The resulting zeptomole detection sensitivity enabled identification of ∼4000 proteins with injection of the equivalent of only 100-200 cells per analysis. The characterization of rare cells in limited volumes of physiological fluids is shown by the isolation and quantitative proteomic profiling of first MCF-7 cells spiked into whole blood as a model system and then two CD133+ endothelial progenitor and hematopoietic cells in whole blood from volunteers.
Collapse
Affiliation(s)
- Siyang Li
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; §Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Brian D Plouffe
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; ¶Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Arseniy M Belov
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; §Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Somak Ray
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts
| | - Xianzhe Wang
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; §Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Shashi K Murthy
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; ¶Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts
| | - Barry L Karger
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; §Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Alexander R Ivanov
- From the ‡Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts; §Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| |
Collapse
|
214
|
Schenck TL, Hopfner U, Chávez MN, Machens HG, Somlai-Schweiger I, Giunta RE, Bohne AV, Nickelsen J, Allende ML, Egaña JT. Photosynthetic biomaterials: a pathway towards autotrophic tissue engineering. Acta Biomater 2015; 15:39-47. [PMID: 25536030 DOI: 10.1016/j.actbio.2014.12.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/02/2014] [Accepted: 12/15/2014] [Indexed: 12/18/2022]
Abstract
Engineered tissues are highly limited by poor vascularization in vivo, leading to hypoxia. In order to overcome this challenge, we propose the use of photosynthetic biomaterials to provide oxygen. Since photosynthesis is the original source of oxygen for living organisms, we suggest that this could be a novel approach to provide a constant source of oxygen supply independently of blood perfusion. In this study we demonstrate that bioartificial scaffolds can be loaded with a solution containing the photosynthetic microalgae Chlamydomonas reinhardtii, showing high biocompatibility and photosynthetic activity in vitro. Furthermore, when photosynthetic biomaterials were engrafted in a mouse full skin defect, we observed that the presence of the microalgae did not trigger a native immune response in the host. Moreover, the analyses showed that the algae survived for at least 5 days in vivo, generating chimeric tissues comprised of algae and murine cells. The results of this study represent a crucial step towards the establishment of autotrophic tissue engineering approaches and suggest the use of photosynthetic cells to treat a broad spectrum of hypoxic conditions.
Collapse
Affiliation(s)
- Thilo Ludwig Schenck
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany
| | - Ursula Hopfner
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany
| | - Myra Noemi Chávez
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany
| | - Hans-Günther Machens
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany
| | - Ian Somlai-Schweiger
- Department of Nuclear Medicine, University Hospital rechts der Isar, Technische Universität München, Germany
| | - Riccardo Enzo Giunta
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany; Handchirurgie, Plastische Chirurgie, Ästhetische Chirurgie der Ludwig-Maximilians Universität München, Germany
| | - Alexandra Viola Bohne
- Molekulare Pflanzenwissenschaften, Biozentrum Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Jörg Nickelsen
- Molekulare Pflanzenwissenschaften, Biozentrum Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile, Santiago, Chile
| | - José Tomás Egaña
- Department of Plastic Surgery and Hand Surgery, University Hospital rechts der Isar, Technische Universität München, Germany; FONDAP Center for Genome Regulation, Faculty of Science, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
215
|
Fu M, Li Z, Tan T, Guo W, Xie N, Liu Q, Zhu H, Xie X, Lei H. Akt/eNOS signaling pathway mediates inhibition of endothelial progenitor cells by palmitate-induced ceramide. Am J Physiol Heart Circ Physiol 2015; 308:H11-7. [PMID: 25380816 DOI: 10.1152/ajpheart.00503.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Palmitate (PA) impairs endothelial progenitor cells (EPCs). However, the molecular mechanism underlying the suppressive function of PA remains largely unknown. Ceramide, a free fatty acid metabolite, mediates multiple cellular signals. We hypothesized that ceramide acts as an intermediate molecule to mediate inhibition of EPCs by PA. We first demonstrated that PA could inhibit the attachment, migration, and tube formation of EPCs through suppression of the Akt/endothelial nitric oxide (NO) synthase (eNOS) signaling pathway. In addition, we observed that PA could induce ceramide accumulation in EPCs. To test whether the accumulation of ceramide causes EPC dysfunction, the ceramide synthesis inhibitors myriocin and fumonisin B1 were used. We that found both inhibitors could effectively abolish PA-mediated EPC inhibition. Furthermore, the ceramide deacylation inhibitor N-oleoylethanolamine could augment the inhibitory effect of PA on EPCs, indicating that it is ceramide, not its metabolites, that mediates the suppression of EPCs by PA. We have previously shown that Akt/eNOS phosphorylation was reduced after PA treatment, which, in turn, hampered the normal bioavailability of NO, leading to impaired functions of EPCs. To test the role for ceramide in this process, a clinically used NO donor, sodium nitroprusside, was used. We found that sodium nitroprusside could rescue the suppressive effects of ceramide on EPCs, suggesting that ceramide-mediated EPC inhibition might be through reduction of NO production. Taken together, our findings indicated that ceramide-induced reduction of NO might be the molecular mechanism for PA-mediated EPC inhibition; thus, targeting either ceramide or NO production might be an effective means for improvement of EPC functions in diseases.
Collapse
Affiliation(s)
- Minghuan Fu
- The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhihong Li
- Division of General Surgery, Chenzhou First People′s Hospital, Chenzhou, Hunan, China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Weixin Guo
- Guangdong Geriatrics Institute, Guangdong General Hospital, Guangdong Academy of Medical sciences, Guangzhou, China; and
| | - Nanzi Xie
- Division of Geriatrics, Tongji Hospital and Tongji University School of Medicine, Shanghai, China
| | - Qing Liu
- The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Xiaoyun Xie
- Division of Geriatrics, Tongji Hospital and Tongji University School of Medicine, Shanghai, China
| | - Han Lei
- The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
216
|
Spiliopoulos S, Kitrou P, Katsanos K, Karnabatidis D. Current Phase II drugs under investigation for the treatment of limb ischemia. Expert Opin Investig Drugs 2015; 24:1447-58. [PMID: 26296189 DOI: 10.1517/13543784.2015.1081894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION More than 20 million people in Europe suffer from peripheral arterial disease and nearly 3% develop critical limb ischemia (CLI). Without any medical treatment, CLI has poor prognosis, resulting in limb loss and high mortality rate. Until today, no systemic drug is available for the treatment of CLI and the gold standard method of treatment includes risk factor modification and open surgical or endovascular revascularization. Endovascular local drug delivery devices and novel antithrombotic agents, currently under investigation, aim to improve outcomes of revascularization procedures. The pioneering concept of therapeutic angiogenesis induced by gene and stem cell therapy has been proposed, in an attempt to increase ischemic tissue perfusion. AREAS COVERED This review summarizes local and systemic pharmacological treatment of CLI using endovascular or pharmaco-biological therapy and focuses on Phase II trials available for these drugs. EXPERT OPINION Novel endovascular technologies combining angioplasty and local drug-delivery continuously improve and will come to be standard of practice for the management of limb ischemia, while new antithrombotic agents will further improve outcomes. Therapeutic angiogenesis represents a safe and promising treatment option. The combination of revascularization with microcirculation improvement induced by gene or stem cell therapy could enhance limb salvage.
