Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Angiogenesis ; 22(3): 411-420, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30929097

RESUMO

Lower numbers of progenitor cells (PCs) in peripheral blood (PB) have been associated with cardiovascular events in high-risk populations. Therapies aiming to increase the numbers of PCs in circulation have been developed, but clinical trials did not result in better outcomes. It is currently unknown what causes the reduction in PB PC numbers: whether it is primary depletion of the progenitor cell reserve, or a reduced mobilization of PCs from the bone marrow (BM). In this study, we examine if PB and BM PC numbers predict Amputation-Free Survival (AFS) in patients with Severe Limb Ischemia (SLI). We obtained PB and BM from 160 patients enrolled in a clinical trial investigating BM cell therapy for SLI. Samples were incubated with antibodies against CD34, KDR, CD133, CD184, CD14, CD105, CD140b, and CD31; PC populations were enumerated by flow cytometry. Higher PB CD34+ and CD133+ PC numbers were related to AFS (Both Hazard Ratio [HRevent] = 0.56, p = 0.003 and p = 0.0007, respectively). AFS was not associated with the other cell populations in PB. BM PC numbers correlated with PB PC numbers and showed similar HRs for AFS. A further subdivision based on relative BM and PB PC numbers showed that BM PC numbers, rather than mobilization, associated with AFS. Both PB and BM PC numbers are associated with AFS independently from traditional risk factor and show very similar risk profiles. Our data suggest that depletion of the progenitor cell reserve, rather than decreased PC mobilization, underlies the association between PB PC numbers and cardiovascular risk.


Assuntos
Células da Medula Óssea/patologia , Extremidades/irrigação sanguínea , Isquemia/patologia , Células-Tronco/patologia , Idoso , Amputação Cirúrgica , Contagem de Células , Feminino , Humanos , Isquemia/sangue , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fatores de Risco
2.
J Tissue Eng Regen Med ; 13(3): 433-445, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30650247

RESUMO

To introduce a functional vascular network into tissue-engineered bone equivalents, human endothelial colony forming cells (ECFCs) and multipotent mesenchymal stromal cells (MSCs) can be cocultured. Here, we studied the impact of donor variation of human bone marrow-derived MSCs and cord blood-derived ECFCs on vasculogenesis and osteogenesis using a 3D in vitro coculture model. Further, to make the step towards cocultures consisting of cells derived from a single donor, we tested how induced pluripotent stem cell (iPSC)-derived human endothelial cells (iECs) performed in coculture models. Cocultures with varying combinations of human donors of MSCs, ECFCs, or iECs were prepared in Matrigel. The constructs were cultured in an osteogenic differentiation medium. Following a 10-day culture period, the length of the prevascular structures and osteogenic differentiation were evaluated for up to 21 days of culture. The particular combination of MSC and ECFC donors influenced the vasculogenic properties significantly and induced variation in osteogenic potential. In addition, the use of iECs in the cocultures resulted in prevascular structure formation in osteogenically differentiated constructs. Together, these results showed that close attention to the source of primary cells, such as ECFCs and MSCs, is critical to address variability in vasculogenic and osteogenic potential. The 3D coculture model appeared to successfully generate prevascularized constructs and were sufficient in exceeding the ~200 µm diffusion limit. In addition, iPSC-derived cell lineages may decrease variability by providing a larger and potentially more uniform source of cells for future preclinical and clinical applications.


Assuntos
Técnicas de Cocultura/métodos , Hidrogéis/farmacologia , Neovascularização Fisiológica , Osteogênese , Doadores de Tecidos , Adulto , Idoso , Calcificação Fisiológica/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Colágeno/farmacologia , Ensaio de Unidades Formadoras de Colônias , Combinação de Medicamentos , Células Endoteliais/citologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Laminina/farmacologia , Masculino , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteonectina/metabolismo , Proteoglicanas/farmacologia , Adulto Jovem
3.
Stem Cells Int ; 2019: 1232810, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31933648