Collapse
Affiliation(s)
- Stavros Spiliopoulos
- a 1 Patras University Hospital, Department of Interventional Radiology , Patras 26504, Greece +30 2613 603 219;
| | - Panagiotis Kitrou
- a 1 Patras University Hospital, Department of Interventional Radiology , Patras 26504, Greece +30 2613 603 219;
| | - Konstantinos Katsanos
- b 2 Guy's and St Thomas' Hospitals, NHS Foundation Trust, Department of Interventional Radiology , London, UK
| | - Dimitris Karnabatidis
- a 1 Patras University Hospital, Department of Interventional Radiology , Patras 26504, Greece +30 2613 603 219;
| |
Collapse
|
217
|
Altabas V. Diabetes, Endothelial Dysfunction, and Vascular Repair: What Should a Diabetologist Keep His Eye on? Int J Endocrinol 2015; 2015:848272. [PMID: 26089898 PMCID: PMC4452196 DOI: 10.1155/2015/848272] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/13/2015] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular complications are the most common complications of diabetes mellitus. A prominent attribute of diabetic cardiovascular complications is accelerated atherosclerosis, considered as a still incurable disease, at least at more advanced stages. The discovery of endothelial progenitor cells (EPCs), able to replace old and injured mature endothelial cells and capable of differentiating into healthy and functional endothelial cells, has offered the prospect of merging the traditional theories on the pathogenesis of atherosclerosis with evolving concepts of vascular biology. The literature supports the notion that EPC alterations are involved in the pathogenesis of vascular diseases in diabetics, but at present many questions remain unanswered. In this review the aspects linking endothelial progenitor cells to the altered vascular biology in diabetes mellitus are discussed.
Collapse
Affiliation(s)
- V. Altabas
- Department for Endocrinology, Diabetes and Metabolic Diseases “Mladen Sekso”, Clinic for Internal Medicine, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
- *V. Altabas:
| |
Collapse
|
218
|
Prisco AR, Prisco MR, Carlson BE, Greene AS. TNF-α increases endothelial progenitor cell adhesion to the endothelium by increasing bond expression and affinity. Am J Physiol Heart Circ Physiol 2014; 308:H1368-81. [PMID: 25539711 DOI: 10.1152/ajpheart.00496.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022]
Abstract
Endothelial progenitor cells (EPCs) are a rare population of cells that participate in angiogenesis. To effectively use EPCs for regenerative therapy, the mechanisms by which they participate in tissue repair must be elucidated. This study focused on the process by which activated EPCs bind to a target tissue. It has been demonstrated that EPCs can bind to endothelial cells (ECs) through the tumore necrosis factor-α (TNF-α)-regulated vascular cell adhesion molecule 1/very-late antigen 4 (VLA4) interaction. VLA4 can bind in a high or low affinity state, a process that is difficult to experimentally isolate from bond expression upregulation. To separate these processes, a new parallel plate flow chamber was built, a detachment assay was developed, and a mathematical model was created that was designed to analyze the detachment assay results. The mathematical model was developed to predict the relative expression of EPC/EC bonds made for a given bond affinity distribution. EPCs treated with TNF-α/vehicle were allowed to bind to TNF-α/vehicle-treated ECs in vitro. Bound cells were subjected to laminar flow, and the cellular adherence was quantified as a function of shear stress. Experimental data were fit to the mathematical model using changes in bond expression or affinity as the only free parameter. It was found that TNF-α treatment of ECs increased adhesion through bond upregulation, whereas TNF-α treatment of EPCs increased adhesion by increasing bond affinity. These data suggest that injured tissue could potentially increase recruitment of EPCs for tissue regeneration via the secretion of TNF-α.
Collapse
Affiliation(s)
- Anthony R Prisco
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael R Prisco
- Exponent Engineering and Scientific Consulting, Biomedical Engineering Practice, Warrenville, Illinois; and
| | - Brian E Carlson
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Andrew S Greene
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin;
| |
Collapse
|
219
|
Qi P, Yan W, Yang Y, Li Y, Fan Y, Chen J, Yang Z, Tu Q, Huang N. Immobilization of DNA aptamers via plasma polymerized allylamine film to construct an endothelial progenitor cell-capture surface. Colloids Surf B Biointerfaces 2014; 126:70-9. [PMID: 25575347 DOI: 10.1016/j.colsurfb.2014.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/25/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023]
Abstract
The endothelial progenitor cells (EPCs) capture stent has drawn increasing attentions and become one of the most promising concepts for the next generation vascular stent. In this regard, it is of great significance to immobilize a molecule with the ability to bind EPC for rapid in vivo endothelialization with high specificity. In this work, a facile two-step method aimed at constructing a coating with specific EPC capturing aptamers is reported. The processes involves as the first-step deposition of plasma polymerized allylamine (PPAam) on a substrate to introduce amine groups, followed by the electrostatic adsorption of a 34 bases single strand DNA sequence to the PPAam surface as a second step (PPAam-DNA). Grazing incidence attenuated total reflection Fourier transform infrared spectroscopy (GATR-FTIR) and X-ray photoelectron spectroscopy (XPS) confirmed the successful immobilization of the aptamers. Quartz crystal microbalance with dissipation (QCM-D) real time monitoring result shows that about 175 ng/cm(2) aptamers were conjugated onto the PPAam surface. The interactions between the modified surfaces and human umbilical vein endothelial cells (ECs), smooth muscle cells (SMCs), and murine induced EPCs derived from mesenchymal stem cells (MSCs) were also investigated. It was demonstrated that PPAam-DNA samples could capture more EPCs, and present a cellular friendly surface for the proliferation of both EPCs and ECs but no effect on the hyperplasia of SMCs. Also, the co-culture results of 3 types of cells confirmed that the aptamer could specifically bond EPCs rather than ECs and SMCs, suggesting the competitive adhesion advantage of EPCs to ECs and SMCs. These data demonstrate that the EPC aptamer has large potential for designing an EPC captured stent and other vascular grafts with targeted in situ endothelialization.
Collapse
Affiliation(s)
- Pengkai Qi
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wei Yan
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Ying Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yalong Li
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; Laboratory of Biosensing and MicroMechatronics, Southwest Jiaotong University, Chengdu 610031, China
| | - Yi Fan
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Junying Chen
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Zhilu Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Qiufen Tu
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; Laboratory of Biosensing and MicroMechatronics, Southwest Jiaotong University, Chengdu 610031, China.
| | - Nan Huang
- Key Lab of Advanced Technology of Materials of Education Ministry, Southwest Jiaotong University, Chengdu 610031, China; School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
220
|
Abstract
Lymphedema is a medically irreversible condition for which currently conservative and surgical therapies are either ineffective or impractical. The potential use of progenitor and stem cell-based therapies has offered a paradigm that may provide alternative treatment options for lymphatic disorders. Moreover, basic research, preclinical studies, as well as clinical trials have evaluated the therapeutic potential of various cell therapies in the field of lymphatic regeneration medicine. Among the available cell approaches, mesenchymal stem cells (MSCs) seem to be the most promising candidate mainly due to their abundant sources and easy availability as well as evitable ethical and immunological issues confronted with embryonic stem cells and induced pluripotent stem cells. In this context, the purpose of this review is to summarize various cell-based therapies for lymphedema, along with strengths and weaknesses of these therapies in the clinical application for lymphedema treatment. Particularly, we will highlight the use of MSCs for lymphatic regeneration medicine. In addition, the future perspectives of MSCs in the field of lymphatic regeneration will be discussed.
Collapse
Affiliation(s)
- Shuqun Qi
- 1 State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University , Chengdu, China
| | | |
Collapse
|
221
|
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: 0.8] [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
|
222
|
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: 1.5] [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
|
223
|
G N, Tan A, Gundogan B, Farhatnia Y, Nayyer L, Mahdibeiraghdar S, Rajadas J, De Coppi P, Davies AH, Seifalian AM. Tissue engineering vascular grafts a fortiori: looking back and going forward. Expert Opin Biol Ther 2014; 15:231-44. [PMID: 25427995 DOI: 10.1517/14712598.2015.980234] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cardiovascular diseases such as coronary heart disease often necessitate the surgical repair using conduits. Although autografts still remain the gold standard, the inconvenience of harvesting and/or insufficient availability in patients with atherosclerotic disease has given impetus to look into alternative sources for vascular grafts. AREAS COVERED There are four main techniques to produce tissue-engineered vascular grafts (TEVGs): i) biodegradable synthetic scaffolds; ii) gel-based scaffolds; iii) decellularised scaffolds and iv) self-assembled cell-sheet-based techniques. The first three techniques can be grouped together as scaffold-guided approach as it involves the use of a construct to function as a supportive framework for the vascular graft. The most significant advantages of TEVGs are that it possesses the ability to grow, remodel and respond to environmental factors. Cell sources for TEVGs include mature somatic cells, stem cells, adult progenitor cells and pluripotent stem cells. EXPERT OPINION TEVG holds great promise with advances in nanotechnology, coupled with important refinements in tissue engineering and decellularisation techniques. This will undoubtedly be an important milestone for cardiovascular medicine when it is eventually translated to clinical use.