RESUMO

BACKGROUND: Cell-based therapies are being developed to meet the need for curative therapy in chronic kidney disease (CKD). Bone marrow- (BM-) derived mesenchymal stromal cells (MSCs) enhance tissue repair and induce neoangiogenesis through paracrine action of secreted proteins and extracellular vesicles (EVs). Administration of allogeneic BM MSCs is less desirable in a patient population likely to require a kidney transplant, but potency of autologous MSCs should be confirmed, given previous indications that CKD-induced dysfunction is present. While the immunomodulatory capacity of CKD BM MSCs has been established, it is unknown whether CKD affects wound healing and angiogenic potential of MSC-derived CM and EVs. METHODS: MSCs were cultured from BM obtained from kidney transplant recipients (N = 15) or kidney donors (N = 17). Passage 3 BM MSCs and BM MSC-conditioned medium (CM) were used for experiments. EVs were isolated from CM by differential ultracentrifugation. BM MSC differentiation capacity, proliferation, and senescence-associated ß-galactosidase activity was assessed. In vitro promigratory and proangiogenic capacity of BM MSC-derived CM and EVs was assessed using an in vitro scratch wound assay and Matrigel angiogenesis assay. RESULTS: Healthy and CKD BM MSCs exhibited similar differentiation capacity, proliferation, and senescence-associated ß-galactosidase activity. Scratch wound migration was not significantly different between healthy and CKD MSCs (P = 0.18). Healthy and CKD BM MSC-derived CM induced similar tubule formation (P = 0.21). There was also no difference in paracrine regenerative function of EVs (scratch wound: P = 0.6; tubulogenesis: P = 0.46). CONCLUSIONS: Our results indicate that MSCs have an intrinsic capacity to produce proangiogenic paracrine factors, including EVs, which is not affected by donor health status regarding CKD. This suggests that autologous MSC-based therapy is a viable option in CKD.

4.
BMJ Open ; 8(8): e020479, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127049

RESUMO

INTRODUCTION: Systemic sclerosis (SSc) is an autoimmune disease characterised by inflammation, fibrosis and vasculopathy. Digital ulcers (DUs) are a frequent manifestation of vasculopathy in patients with SSc. Despite recent advances in pharmacological treatments, DU still have major health and economic implications. As there is currently no proven therapeutic strategy to promote DU healing, new treatments are urgently needed. Mesenchymal stem or stromal cells (MSCs) may provide a novel therapy for DU in SSc, because of their immunomodulatory and vasculoregenerative properties. Allogeneic MSC therapy involves functionally competent MSCs from healthy donors and may be used as 'off-the-shelf' available treatment. This study will evaluate whether allogeneic MSC therapy is a safe and potentially efficacious treatment for DU of SSc. METHODS AND ANALYSIS: The MANUS (Mesenchymal stromal cells for Angiogenesis and Neovascularization in digital Ulcers of Systemic Sclerosis) Trial is a double-blind randomised placebo-controlled trial. 20 patients with SSc with refractory DU will be randomised to receive eight intramuscular injections with either placebo or 50*106 MSCs. The primary outcome is the toxicity of the treatment at 12 weeks after administration. Secondary outcomes include (serious) adverse events, number and time to healing of DU, pain, reported hand function, quality of life and SSc disease activity. We will also evaluate changes in nailfold capillaroscopy pattern, as well as biochemical parameters and biomarkers in peripheral blood and skin biopsies. Follow-up visits will be scheduled at 48 hours and 2, 4, 8, 12, 24 and 52 weeks post-treatment. If the results confirm safety, feasibility and potential efficacy, a large multicentre randomised controlled trial with longer follow-up will be initiated focusing on efficacy. ETHICS AND DISSEMINATION: The study has been approved by the Dutch Central Committee on Research Concerning Human Subjects (protocol no: NL51705.000.15). The results will be disseminated through patient associations and conventional scientific channels. TRIAL REGISTRATION NUMBER: NCT03211793; Pre-results.