Collapse
Affiliation(s)
- Natasha G
- University College London (UCL), Centre for Nanotechnology and Regenerative Medicine, UCL Division of Surgery and Interventional Science, Research Department of Nanotechnology , London NW3 2QG , UK +44 207 830 2901 ;
| | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Rose JA, Erzurum S, Asosingh K. Biology and flow cytometry of proangiogenic hematopoietic progenitors cells. Cytometry A 2014; 87:5-19. [PMID: 25418030 DOI: 10.1002/cyto.a.22596] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/21/2014] [Accepted: 11/06/2014] [Indexed: 12/25/2022]
Abstract
During development, hematopoiesis and neovascularization are closely linked to each other via a common bipotent stem cell called the hemangioblast that gives rise to both hematopoietic cells and endothelial cells. In postnatal life, this functional connection between the vasculature and hematopoiesis is maintained by a subset of hematopoietic progenitor cells endowed with the capacity to differentiate into potent proangiogenic cells. These proangiogenic hematopoietic progenitors comprise a specific subset of bone marrow (BM)-derived cells that homes to sites of neovascularization and possess potent paracrine angiogenic activity. There is emerging evidence that this subpopulation of hematopoietic progenitors plays a critical role in vascular health and disease. Their angiogenic activity is distinct from putative "endothelial progenitor cells" that become structural cells of the endothelium by differentiation into endothelial cells. Proangiogenic hematopoietic progenitor cell research requires multidisciplinary expertise in flow cytometry, hematology, and vascular biology. This review provides a comprehensive overview of proangiogenic hematopoietic progenitor cell biology and flow cytometric methods to detect these cells in the peripheral blood circulation and BM.
Collapse
Affiliation(s)
- Jonathan A Rose
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | | | | |
Collapse
|
225
|
Sándor N, Walter FR, Bocsik A, Sántha P, Schilling-Tóth B, Léner V, Varga Z, Kahán Z, Deli MA, Sáfrány G, Hegyesi H. Low dose cranial irradiation-induced cerebrovascular damage is reversible in mice. PLoS One 2014; 9:e112397. [PMID: 25393626 PMCID: PMC4231057 DOI: 10.1371/journal.pone.0112397] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/03/2014] [Indexed: 11/21/2022] Open
Abstract
Background High-dose radiation-induced blood-brain barrier breakdown contributes to acute radiation toxicity syndrome and delayed brain injury, but there are few data on the effects of low dose cranial irradiation. Our goal was to measure blood-brain barrier changes after low (0.1 Gy), moderate (2 Gy) and high (10 Gy) dose irradiation under in vivo and in vitro conditions. Methodology Cranial irradiation was performed on 10-day-old and 10-week-old mice. Blood-brain barrier permeability for Evans blue, body weight and number of peripheral mononuclear and circulating endothelial progenitor cells were evaluated 1, 4 and 26 weeks postirradiation. Barrier properties of primary mouse brain endothelial cells co-cultured with glial cells were determined by measurement of resistance and permeability for marker molecules and staining for interendothelial junctions. Endothelial senescence was determined by senescence associated β-galactosidase staining. Principle Findings Extravasation of Evans blue increased in cerebrum and cerebellum in adult mice 1 week and in infant mice 4 weeks postirradiation at all treatment doses. Head irradiation with 10 Gy decreased body weight. The number of circulating endothelial progenitor cells in blood was decreased 1 day after irradiation with 0.1 and 2 Gy. Increase in the permeability of cultured brain endothelial monolayers for fluorescein and albumin was time- and radiation dose dependent and accompanied by changes in junctional immunostaining for claudin-5, ZO-1 and β-catenin. The number of cultured brain endothelial and glial cells decreased from third day of postirradiation and senescence in endothelial cells increased at 2 and 10 Gy. Conclusion Not only high but low and moderate doses of cranial irradiation increase permeability of cerebral vessels in mice, but this effect is reversible by 6 months. In-vitro experiments suggest that irradiation changes junctional morphology, decreases cell number and causes senescence in brain endothelial cells.
Collapse
Affiliation(s)
- Nikolett Sándor
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Doctoral Schools of Pathological Sciences, Semmelweis University, Budapest, Hungary
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Alexandra Bocsik
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Petra Sántha
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Boglárka Schilling-Tóth
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Violetta Léner
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Care, Semmelweis University, Budapest, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Mária A. Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Géza Sáfrány
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Hargita Hegyesi
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Care, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
226
|
Yang D, Zhang JN, Zhou HF. Endothelial progenitor cells in patients with age-related hearing loss. Am J Otolaryngol 2014; 35:695-8. [PMID: 25212104 DOI: 10.1016/j.amjoto.2014.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/04/2014] [Accepted: 08/14/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE This study was conducted to determine the concentration of EPCs in patients with ARHL. METHODS Twenty patients with ARHL were evaluated. The number of EPCs was analyzed by flow cytometry analysis of peripheral blood CD34(+)/CD133(+) cells. RESULTS The concentration of circulating EPCs, both for CD34(+)/CD133(+) cells, was significantly lower in ARHL patients compared to controls (P<0.05). No statistically significant differences were found between these two groups in terms of the level of total cholesterol, LDL, HDL, triglycerides and GLU. CONCLUSIONS The possible role of circulating epithelial progenitor cells in the pathogenesis of age related hearing loss should be considered based on their significant reduction in patients with ARHL, although the association alone does not prove causality. Further studies were warranted to confirm the role of circulating EPCs in the pathogenesis of ARHL.
Collapse
|
227
|
Jeong HS, Lee HK, Tark KC, Lew DH, Koh YW, Kim CH, Seo IS. Effect of endogenous bone marrow derived stem cells induced by AMD-3100 on expanded ischemic flap. J Korean Med Sci 2014; 29 Suppl 3:S237-48. [PMID: 25473215 PMCID: PMC4248011 DOI: 10.3346/jkms.2014.29.s3.s237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/17/2014] [Indexed: 11/20/2022] Open
Abstract
The purpose of this study was to devise an expanded ischemic flap model and to investigate the role of AMD-3100 (Plerixafor, chemokine receptor 4 inhibitor) in this model by confirming its effect on mobilization of stem cells from the bone marrow. Male Sprague-Dawley rats were used as an animal research model. The mobilization of stem cells from the bone marrow was confirmed in the AMD-3100-treated group. The fractions of endothelial progenitor cells (EPC) and the vascular endothelial growth factor receptor (VEGFR) 2+ cells in the peripheral blood were increased in groups treated with AMD-3100. The expression of vascular endothelial growth factor (VEGF) was increased in response to expansion or AMD injection. The expression of stromal cell derived factor (SDF)-1 and VEGFR2 were increased only in unexpanded flap treated with AMD-3100. Treatment with AMD-3100 increased both the number and area of blood vessels. However, there were no statistically significant differences in the survival area or physiologic microcirculation in rats from the other groups. This endogenous neovascularization induced by AMD-3100 may be a result of the increase in both the area and number of vessels, as well as paracrine augmentation of the expression of VEGF and EPCs. However, the presence of a tissue expander under the flap could block the neovascularization between the flap and the recipient regardless of AMD-3100 treatment and expansion.