Assuntos
Transplante de Células-Tronco Mesenquimais , Escleroderma Sistêmico/complicações , Úlcera Cutânea/cirurgia , Adulto , Aloenxertos , Protocolos Clínicos , Método Duplo-Cego , Humanos , Injeções Intramusculares , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Úlcera Cutânea/etiologia
5.
Curr Rheumatol Rep ; 18(2): 12, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26943351

RESUMO

Systemic sclerosis (SSc) is a rare autoimmune connective tissue disease with a high mortality and morbidity. While progress has been made in terms of identifying high-risk patients and implementing new treatment strategies, therapeutic options remain limited. In the past few decades, various cellular therapies have emerged, which have been studied in SSc and other conditions. Here, we provide a comprehensive review of currently available cellular therapies and critically assess their merit as disease-modifying treatment for SSc. Currently, hematopoietic stem cell transplantation is the only cellular therapy that has demonstrated clinical effects on the immune system, neoangiogenesis, and fibrosis. Robust mechanistic studies as well as clinical trials are essential to move the field forward.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Escleroderma Sistêmico/terapia , Ensaios Clínicos como Assunto/métodos , Células Dendríticas/transplante , Transplante de Células-Tronco Hematopoéticas/tendências , Humanos , Transplante de Células-Tronco Mesenquimais/tendências , Linfócitos T Reguladores/transplante
6.
Biomaterials ; 76: 187-95, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26524538

RESUMO

In an in-situ approach towards tissue engineered cardiovascular replacement grafts, cell-free scaffolds are implanted that engage in endogenous tissue formation. Bioactive molecules can be incorporated into such grafts to facilitate cellular recruitment. Stromal cell derived factor 1α (SDF1α) is a powerful chemoattractant of lymphocytes, monocytes and progenitor cells and plays an important role in cellular signaling and tissue repair. Short SDF1α-peptides derived from its receptor-activating domain are capable of activating the SDF1α-specific receptor CXCR4. Here, we show that SDF1α-derived peptides can be chemically modified with a supramolecular four-fold hydrogen bonding ureido-pyrimidinone (UPy) moiety, that allows for the convenient incorporation of the UPy-SDF1α-derived peptides into a UPy-modified polymer scaffold. We hypothesized that a UPy-modified material bioactivated with these UPy-SDF1α-derived peptides can retain and stimulate circulating cells in an anti-inflammatory, pro-tissue formation signaling environment. First, the early recruitment of human peripheral blood mononuclear cells to the scaffolds was analyzed in vitro in a custom-made mesofluidic device applying physiological pulsatile fluid flow. Preferential adhesion of lymphocytes with reduced expression of inflammatory factors TNFα, MCP1 and lymphocyte activation marker CD25 was found in the bioactivated scaffolds, indicating a reduction in inflammatory signaling. As a proof of concept, in-vivo implantation of the bioactivated scaffolds as rat abdominal aorta interposition grafts showed increased cellularity by CD68+ cells after 7 days. These results indicate that a completely synthetic, cell-free biomaterial can attract and stimulate specific leukocyte populations through supramolecular incorporation of short bioactive SDF1α derived peptides.


Assuntos
Prótese Vascular , Quimiocina CXCL12/química , Peptídeos/química , Humanos , Ligação de Hidrogênio , Microscopia Eletrônica de Varredura , Proteólise , Engenharia Tecidual
7.
Stem Cell Res Ther ; 6: 63, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25889756

RESUMO

INTRODUCTION: Healthy bone marrow cell (BMC) infusion improves renal function and limits renal injury in a model of chronic kidney disease (CKD) in rats. However, BMCs derived from rats with CKD fail to retain beneficial effects, demonstrating limited therapeutic efficacy. Statins have been reported to improve cellular repair mechanisms. METHODS: We studied whether exposing CKD rat BMCs ex vivo to pravastatin improved their in vivo therapeutic efficacy in CKD and compared this to systemic in vivo treatment. Six weeks after CKD induction, healthy BMCs, healthy pravastatin-pretreated BMCs, CKD BMCs or CKD pravastatin-pretreated BMCs were injected into the renal artery of CKD rats. RESULTS: At 6 weeks after BMC injection renal injury was reduced in pravastatin-pretreated CKD BMC recipients vs. CKD BMC recipients. Effective renal plasma flow was lower and filtration fraction was higher in CKD BMC recipients compared to all groups whereas there was no difference between pravastatin-pretreated CKD BMC and healthy BMC recipients. Mean arterial pressure was higher in CKD BMC recipients compared to all other groups. In contrast, 6 weeks of systemic in vivo pravastatin treatment had no effect. In vitro results showed improved migration, decreased apoptosis and lower excretion of pro-inflammatory Chemokine (C-X-C Motif) Ligand 5 in pravastatin-pretreated CKD BMCs. CONCLUSIONS: Short ex vivo exposure of CKD BMC to pravastatin improves CKD BMC function and their subsequent therapeutic efficacy in a CKD setting, whereas systemic statin treatment did not provide renal protection.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Pravastatina/farmacologia , Insuficiência Renal Crônica/patologia , Animais , Apoptose , Pressão Sanguínea , Peso Corporal , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Movimento Celular , Quimiocina CXCL5/análise , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Taxa de Filtração Glomerular , Rim/irrigação sanguínea , Rim/fisiologia , Masculino , Ratos , Ratos Endogâmicos Lew , Insuficiência Renal Crônica/terapia , Insuficiência Renal Crônica/veterinária , Doadores de Tecidos , Transplante Homólogo
8.
PLoS One ; 10(2): e0118653, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25689740

RESUMO

BACKGROUND: Techniques to treat urethral stricture and hypospadias are restricted, as substitution of the unhealthy urethra with tissue from other origins (skin, bladder or buccal mucosa) has some limitations. Therefore, alternative sources of tissue for use in urethral reconstructions are considered, such as ex vivo engineered constructs. PURPOSE: To review recent literature on tissue engineering for human urethral reconstruction. METHODS: A search was made in the PubMed and Embase databases restricted to the last 25 years and the English language. RESULTS: A total of 45 articles were selected describing the use of tissue engineering in urethral reconstruction. The results are discussed in four groups: autologous cell cultures, matrices/scaffolds, cell-seeded scaffolds, and clinical results of urethral reconstructions using these materials. Different progenitor cells were used, isolated from either urine or adipose tissue, but slightly better results were obtained with in vitro expansion of urothelial cells from bladder washings, tissue biopsies from the bladder (urothelium) or the oral cavity (buccal mucosa). Compared with a synthetic scaffold, a biological scaffold has the advantage of bioactive extracellular matrix proteins on its surface. When applied clinically, a non-seeded matrix only seems suited for use as an onlay graft. When a tubularized substitution is the aim, a cell-seeded construct seems more beneficial. CONCLUSIONS: Considerable experience is available with tissue engineering of urethral tissue in vitro, produced with cells of different origin. Clinical and in vivo experiments show promising results.


Assuntos
Procedimentos de Cirurgia Plástica/métodos , Engenharia Tecidual/métodos , Uretra/cirurgia , Humanos , Alicerces Teciduais , Uretra/citologia
9.
Br J Pharmacol ; 172(6): 1607-19, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24597655

RESUMO

BACKGROUND AND PURPOSE: Whether NO, carbon monoxide (CO) and hydrogen sulfide (H2 S) compensate for each other when one or more is depleted is unclear. Inhibiting NOS causes hypertension and kidney injury. Both global depletion of H2 S by cystathionine γ-lyase (CSE) gene deletion and low levels of exogenous H2 S cause hypertension. Inhibiting CO-producing enzyme haeme oxygenase-1 (HO-1) makes rodents hypersensitive to hypertensive stimuli. We hypothesized that combined inhibition of NOS and HO-1 exacerbates hypertension and renal injury, but how combined inhibition of NOS and CSE affect hypertension and renal injury was unclear. EXPERIMENTAL APPROACH: Rats were treated with inhibitors of NOS (L-nitroarginine; LNNA), CSE (DL-propargylglycine; PAG), or HO-1 (tin protoporphyrin; SnPP) singly for 1 or 4 weeks or in combinations for 4 weeks. KEY RESULTS: LNNA always reduced NO, decreased H2 S and increased CO after 4 weeks. PAG abolished H2 S, always enhanced CO and reduced NO, but not when used in combination with other inhibitors. SnPP always increased NO, enhanced H2 S and inhibited CO after 1 week. Rats treated with LNNA, but not PAG and SnPP, rapidly developed hypertension followed by renal dysfunction. LNNA-induced hypertension was ameliorated and renal dysfunction prevented by all additional treatments. Renal HO-1 expression was increased by LNNA in injured tubules and increased in all tubules by all other treatments. CONCLUSIONS AND IMPLICATIONS: The amelioration of LNNA-induced hypertension and renal injury by additional inhibition of H2 S and/or CO-producing enzymes appeared to be associated with secondary increases in renal CO or NO production.