Collapse
Affiliation(s)
- Hii-Sun Jeong
- Department of Plastic & Reconstructive Surgery, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Halllym University College of Medicine, Seoul, Korea
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Kyung Lee
- Department of Plastic and Reconstructive Surgery, Eulji General Hospital, Eulji University School of Medicine, Seoul, Korea
| | - Kwan-Chul Tark
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Dae-Hyun Lew
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon-Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| | - Chul-Hoon Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | - In-Suck Seo
- Department of Plastic & Reconstructive Surgery, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Halllym University College of Medicine, Seoul, Korea
| |
Collapse
|
228
|
May the remodeling of the Ca²⁺ toolkit in endothelial progenitor cells derived from cancer patients suggest alternative targets for anti-angiogenic treatment? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1958-73. [PMID: 25447551 DOI: 10.1016/j.bbamcr.2014.10.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/16/2014] [Accepted: 10/28/2014] [Indexed: 01/10/2023]
Abstract
Endothelial progenitor cells (EPCs) may be recruited from bone marrow to sustain the metastatic switch in a number of solid cancers, including breast cancer (BC) and renal cellular carcinoma (RCC). Preventing EPC mobilization causes tumor shrinkage. Novel anti-angiogenic treatments have been introduced in therapy to inhibit VEGFR-2 signaling; unfortunately, these drugs blocked tumor angiogenesis in pre-clinical murine models, but resulted far less effective in human patients. Understanding the molecular mechanisms driving EPC proliferation and tubulogenesis in cancer patients could outline novel targets for alternative anti-angiogenic treatments. Store-operated Ca²⁺ entry (SOCE) regulates the growth of human EPCs, and it is mediated by the interaction between the endoplasmic reticulum Ca²⁺-sensor, Stim1, and the plasmalemmal Ca²⁺ channels, Orai1 and TRPC1. EPCs do not belong to the neoplastic clone: thus, unlike tumor endothelium and neoplastic cells, they should not remodel their Ca²⁺ toolkit in response to tumor microenvironment. However, our recent work demonstrated that EPCs isolated from naïve RCC patients (RCC-EPCs) undergo a dramatic remodeling of their Ca²⁺ toolkit by displaying a remarkable drop in the endoplasmic reticulum Ca²⁺ content, by down-regulating the expression of inositol-1,4,5-receptors (InsP3Rs), and by up-regulating Stim1, Orai1 and TRPC1. Moreover, EPCs are dramatically less sensitive to VEGF stimulation both in terms of Ca²⁺ signaling and of gene expression when isolated from tumor patients. Conversely, the pharmacological abolition of SOCE suppresses proliferation in these cells. These results question the suitability of VEGFR-2 as a therapeutically relevant target for anti-angiogenic treatments and hint at Orai1 and TRPC1 as more promising alternatives. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
Collapse
|
229
|
Piatkowski A, Grieb G, Simons D, Bernhagen J, van der Hulst RR. Endothelial progenitor cells--potential new avenues to improve neoangiogenesis and reendothelialization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 306:43-81. [PMID: 24016523 DOI: 10.1016/b978-0-12-407694-5.00002-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term endothelial progenitor cell (EPC) was established more than 10 years ago and is used to refer to a group of circulating cells that display endothelial lineage qualities and are able to home to areas of ischemia or vascular injury and to facilitate the repair of damaged blood vessels or develop new vessels as needed. This chapter reviews the current lineage relationships among all the cells called EPC and will clear the terminology used in EPC research. Furthermore, an overview of the clinical and in vitro research, as well as cytokine and drug interactions and potential EPC applications, is given.
Collapse
Affiliation(s)
- Andrzej Piatkowski
- Department of Plastic Surgery, academisch ziekenhuis Maastricht, MUMC+, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
230
|
Kadomatsu K, Bencsik P, Görbe A, Csonka C, Sakamoto K, Kishida S, Ferdinandy P. Therapeutic potential of midkine in cardiovascular disease. Br J Pharmacol 2014; 171:936-44. [PMID: 24286213 DOI: 10.1111/bph.12537] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/12/2013] [Accepted: 11/20/2013] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Ischaemic heart disease, stroke and their pathological consequences are life-threatening conditions that account for about half of deaths in developed countries. Pathology of these diseases includes cell death due to ischaemia/reperfusion injury, vascular stenosis and cardiac remodelling. The growth factor midkine plays a pivotal role in these events. Midkine shows an acute cytoprotective effect in ischaemia/reperfusion injury at least in part via its anti-apoptotic effect. Moreover, while midkine promotes endothelial cell proliferation, it also recruits inflammatory cells to lesions. These activities eventually enhance angiogenesis, thereby preventing cardiac tissue remodelling. However, midkine's activity in recruiting inflammatory cells into the vascular wall also triggers neointima formation, and consequently, vascular stenosis. Moreover, midkine is induced in cancer tissues where it enhances angiogenesis. Therefore, midkine may promote tumour formation through its angiogenic and anti-apoptotic activity. This review focuses on the roles of midkine in ischaemic cardiovascular disease and their pathological consequences, that is angiogenesis, vascular stenosis, and cardiac remodelling, and discusses the possible therapeutic potential of modulation of midkine in these diseases. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
231
|
Widemann A, Pasero C, Arnaud L, Poullin P, Loundou AD, Choukroun G, Sanderson F, Lacroix R, Sabatier F, Coppo P, Dignat-George F, Kaplanski G. Circulating endothelial cells and progenitors as prognostic factors during autoimmune thrombotic thrombocytopenic purpura: results of a prospective multicenter French study. J Thromb Haemost 2014; 12:1601-9. [PMID: 25088020 DOI: 10.1111/jth.12681] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/06/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND Autoimmune thrombotic thrombocytopenic purpura (AI-TTP) is characterized by an excess of circulating ultralarge von Willebrand factor (VWF) caused by anti-ADAMTS-13 autoantibodies. Animal studies, however, have shown that endothelial cell activation may also be an important trigger of AI-TTP. OBJECTIVES To prospectively study circulating biomarkers of endothelial lesion and activation, such as circulating endothelial cells (CECs), soluble P-selectin (sP-selectin), or VWF, and of endothelial repair, such as circulating progenitor cells (CPCs) and endothelial progenitor cells (EPCs), in AI-TTP, in relation to disease severity and prognosis. RESULTS Twenty-two patients were included in this study. CEC (P < 0.01), VWF (P < 0.05) and sP-selectin (P < 0.01) levels were significantly increased during crisis, and returned to baseline levels during remission. Both CEC (P < 0.05) and sP-selectin (P < 0.05) levels were significantly higher in patients who died or developed neurologic sequelae. CPC levels were substantially increased during the acute phase of the disease (P < 0.001), and returned to baseline levels during remission. Among CPCs, EPC levels were also increased during crisis (P < 0.05) and significantly decreased during remission. Patients who received < 16 plasma exchanges (PEs) had significantly higher EPC counts (P < 0.05) than those who needed more numerous PEs to obtain remission, suggesting that initial EPC counts may be associated with faster endothelial repair. CONCLUSION The profile of circulating endothelial markers shows massive endothelial activation and repair/remodeling during AI-TTP, and suggests that CECs and EPCs may be promising prognostic biomarkers of the disease.
Collapse
Affiliation(s)
- A Widemann
- Vascular Research Center of Marseille, Inserm, UMRS_1076, Aix-Marseille Université, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Jang IH, Heo SC, Kwon YW, Choi EJ, Kim JH. Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis. Adv Biol Regul 2014; 57:162-72. [PMID: 25304660 DOI: 10.1016/j.jbior.2014.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/03/2014] [Indexed: 12/30/2022]
Abstract
Endothelial progenitor cells (EPCs) hold a great promise as a therapeutic mediator in treatment of ischemic disease conditions. The discovery of EPCs in adult blood has been a cause of significant enthusiasm in the field of endothelial cell research and numerous clinical trials have been expedited. After more than a decade of research in basic science and clinical applications, limitations and new strategies of EPC therapeutics have emerged. With various phenotypes, vague definitions, and uncertain distinction from hematopoietic cells, understanding EPC biology remains challenging. However, EPCs, still hold great hope for treatment of critical ischemic injury as low concern regarding safety can accelerate the clinical applications from basic findings. This review provides an introduction to EPC as cellular therapeutics, which highlights a recent finding that EPC homing was promoted through FPR2 signaling.
Collapse
Affiliation(s)
- Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Soon Chul Heo
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Yang Woo Kwon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Eun Jung Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea; Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 626-770, Gyeongsangnam-do, Republic of Korea.
| |
Collapse
|
233
|
Expansion of endothelial progenitor cells in high density dot culture of rat bone marrow cells. PLoS One 2014; 9:e107127. [PMID: 25254487 PMCID: PMC4177845 DOI: 10.1371/journal.pone.0107127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 08/09/2014] [Indexed: 02/04/2023] Open
Abstract
In vitro expansion of endothelial progenitor cells (EPCs) remains a challenge in stem cell research and its application. We hypothesize that high density culture is able to expand EPCs from bone marrow by mimicking cell-cell interactions of the bone marrow niche. To test the hypothesis, rat bone marrow cells were either cultured in high density (2×105 cells/cm2) by seeding total 9×105 cells into six high density dots or cultured in regular density (1.6×104 cells/cm2) with the same total number of cells. Flow cytometric analyses of the cells cultured for 15 days showed that high density cells exhibited smaller cell size and higher levels of marker expression related to EPCs when compared to regular density cultured cells. Functionally, these cells exhibited strong angiogenic potentials with better tubal formation in vitro and potent rescue of mouse ischemic limbs in vivo with their integration into neo-capillary structure. Global gene chip and ELISA analyses revealed up-regulated gene expression of adhesion molecules and enhanced protein release of pro-angiogenic growth factors in high density cultured cells. In summary, high density cell culture promotes expansion of bone marrow contained EPCs that are able to enhance tissue angiogenesis via paracrine growth factors and direct differentiation into endothelial cells.
Collapse
|
234
|
Lin RZ, Hatch A, Antontsev VG, Murthy SK, Melero-Martin JM. Microfluidic capture of endothelial colony-forming cells from human adult peripheral blood: phenotypic and functional validation in vivo. Tissue Eng Part C Methods 2014; 21:274-83. [PMID: 25091645 DOI: 10.1089/ten.tec.2014.0323] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Endothelial colony-forming cells (ECFCs) are endothelial progenitors that circulate in peripheral blood and are currently the subject of intensive investigation due to their therapeutic potential. However, in adults, ECFCs comprise a very small subset among circulating cells, which makes their isolation a challenge. MATERIALS AND METHODS Currently, the standard method for ECFC isolation relies on the separation of mononuclear cells and erythrocyte lysis, steps that are time consuming and known to increase cell loss. Alternatively, we previously developed a novel disposable microfluidic platform containing antibody-functionalized degradable hydrogel coatings that is ideally suited for capturing low-abundance circulating cells from unprocessed blood. In this study, we reasoned that this microfluidic approach could effectively isolate rare ECFCs by virtue of their CD34 expression. RESULTS We conducted preclinical experiments with peripheral blood from four adult volunteers and demonstrated that the actual microfluidic capture of circulating CD34(+) cells from unprocessed blood was compatible with the subsequent differentiation of these cells into ECFCs. Moreover, the ECFC yield obtained with the microfluidic system was comparable to that of the standard method. Importantly, we unequivocally validated the phenotypical and functional properties of the captured ECFCs, including the ability to form microvascular networks following transplantation into immunodeficient mice. DISCUSSION We showed that the simplicity and versatility of our microfluidic system could be very instrumental for ECFC isolation while preserving their therapeutic potential. We anticipate our results will facilitate additional development of clinically suitable microfluidic devices by the vascular therapeutic and diagnostic industry.
Collapse
Affiliation(s)
- Ruei-Zeng Lin
- 1 Department of Cardiac Surgery, Boston Children's Hospital , Boston, Massachusetts
| | | | | | | | | |
Collapse
|
235
|
Neels JG, Grimaldi PA. Physiological functions of peroxisome proliferator-activated receptor β. Physiol Rev 2014; 94:795-858. [PMID: 24987006 DOI: 10.1152/physrev.00027.2013] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptors, PPARα, PPARβ, and PPARγ, are a family of transcription factors activated by a diversity of molecules including fatty acids and fatty acid metabolites. PPARs regulate the transcription of a large variety of genes implicated in metabolism, inflammation, proliferation, and differentiation in different cell types. These transcriptional regulations involve both direct transactivation and interaction with other transcriptional regulatory pathways. The functions of PPARα and PPARγ have been extensively documented mainly because these isoforms are activated by molecules clinically used as hypolipidemic and antidiabetic compounds. The physiological functions of PPARβ remained for a while less investigated, but the finding that specific synthetic agonists exert beneficial actions in obese subjects uplifted the studies aimed to elucidate the roles of this PPAR isoform. Intensive work based on pharmacological and genetic approaches and on the use of both in vitro and in vivo models has considerably improved our knowledge on the physiological roles of PPARβ in various cell types. This review will summarize the accumulated evidence for the implication of PPARβ in the regulation of development, metabolism, and inflammation in several tissues, including skeletal muscle, heart, skin, and intestine. Some of these findings indicate that pharmacological activation of PPARβ could be envisioned as a therapeutic option for the correction of metabolic disorders and a variety of inflammatory conditions. However, other experimental data suggesting that activation of PPARβ could result in serious adverse effects, such as carcinogenesis and psoriasis, raise concerns about the clinical use of potent PPARβ agonists.
Collapse
Affiliation(s)
- Jaap G Neels
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| | - Paul A Grimaldi
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| |
Collapse
|
236
|
López-Ruiz E, Perán M, Picón-Ruiz M, García MA, Carrillo E, Jiménez-Navarro M, Hernández MC, Prat I, De Teresa E, Marchal JA. Cardiomyogenic differentiation potential of human endothelial progenitor cells isolated from patients with myocardial infarction. Cytotherapy 2014; 16:1229-37. [DOI: 10.1016/j.jcyt.2014.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/09/2014] [Accepted: 05/12/2014] [Indexed: 11/16/2022]
|
237
|
Kwon SM, Lee JH, Lee SH, Jung SY, Kim DY, Kang SH, Yoo SY, Hong JK, Park JH, Kim JH, Kim SW, Kim YJ, Lee SJ, Kim HG, Asahara T. Cross talk with hematopoietic cells regulates the endothelial progenitor cell differentiation of CD34 positive cells. PLoS One 2014; 9:e106310. [PMID: 25166961 PMCID: PMC4148437 DOI: 10.1371/journal.pone.0106310] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Despite the crucial role of endothelial progenitor cells (EPCs) in vascular regeneration, the specific interactions between EPCs and hematopoietic cells remain unclear. METHODS In EPC colony forming assays, we first demonstrated that the formation of EPC colonies was drastically increased in the coculture of CD34+ and CD34- cells, and determined the optimal concentrations of CD34+ cells and CD34- cells for spindle-shaped EPC differentiation. RESULTS Functionally, the coculture of CD34+ and CD34- cells resulted in a significant enhancement of adhesion, tube formation, and migration capacity compared with culture of CD34+ cells alone. Furthermore, blood flow recovery and capillary formation were remarkably increased by the coculture of CD34+ and CD34- cells in a murine hind-limb ischemia model. To elucidate further the role of hematopoietic cells in EPC differentiation, we isolated different populations of hematopoietic cells. T lymphocytes (CD3+) markedly accelerated the early EPC status of CD34+ cells, while macrophages (CD11b+) or megakaryocytes (CD41+) specifically promoted large EPC colonies. CONCLUSION Our results suggest that specific populations of hematopoietic cells play a role in the EPC differentiation of CD34+ cells, a finding that may aid in the development of a novel cell therapy strategy to overcome the quantitative and qualitative limitations of EPC therapy.
Collapse
Affiliation(s)
- Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
- * E-mail: (SMK); (TA)
| | - Jun-Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sang-Hun Lee
- Soonchunhyang Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Seok-Yun Jung
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Da-Yeon Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Song-Hwa Kang
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - So-Young Yoo
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jong-Kyu Hong
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Ji-Hye Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jung-Hee Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sung-Wook Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yeon-Ju Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Sun-Jin Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Hwi-Gon Kim
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Takayuki Asahara
- Department Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
- * E-mail: (SMK); (TA)
| |
Collapse
|
238
|
Yunt ZX, Mohning MP, Barthel L, Kearns MT, Tuder RM, Hyde DM, Henson PM, Janssen WJ. Kinetics of the angiogenic response in lung endothelium following acute inflammatory injury with bleomycin. Exp Lung Res 2014; 40:415-25. [PMID: 25153689 DOI: 10.3109/01902148.2014.938202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE/AIM Angiogenesis is a central component of normal wound healing but it has not been fully characterized in lung repair following acute inflammatory injury. The current literature lacks vital information pertaining to the extent, timing, and location of this process. This information is necessary for examining mechanisms that drive normal lung repair in resolving acute inflammatory injury. The goal of our study was to formally characterize lung angiogenesis over a time course of bleomycin-induced lung injury. MATERIALS AND METHODS Female C57BL/6 mice age 8-12 weeks were treated with a single dose of intratracheal bleomycin. Total lung endothelial cells were quantified with flow cytometry 0, 7, 14, 21, and 28 days following bleomycin administration, and endothelial cell replication was assessed using bromodeoxyuridine (BrdU) incorporation. RESULTS Endothelial cell replication was maximal 14 days after bleomycin administration, while total lung endothelial cells peaked at day 21. Tissue analysis with stereology was performed to measure total lung vascular surface area in bleomycin at day 21 relative to controls and demonstrated a trend toward increased vasculature in the bleomycin group. CONCLUSIONS Angiogenesis begins shortly after injury in the bleomycin model and leads to an expansion in the lung endothelial cell population that peaks at day 21. This study offers the first longitudinal examination of angiogenesis following acute inflammatory lung injury induced by bleomycin. Information provided in this study will be vital for further investigating mechanisms of angiogenesis in both normal and abnormal lung repair.
Collapse
|
239
|
Fadini GP, Ferraro F, Quaini F, Asahara T, Madeddu P. Concise review: diabetes, the bone marrow niche, and impaired vascular regeneration. Stem Cells Transl Med 2014; 3:949-57. [PMID: 24944206 PMCID: PMC4116251 DOI: 10.5966/sctm.2014-0052] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/15/2014] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is a global health problem that results in multiorgan complications leading to high morbidity and mortality. Until recently, the effects of diabetes and hyperglycemia on the bone marrow microenvironment-a site where multiple organ systems converge and communicate-have been underappreciated. However, several new studies in mice, rats, and humans reveal that diabetes leads to multiple bone marrow microenvironmental defects, such as small vessel disease (microangiopathy), nerve terminal pauperization (neuropathy), and impaired stem cell mobilization (mobilopathy). The discovery that diabetes involves bone marrow-derived progenitors implicated in maintaining cardiovascular homeostasis has been proposed as a bridging mechanism between micro- and macroangiopathy in distant organs. Herein, we review the physiological and molecular bone marrow abnormalities associated with diabetes and discuss how bone marrow dysfunction represents a potential root for the development of the multiorgan failure characteristic of advanced diabetes. The notion of diabetes as a bone marrow and stem cell disease opens new avenues for therapeutic interventions ultimately aimed at improving the outcome of diabetic patients.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Francesca Ferraro
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Federico Quaini
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Takayuki Asahara
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
240
|
Lin LY, Huang CC, Chen JS, Wu TC, Leu HB, Huang PH, Chang TT, Lin SJ, Chen JW. Effects of pitavastatin versus atorvastatin on the peripheral endothelial progenitor cells and vascular endothelial growth factor in high-risk patients: a pilot prospective, double-blind, randomized study. Cardiovasc Diabetol 2014; 13:111. [PMID: 25027585 PMCID: PMC4223413 DOI: 10.1186/s12933-014-0111-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 07/07/2014] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Circulating endothelial progenitor cells (EPCs) reflect endothelial repair capacity and may be a significant marker for the clinical outcomes of cardiovascular disease. While some high-dose statin treatments may improve endothelial function, it is not known whether different statins may have similar effects on EPCs.This study aimed to investigate the potential class effects of different statin treatment including pitavastatin and atorvastatin on circulating EPCs in clinical setting. METHODS A pilot prospective, double-blind, randomized study was conducted to evaluate the ordinary dose of pitavastatin (2 mg daily) or atorvastatin (10 mg daily) treatment for 12 weeks on circulating EPCs in patients with cardiovascular risk such as hypercholesterolemia and type 2 diabetes mellitus (T2DM). Additional in vitro study was conducted to clarify the direct effects of both statins on EPCs from the patients. RESULTS A total of 26 patients (19 with T2DM) completed the study. While the lipid-lowering effects were similar in both treatments, the counts of circulating CD34+KDR+EPCs were significantly increased (from 0.021 ± 0.015 to 0.054 ± 0.044% of gated mononuclear cells, P < 0.05) only by pitavastatin treatment. Besides, plasma asymmetric dimethylarginine level was reduced (from 0.68 ± 0.10 to 0.53 ± 0.12 μmol/L, P < 0.05) by atorvastatin, and plasma vascular endothelial growth factor (VEGF) level was increased (from 74.33 ± 32.26 to 98.65 ± 46.64 pg/mL, P < 0.05) by pitavastatin. In the in vitro study, while both statins increased endothelial nitric oxide synthase (eNOS) expression, only pitavastatin increased the phosphorylation of eNOS in EPCs. Pitavastatin but not atorvastatin ameliorated the adhesion ability of early EPCs and the migration and tube formation capacities of late EPCs. CONCLUSIONS While both statins similarly reduced plasma lipids, only pitavastatin increased plasma VEGF level and circulating EPCs in high-risk patients, which is probably related to the differential pleiotropic effects of different statins. TRIAL REGISTRATION This trial is registered at ClinicalTrials.gov, NCT01386853.
Collapse
|
241
|
Bone marrow-derived mesenchymal stromal cells improve vascular regeneration and reduce leukocyte-endothelium activation in critical ischemic murine skin in a dose-dependent manner. Cytotherapy 2014; 16:1345-60. [PMID: 24972742 DOI: 10.1016/j.jcyt.2014.05.008] [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: 01/16/2014] [Revised: 04/26/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Stem cells participate in vascular regeneration following critical ischemia. However, their angiogenic and remodeling properties, as well as their role in ischemia-related endothelial leukocyte activation, need to be further elucidated. Herein, we investigated the effect of bone marrow-derived mesenchymal stromal cells (BM-MSCs) in a critically ischemic murine skin flap model. METHODS Groups received either 1 × 10(5), 5 × 10(5), or 1 × 10(6) BM-MSCs or cell-free conditioned medium (CM). Controls received sodium chloride. Intravital fluorescence microscopy was performed for morphological and quantitative assessment of micro-hemodynamic parameters over 12 days. RESULTS Tortuosity and diameter of conduit-arterioles were pronounced in the MSC groups (P < 0.01), whereas vasodilation was shifted to the end arteriolar level in the CM group (P < 0.01). These effects were accompanied by angiopoietin-2 expression. Functional capillary density and red blood cell velocity were enhanced in all treatment groups (P < 0.01). Although a significant reduction of rolling and sticking leukocytes was observed in the MSC groups with a reduction of diameter in postcapillary venules (P < 0.01), animals receiving CM exhibited a leukocyte-endothelium interaction similar to controls. This correlated with leukocyte common antigen expression in tissue sections (P < 0.01) and p38 mitogen-activated protein kinase expression from tissue samples. Cytokine analysis from BM-MSC culture medium revealed a 50% reduction of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, IL-12, tumor necrosis factor-α, interferon-γ) and chemokines (keratinocyte chemoattractant, granulocyte colony-stimulating factor) under hypoxic conditions. DISCUSSION We demonstrated positive effects of BM-MSCs on vascular regeneration and modulation of endothelial leukocyte adhesion in critical ischemic skin. The improvements after MSC application were dose-dependent and superior to the use of CM alone.