Assuntos
Monóxido de Carbono/metabolismo , Sulfeto de Hidrogênio/metabolismo , Hipertensão/fisiopatologia , Óxido Nítrico/metabolismo , Alcinos/farmacologia , Animais , Cistationina gama-Liase/antagonistas & inibidores , Glicina/análogos & derivados , Glicina/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Rim/metabolismo , Rim/patologia , Masculino , Metaloporfirinas/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroarginina/farmacologia , Protoporfirinas/farmacologia , Ratos , Ratos Sprague-Dawley
11.
Mol Ther ; 22(11): 1960-70, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25174586

RESUMO

Critical limb ischemia (CLI) is often poorly treatable by conventional management and alternatives such as autologous cell therapy are increasingly investigated. Whereas previous studies showed a substantial impairment of neovascularization capacity in primary bone-marrow (BM) isolates from patients, little is known about dysfunction in patient-derived BM mesenchymal stromal cells (MSCs). In this study, we have compared CLI-MSCs to healthy controls using gene expression profiling and functional assays for differentiation, senescence and in vitro and in vivo pro-angiogenic ability. Whereas no differentially expressed genes were found and adipogenic and osteogenic differentiation did not significantly differ between groups, chondrogenic differentiation was impaired in CLI-MSCs, potentially as a consequence of increased senescence. Migration experiments showed no differences in growth factor sensitivity and secretion between CLI- and control MSCs. In a murine hind-limb ischemia model, recovery of perfusion was enhanced in MSC-treated mice compared to vehicle controls (71 ± 24% versus 44 ± 11%; P < 1 × 10(-6)). CLI-MSC- and control-MSC-treated animals showed nearly identical amounts of reperfusion (ratio CLI:Control = 0.98, 95% CI = 0.82-1.14), meeting our criteria for statistical equivalence. The neovascularization capacity of MSCs derived from CLI-patients is not compromised and equivalent to that of control MSCs, suggesting that autologous MSCs are suitable for cell therapy in CLI patients.


Assuntos
Isquemia/patologia , Isquemia/terapia , Perna (Membro)/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Animais , Diferenciação Celular , Células Cultivadas , Senescência Celular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Perna (Membro)/irrigação sanguínea , Camundongos
12.
J Control Release ; 184: 58-66, 2014 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-24727077

RESUMO

Timely vascularization is essential for optimal performance of bone regenerative constructs. Vascularization is efficiently stimulated by vascular endothelial growth factor (VEGF), a substance with a short half-life time. This study investigates the controlled release of VEGF from gelatin microparticles (GMPs) as a means to prolong VEGF activity at the preferred location within 3D bioprinted scaffolds, and the effects on subsequent vascularization. The release of VEGF from GMPs was continuous for 3 weeks during in vitro studies, and bioactivity was confirmed using human endothelial progenitor cells (EPCs) in migration assays. Traditional and real-time migration assays showed immediate and efficient EPC migration in the presence of GMP-released VEGF, indistinguishable from VEGF-solution that was added to the medium. Matrigel scaffolds containing EPCs and VEGF, which was released either in a fast or sustained fashion by application of GMPs, were investigated for their in vivo vasculogenic capacity. Implantation in subcutaneous pockets in nude mice for one week demonstrated that vessel formation was significantly higher in the VEGF sustained-release group compared to the fast release group. In addition, regional differences with respect to VEGF release were introduced in 3D bioprinted EPC-laden scaffolds and their influence on vasculogenesis was investigated in vivo. The different regions were retained and vessel formation occurred analogous with the results seen in the Matrigel plugs. We conclude that GMPs are suitable to generate sustained release profiles of bioactive VEGF, and that they can be used to create defined differentiation regions in 3D bioprinted heterogeneous constructs, allowing a new generation of smart scaffold design. The prolonged presence of VEGF led to a significant increase in scaffold vascularization when applied in vivo.