Collapse
|
242
|
Parri M, Pietrovito L, Grandi A, Campagnoli S, De Camilli E, Bianchini F, Marchiò S, Bussolino F, Jin B, Sarmientos P, Grandi G, Viale G, Pileri P, Chiarugi P, Grifantini R. Angiopoietin-like 7, a novel pro-angiogenetic factor over-expressed in cancer. Angiogenesis 2014; 17:881-96. [PMID: 24903490 DOI: 10.1007/s10456-014-9435-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/22/2014] [Indexed: 12/21/2022]
Abstract
Angiopoietin-like (ANGPTL) proteins are secreted proteins showing structural similarity to members of the angiopoietin family. Some ANGPTL proteins possess pleiotropic activities, being involved in cancer lipid, glucose energy metabolisms, and angiogenesis. ANGPTL7 is the less characterized member of the family whose functional role is only marginally known. In this study, we provide experimental evidences that ANGPTL7 is over-expressed in different human cancers. To understand the role played by ANGPTL7 in tumor biology, we asked whether ANGPTL7 is endogenously expressed by malignant cells or in response to environmental stimuli. We found that ANGPTL7 is marginally expressed under standard growth condition while it is specifically up-regulated by hypoxia. Interestingly, the protein is secreted and partially associated with the exosomal fraction, suggesting that it could be found in the systemic circulation of oncologic patients and act in an endocrine way. Moreover, we found that ANGPTL7 exerts a pro-angiogenetic effect on human differentiated endothelial cells by stimulating their proliferation, motility, invasiveness, and capability to form capillary-like networks while it does not stimulate progenitor endothelial cells. Finally, we showed that ANGPTL7 promotes vascularization in vivo in the mouse Matrigel sponge assay, thereby accrediting this molecule as a pro-angiogenic factor.
Collapse
Affiliation(s)
- Matteo Parri
- Externautics SpA, Via Fiorentina 1, 53100, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Sahara M, Hansson EM, Wernet O, Lui KO, Später D, Chien KR. Manipulation of a VEGF-Notch signaling circuit drives formation of functional vascular endothelial progenitors from human pluripotent stem cells. Cell Res 2014; 24:820-41. [PMID: 24810299 PMCID: PMC4085760 DOI: 10.1038/cr.2014.59] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/10/2014] [Accepted: 03/31/2014] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial lineage cells constitutes a promising source for therapeutic revascularization, but progress in this arena has been hampered by a lack of clinically-scalable differentiation protocols and inefficient formation of a functional vessel network integrating with the host circulation upon transplantation. Using a human embryonic stem cell reporter cell line, where green fluorescent protein expression is driven by an endothelial cell-specific VE-cadherin (VEC) promoter, we screened for > 60 bioactive small molecules that would promote endothelial differentiation, and found that administration of BMP4 and a GSK-3β inhibitor in an early phase and treatment with VEGF-A and inhibition of the Notch signaling pathway in a later phase led to efficient differentiation of hPSCs to the endothelial lineage within six days. This sequential approach generated > 50% conversion of hPSCs to endothelial cells (ECs), specifically VEC+CD31+CD34+CD14−KDRhigh endothelial progenitors (EPs) that exhibited higher angiogenic and clonogenic proliferation potential among endothelial lineage cells. Pharmaceutical inhibition or genetical knockdown of Notch signaling, in combination with VEGF-A treatment, resulted in efficient formation of EPs via KDR+ mesodermal precursors and blockade of the conversion of EPs to mature ECs. The generated EPs successfully formed functional capillary vessels in vivo with anastomosis to the host vessels when transplanted into immunocompromised mice. Manipulation of this VEGF-A-Notch signaling circuit in our protocol leads to rapid large-scale production of the hPSC-derived EPs by 12- to 20-fold vs current methods, which may serve as an attractive cell population for regenerative vascularization with superior vessel forming capability compared to mature ECs.
Collapse
Affiliation(s)
- Makoto Sahara
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA [4] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Emil M Hansson
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Oliver Wernet
- Department of Anesthesiology and Intensive Care Medicine, Charité-University Medicine Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Kathy O Lui
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Daniela Später
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Kenneth R Chien
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
244
|
Cervio M, Scudeller L, Viarengo G, Monti M, Del Fante C, Arici V, Perotti C. γ-Irradiated cord blood MNCs: different paracrine effects on mature and progenitor endothelial cells. Microvasc Res 2014; 94:9-16. [PMID: 24788073 DOI: 10.1016/j.mvr.2014.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/16/2014] [Accepted: 04/19/2014] [Indexed: 11/18/2022]
Abstract
Cell-based therapies have been employed to promote neovascularization mainly through the release of paracrine factors inhibiting apoptosis and supporting migration and proliferation of resident differentiated cells. We tested in vitro pro-angiogenic effects of apoptotic cord blood-derived mononuclear cells (CB-MNCs) and their conditioned medium (CM) on mature endothelial cells (HUVECs) and peripheral blood-derived endothelial progenitor cells (ECFCs). CB-MNCs were γ-irradiated to induce apoptosis and cultured for 72 h to obtain the release of CM. MNCs viability, evaluated by flow cytometry, decreased progressively after γ-irradiation reaching 41% at 72 h. γ-Irradiated MNCs (γMNCs) released increasing amounts of EGF, PDGF-AB and VEGF in their CM over time, as assessed by ELISA. γ-MNCs and their CM enhanced capillary-like network formation (in a dose-dependent and time-persistent manner), proliferation and migration of HUVECs in vitro, while they primed capillary-like network formation (dose-independent and not time-persistent) and induced migration but did not support proliferation of ECFCs. Our data support the hypothesis of paracrine mechanism as prevalent in regenerative medicine and demonstrate the efficacy of MNCs secretome in inducing neovascularization. To our knowledge, this is the first paper highlighting differential pro-angiogenic effects of CM on mature and progenitor endothelial cells, adding a tile in the understanding of mechanisms involved in neovascularization.
Collapse
Affiliation(s)
- Marila Cervio
- Immunohematology and Transfusion Service, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy.
| | - Luigia Scudeller
- Service of Biometry and Statistics, Scientific Direction, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Gianluca Viarengo
- Immunohematology and Transfusion Service, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Manuela Monti
- Research Center for Regenerative Medicine, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Claudia Del Fante
- Immunohematology and Transfusion Service, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Vittorio Arici
- Vascular Surgery Unit, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Cesare Perotti
- Immunohematology and Transfusion Service, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
245
|
Di Santo S, Seiler S, Fuchs AL, Staudigl J, Widmer HR. The secretome of endothelial progenitor cells promotes brain endothelial cell activity through PI3-kinase and MAP-kinase. PLoS One 2014; 9:e95731. [PMID: 24755675 PMCID: PMC3995762 DOI: 10.1371/journal.pone.0095731] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/31/2014] [Indexed: 11/19/2022] Open
Abstract
Background Angiogenesis and vascular remodelling are crucial events in tissue repair mechanisms promoted by cell transplantation. Current evidence underscores the importance of the soluble factors secreted by stem cells in tissue regeneration. In the present study we investigated the effects of paracrine factors derived from cultured endothelial progenitor cells (EPC) on rat brain endothelial cell properties and addressed the signaling pathways involved. Methods Endothelial cells derived from rat brain (rBCEC4) were incubated with EPC-derived conditioned medium (EPC-CM). The angiogenic response of rBCEC4 to EPC-CM was assessed as effect on cell number, migration and tubular network formation. In addition, we have compared the outcome of the in vitro experiments with the effects on capillary sprouting from rat aortic rings. The specific PI3K/AKT inhibitor LY294002 and the MEK/ERK inhibitor PD98059 were used to study the involvement of these two signaling pathways in the transduction of the angiogenic effects of EPC-CM. Results Viable cell number, migration and tubule network formation were significantly augmented upon incubation with EPC-CM. Similar findings were observed for aortic ring outgrowth with significantly longer sprouts. The EPC-CM-induced activities were significantly reduced by the blockage of the PI3K/AKT and MEK/ERK signaling pathways. Similarly to the outcome of the rBCEC4 experiments, inhibition of the PI3K/AKT and MEK/ERK pathways significantly interfered with capillary sprouting induced by EPC-CM. Conclusion The present study demonstrates that EPC-derived paracrine factors substantially promote the angiogenic response of brain microvascular endothelial cells. In addition, our findings identified the PI3K/AKT and MEK/ERK pathways to play a central role in mediating these effects.
Collapse
Affiliation(s)
- Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
| | - Stefanie Seiler
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
| | - Anna-Lena Fuchs
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
| | - Jennifer Staudigl
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University of Bern, Inselspital, Bern, Switzerland
- * E-mail:
| |
Collapse
|
246
|
Kozakowska M, Szade K, Dulak J, Jozkowicz A. Role of heme oxygenase-1 in postnatal differentiation of stem cells: a possible cross-talk with microRNAs. Antioxid Redox Signal 2014; 20:1827-50. [PMID: 24053682 PMCID: PMC3961774 DOI: 10.1089/ars.2013.5341] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. HO-1 via heme removal and degradation products acts as a cytoprotective, anti-inflammatory, immunomodulatory, and proangiogenic protein, regulating also a cell cycle. Additionally, HO-1 can translocate to nucleus and regulate transcription factors, so it can also act independently of enzymatic function. RECENT ADVANCES Recently, a body of evidence has emerged indicating a role for HO-1 in postnatal differentiation of stem and progenitor cells. Maturation of satellite cells, skeletal myoblasts, adipocytes, and osteoclasts is inhibited by HO-1, whereas neurogenic differentiation and formation of cardiomyocytes perhaps can be enhanced. Moreover, HO-1 influences a lineage commitment in pluripotent stem cells and maturation of hematopoietic cells. It may play a role in development of osteoblasts, but descriptions of its exact effects are inconsistent. CRITICAL ISSUES In this review we discuss a role of HO-1 in cell differentiation, and possible HO-1-dependent signal transduction pathways. Among the potential mediators, we focused on microRNA (miRNA). These small, noncoding RNAs are critical for cell differentiation. Recently we have found that HO-1 not only influences expression of specific miRNAs but also regulates miRNA processing enzymes. FUTURE DIRECTIONS It seems that interplay between HO-1 and miRNAs may be important in regulating fates of stem and progenitor cells and needs further intensive studies.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- 1 Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University , Krakow, Poland
| | | | | | | |
Collapse
|
247
|
Ando Y, Matsubara K, Ishikawa J, Fujio M, Shohara R, Hibi H, Ueda M, Yamamoto A. Stem cell-conditioned medium accelerates distraction osteogenesis through multiple regenerative mechanisms. Bone 2014; 61:82-90. [PMID: 24389414 DOI: 10.1016/j.bone.2013.12.029] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 12/01/2013] [Accepted: 12/24/2013] [Indexed: 12/13/2022]
Abstract
Distraction osteogenesis (DO) successfully induces large-scale skeletal tissue regeneration, but it involves an undesirably long treatment period. A high-speed DO mouse model (H-DO) with a distraction speed twice that of a control DO model failed to generate new bone callus in the distraction gap. Here we demonstrate that the local administration of serum-free conditioned medium from human mesenchymal stem cells (MSC-CM) accelerated callus formation in the mouse H-DO model. Secretomic analysis identified factors contained in MSC-CM that recruit murine bone marrow stromal cells (mBMSCs) and endothelial cells/endothelial progenitor cells (EC/EPCs), inhibit inflammation and apoptosis, and promote osteoblast differentiation, angiogenesis, and cell proliferation. Functional assays identified MCP-1/-3 and IL-3/-6 as essential factors in recruiting mBMSCs and EC/EPCs. IL-3/-6 also enhanced the osteogenic differentiation of mBMSCs. MSC-CM that had been depleted of MCP-1/-3 failed to recruit mBMSCs, and consequently failed to promote callus formation. Taken together, our data suggest that MSCs produce a broad repertoire of trophic factors with tissue-regenerative activities that accelerate healing in the DO process.
Collapse
Affiliation(s)
- Yuji Ando
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kohki Matsubara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Jun Ishikawa
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masahito Fujio
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Ryutaro Shohara
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| |
Collapse
|
248
|
Kojima H, Kim J, Chan L. Emerging roles of hematopoietic cells in the pathobiology of diabetic complications. Trends Endocrinol Metab 2014; 25:178-87. [PMID: 24507996 PMCID: PMC3975817 DOI: 10.1016/j.tem.2014.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/21/2013] [Accepted: 01/09/2014] [Indexed: 02/08/2023]
Abstract
Diabetic complications encompass macrovascular events, mainly the result of accelerated atherosclerosis, and microvascular events that strike the eye (retinopathy), kidney (nephropathy), and nervous system (neuropathy). The traditional view is that hyperglycemia-induced dysregulated biochemical pathways cause injury and death of cells intrinsic to the organs affected. There is emerging evidence that diabetes compromises the function of the bone marrow (BM), producing a stem cell niche-dependent defect in hematopoietic stem cell mobilization. Furthermore, dysfunctional BM-derived hematopoietic cells contribute to diabetic complications. Thus, BM cells are not only a victim but also an accomplice in diabetes and diabetic complications. Understanding the underlying molecular mechanisms may lead to the development of new therapies to prevent and/or treat diabetic complications by specifically targeting these perpetrators.
Collapse
Affiliation(s)
- Hideto Kojima
- Departments of Medicine and Molecular and Cellular Biology, and the Diabetes and Endocrinology Research Center, Baylor College of Medicine, Houston, Texas 77030, USA; Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Jongoh Kim
- Departments of Medicine and Molecular and Cellular Biology, and the Diabetes and Endocrinology Research Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Lawrence Chan
- Departments of Medicine and Molecular and Cellular Biology, and the Diabetes and Endocrinology Research Center, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
249
|
The role of fucosylation in the promotion of endothelial progenitor cells in neovascularization and bone repair. Biomaterials 2014; 35:3777-85. [DOI: 10.1016/j.biomaterials.2014.01.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 01/09/2014] [Indexed: 01/07/2023]
|
250
|
Muniyappa R, Sowers JR. Glycogen synthase kinase-3β and cathepsin B in diabetic endothelial progenitor cell dysfunction: an old player finds a new partner. Diabetes 2014; 63:1194-7. [PMID: 24651804 PMCID: PMC3964509 DOI: 10.2337/db14-0004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Ranganath Muniyappa
- Clinical Endocrine Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - James R. Sowers
- Department of Internal Medicine, Department of Medical Pharmacology and Physiology, and Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO
- Corresponding author: James R. Sowers,
| |
Collapse
|