Assuntos
Neovascularização Fisiológica/efeitos dos fármacos , Impressão Tridimensional , Alicerces Teciduais , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Animais , Movimento Celular/efeitos dos fármacos , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/fisiologia , Feminino , Gelatina/química , Humanos , Camundongos Nus , Fator A de Crescimento do Endotélio Vascular/química
13.
Circ Res ; 114(2): 311-4, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24218170

RESUMO

RATIONALE: The impact of severe cardiovascular disease and critical limb ischemia (CLI) on the bone marrow (BM) is largely unknown. OBJECTIVE: To investigate microvascular and neuropathic changes in BM of patients with CLI. METHODS AND RESULTS: BM biopsies were obtained from patients with CLI (n=33) included in the Rejuvenating Endothelial Progenitor Cells via Transcutaneous Intra-arterial Supplementation (JUVENTAS) trial (NCT00371371) and controls (n=12). We performed immunohistochemistry and histomorphometry of the BM to assess microvascular density and to evaluate pan-neuronal and sympathetic innervation, which is involved in progenitor cell mobilization. Microvascular density was reduced significantly in CLI compared with controls (P=0.01), as was sympathetic (P=0.047) and pan-neuronal innervation (P=0.006). No differences in microvascular density and sympathetic or pan-neuronal innervation were observed between patients with CLI with and without diabetes mellitus. CONCLUSIONS: CLI is associated with BM microvascular and neuropathic changes, both in patients with and without diabetes mellitus.


Assuntos
Medula Óssea/irrigação sanguínea , Medula Óssea/inervação , Isquemia/patologia , Microvasos/patologia , Sistema Nervoso Simpático/patologia , Idoso , Biomarcadores/análise , Biópsia , Exame de Medula Óssea , Estudos de Casos e Controles , Estado Terminal , Diabetes Mellitus/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microvasos/química , Pessoa de Meia-Idade , Sistema Nervoso Simpático/química
14.
Stem Cell Res Ther ; 4(6): 140, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24246031

RESUMO

Cell therapy using mesenchymal stromal cells (MSCs) is a promising new avenue of treatment for critical limb ischemia (CLI). Preclinical studies have suggested that MSCs enhance neovascularization in ischemic limbs. In this commentary, we discuss a recent study by Gupta and colleagues, one of the first human trials using allogeneic MSCs for CLI, in relation to the current state of knowledge regarding cell therapy for CLI.


Assuntos
Transplante de Medula Óssea/métodos , Isquemia/patologia , Perna (Membro)/patologia , Células-Tronco Mesenquimais/citologia , Doenças Vasculares Periféricas/terapia , Feminino , Humanos , Masculino
16.
PLoS One ; 8(9): e73161, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023827

RESUMO

AIMS: Tissue engineering is an innovative method to restore cardiovascular tissue function by implanting either an in vitro cultured tissue or a degradable, mechanically functional scaffold that gradually transforms into a living neo-tissue by recruiting tissue forming cells at the site of implantation. Circulating endothelial colony forming cells (ECFCs) are capable of differentiating into endothelial cells as well as a mesenchymal ECM-producing phenotype, undergoing Endothelial-to-Mesenchymal-transition (EndoMT). We investigated the potential of ECFCs to produce and organize ECM under the influence of static and cyclic mechanical strain, as well as stimulation with transforming growth factor ß1 (TGFß1). METHODS AND RESULTS: A fibrin-based 3D tissue model was used to simulate neo-tissue formation. Extracellular matrix organization was monitored using confocal laser-scanning microscopy. ECFCs produced collagen and also elastin, but did not form an organized matrix, except when cultured with TGFß1 under static strain. Here, collagen was aligned more parallel to the strain direction, similar to Human Vena Saphena Cell-seeded controls. Priming ECFC with TGFß1 before exposing them to strain led to more homogenous matrix production. CONCLUSIONS: Biochemical and mechanical cues can induce extracellular matrix formation by ECFCs in tissue models that mimic early tissue formation. Our findings suggest that priming with bioactives may be required to optimize neo-tissue development with ECFCs and has important consequences for the timing of stimuli applied to scaffold designs for both in vitro and in situ cardiovascular tissue engineering. The results obtained with ECFCs differ from those obtained with other cell sources, such as vena saphena-derived myofibroblasts, underlining the need for experimental models like ours to test novel cell sources for cardiovascular tissue engineering.


Assuntos
Células Endoteliais/citologia , Matriz Extracelular/metabolismo , Células-Tronco/citologia , Estresse Mecânico , Engenharia Tecidual , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Colágeno/metabolismo , Elastina/metabolismo , Células Endoteliais/efeitos dos fármacos , Fatores de Crescimento Endotelial/farmacologia , Matriz Extracelular/efeitos dos fármacos , Humanos , Imagem Molecular , Células-Tronco/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia
17.
Tissue Eng Part A ; 18(1-2): 208-18, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21859278

RESUMO

To ensure the survival of engineered bone after implantation, we combined human endothelial colony forming cells (ECFCs) and multipotent stromal cells (MSCs) as a proof of concept in a co-culture model to create in vitro prevascularized bone constructs. We hypothesized that a hypoxic stimulus will contribute to prevascularization of engineered bone. Bone marrow-derived MSCs and ECFCs from human adult peripheral blood were allowed to form co-culture pellets containing ECFCs and MSCs (1:4) or MSCs only in controls. After culture under normoxia or hypoxia (5%), pellets were harvested and processed for immunohistochemistry of CD31, α-smooth muscle actin, and osteocalcin. Expression of vascular endothelial growth factor and SDF-1α was analyzed by PCR to elucidate their involvement in hypoxic stimulation of prevascularization. The normoxic condition in co-cultures of MSCs and ECFCs supported the formation and maintenance of prevascular structures, including organized CD31-positive cells embraced by differentiated mural cells. These structures failed to form in hypoxic conditions, thereby rejecting the hypothesis that hypoxia stimulates prevasculogenesis in three-dimensional engineered bone constructs. Further, the formation of prevascular structures was paralleled by increased SDF-1α expression. It is suggested that actual oxygen levels were below 5% in the hypoxic co-cultures, which prevented prevascular structure formation. In conclusion, our normoxic co-culture model containing cells from clinically relevant sources sustained simultaneous endothelial, smooth muscle, and osteogenic differentiation.


Assuntos
Osso e Ossos/irrigação sanguínea , Osso e Ossos/fisiologia , Hipóxia/patologia , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Adulto , Idoso , Benzilaminas , Biomarcadores/metabolismo , Osso e Ossos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Meios de Cultura/farmacologia , Ciclamos , Feminino , Compostos Heterocíclicos/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos
18.
J Biol Chem ; 285(45): 34677-85, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20736166

RESUMO

Increased interferon (IFN)-ß signaling in patients with insufficient coronary collateralization and an inhibitory effect of IFNß on collateral artery growth in mice have been reported. The mechanisms of IFNß-induced inhibition of arteriogenesis are unknown. In stimulated monocytes from patients with chronic total coronary artery occlusion and decreased arteriogenic response, whole genome expression analysis showed increased expression of IFNß-regulated genes. Immunohistochemically, the IFNß receptor was localized in the vascular media of murine collateral arteries. Treatment of vascular smooth muscle cells (VSMC) with IFNß resulted in an attenuated proliferation, cell-cycle arrest, and increased expression of cyclin-dependent kinase inhibitor-1A (p21). The growth inhibitory effect of IFNß was attenuated by inhibition of p21 by RNA interference. IFNß-treated THP1 monocytes showed enhanced apoptosis. Subsequently, we tested if collateral artery growth can be stimulated by inhibition of IFNß-signaling. RNA interference of the IFNß receptor-1 (IFNAR1) increased VSMC proliferation, cell cycle progression, and reduced p21 gene expression. IFNß signaling and FAS and TRAIL expression were attenuated in monocytes from IFNAR1(-/-) mice, indicating reduced monocyte apoptosis. Hindlimb perfusion restoration 1 week after femoral artery ligation was improved in IFNAR1(-/-) mice compared with wild-type mice as assessed by infusion of fluorescent microspheres. These results demonstrate that IFNß inhibits collateral artery growth and VSMC proliferation through p21-dependent cell cycle arrest and induction of monocyte apoptosis. Inhibition of IFNß stimulates VSMC proliferation and collateral artery growth.


Assuntos
Ciclo Celular , Oclusão Coronária/metabolismo , Interferon beta/antagonistas & inibidores , Monócitos/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neovascularização Fisiológica , Animais , Apoptose/genética , Células Cultivadas , Oclusão Coronária/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Knockout , Interferência de RNA , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
19.
Blood ; 115(12): 2533-42, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20032497

RESUMO

The shear stress-induced transcription factor Krüppel-like factor 2 (KLF2) confers antiinflammatory properties to endothelial cells through the inhibition of activator protein 1, presumably by interfering with mitogen-activated protein kinase (MAPK) cascades. To gain insight into the regulation of these cascades by KLF2, we used antibody arrays in combination with time-course mRNA microarray analysis. No gross changes in MAPKs were detected; rather, phosphorylation of actin cytoskeleton-associated proteins, including focal adhesion kinase, was markedly repressed by KLF2. Furthermore, we demonstrate that KLF2-mediated inhibition of Jun NH(2)-terminal kinase (JNK) and its downstream targets ATF2/c-Jun is dependent on the cytoskeleton. Specifically, KLF2 directs the formation of typical short basal actin filaments, termed shear fibers by us, which are distinct from thrombin- or tumor necrosis factor-alpha-induced stress fibers. KLF2 is shown to be essential for shear stress-induced cell alignment, concomitant shear fiber assembly, and inhibition of JNK signaling. These findings link the specific effects of shear-induced KLF2 on endothelial morphology to the suppression of JNK MAPK signaling in vascular homeostasis via novel actin shear fibers.


Assuntos
Citoesqueleto de Actina/metabolismo , Células Endoteliais/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fator 2 Ativador da Transcrição/metabolismo , Animais , Aorta/citologia , Células Cultivadas , Células Endoteliais/citologia , Artéria Femoral/citologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fosforilação/fisiologia , Ratos , Fluxo Sanguíneo Regional/fisiologia , Veia Safena/citologia , Estresse Mecânico , Transdução Genética , Veias Umbilicais/citologia , Quinases Associadas a rho/metabolismo
20.
Curr Stem Cell Res Ther ; 4(2): 131-40, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19442197

RESUMO

The origin of vascular smooth muscle cells that accumulate in the neointima in vascular diseases such as transplant arteriosclerosis, atherosclerosis and restenosis remains subject to much debate. Smooth muscle cells are a highly heterogeneous cell population with different characteristics and markers, and distinct phenotypes in physiological and pathological conditions. Several studies have reported a role for bone marrow-derived progenitor cells in vascular maintenance and repair. Moreover, bone marrow-derived smooth muscle progenitor cells have been detected in human atherosclerotic tissue as well as in in vivo mouse models of vascular disease. However, it is not clear whether smooth muscle progenitor cells can be regarded as a 'friend' or 'foe' in neointima formation. In this review we will discuss the heterogeneity of smooth muscle cells, the role of smooth muscle progenitor cells in vascular disease, potential mechanisms that could regulate smooth muscle progenitor cell contribution and the implications this may have on designing novel therapeutic tools to prevent development and progression of vascular disease.


Assuntos
Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Células-Tronco/fisiologia , Doenças Vasculares/fisiopatologia , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Humanos , Miócitos de Músculo Liso/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Doenças Vasculares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA