Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell Biol Int ; 46(12): 2132-2141, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36073008

RESUMO

Bone marrow-derived mesenchymal stromal cells (BMSCs) physically associate with the hematopoietic stem cells (HSCs), forming a unique HSC niche. Owing to this proximity, the signaling mechanisms prevailing in the BMSCs affect the fate of the HSCs. In addition to cell-cell and cell-extracellular matrix interactions, various cytokines and growth factors present in the BM milieu evoke signaling mechanisms in the BMSCs. Previously, I have shown that priming of human BMSCs with transforming growth factor ß1 (TGFß1), a cytokine consistently found at active sites of hematopoiesis, boosts their hematopoiesis-supportive ability. Basic fibroblast growth factor (bFGF), another cytokine present in the marrow microenvironment, positively regulates hematopoiesis. Hence, I examined whether priming human BMSCs with bFGF improves their hematopoiesis-supportive ability. I found that bFGF-primed BMSCs stimulate hematopoiesis, as seen by a significant increase in colony formation from the bone marrow cells briefly interacted with them and the extensive proliferation of CD34+ HSCs cocultured with them. However, contrary to my expectation, I found that chimeric feeders comprising a mixture of TGF-primed and bFGF-primed BMSCs exerted a suppressive effect. These data demonstrate that though the TGF- and bFGF-primed BMSCs exert a salutary effect on hematopoiesis when used independently, they exert a suppressive effect when presented as a chimera. These findings suggest that the combinatorial effect of various priming agents and cytokines on the functionality of BMSCs toward the target tissues needs to be critically evaluated before they are clinically applied.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Humanos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células da Medula Óssea
2.
Adv Exp Med Biol ; 1347: 29-43, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34114129

RESUMO

Hematopoietic stem cells (HSCs) and their development are one of the most widely studied model systems in mammals. In adults, HSCs are predominantly found in the bone marrow, from where they maintain homeostasis. Besides bone marrow and mobilized peripheral blood, cord blood is also being used as an alternate allogenic source of transplantable HSCs. HSCs from both autologous and allogenic sources are being applied for the treatment of various conditions like blood cancers, anemia, etc. HSCs can further differentiate to mature blood cells. Differentiation process of HSCs is being extensively studied so as to obtain a large number of pure populations of various differentiated cells in vitro so that they can be taken up for clinical trials. The ability to generate sufficient quantity of clinical-grade specialized blood cells in vitro would take the field of hematology a step ahead in translational medicine.


Assuntos
Células-Tronco Hematopoéticas , Ciência Translacional Biomédica , Animais , Medula Óssea , Diferenciação Celular , Sangue Fetal
3.
Cell Biol Int ; 44(11): 2293-2306, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32749730

RESUMO

Transforming growth factor ß1 (TGFß1) is a negative regulator of hematopoiesis, and yet, it is frequently found at the active sites of hematopoiesis. Here, we show for the first time that bone marrow-derived mononuclear cells (BM MNCs) secrete TGFß1 in response to erythropoietin (EPO). We further show that human bone marrow-derived mesenchymal stromal cells (BMSCs) briefly exposed to the conditioned medium of EPO-primed MNCs, or purified TGFß1, gain significantly increased hematopoiesis-supportive ability. Mechanistically, we show that this phenomenon involves TGFß1-mediated activation of nitric oxide (NO) signalling pathway in the BMSCs. The data suggest that EPO-MNC-TGFß1 could be one of the regulatory axes operative in the bone marrow microenvironment involved in maintaining the functionality of the resident BMSCs.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Células Cultivadas , Eritropoetina/farmacologia , Hematopoese/genética , Hematopoese/fisiologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/fisiologia
4.
Stem Cells Dev ; 28(22): 1473-1479, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31559908

RESUMO

Regenerative potential of mesenchymal stem/stromal cells (MSCs) has led to their application in various cellular therapies. Since in vivo these cells are present in very low numbers, they need expansion in culture to get clinically relevant numbers; however, such long-term ex vivo manipulation leads to loss of their regenerative capacity. Although use of naïve MSCs is still the most common approach used in various therapies, several strategies, both genetic and pharmacological, are being tried out to boost the regenerative capacity of in vitro expanded MSCs. Such manipulations are very commonly reported for regeneration of various tissues like bone, cartilage, kidney, pancreas, and others. Likewise, several efforts have been made to investigate priming of MSCs to enhance their immunoregulatory activity, but such efforts have not been made to the same extent for enhancing the efficacy of hematopoietic stem cell transplantation (HSCT). Development of such approaches for HSCT would not only be useful for enhancing the transplantation efficacy of cord blood cells, which are fewer in numbers, and aged HSCs, which could be functionally compromised, but also for genetically modified HSCs, which are likely to be both, fewer in number and functionally compromised. This review specifically deals with application of "primed" MSCs in the scenario of HSCT.


Assuntos
Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/tendências , Transplante de Células-Tronco Mesenquimais/tendências , Vesículas Extracelulares/transplante , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Mesenquimais/citologia
5.
Methods Mol Biol ; 2029: 235-246, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31273746

RESUMO

Flow cytometry is a widely used laser-based technology for rapid analysis of the expression of cell surface antigens and intracellular molecules in various cell types including hematopoietic stem/progenitor cells (HSPCs). Multiparametric analysis of individual cells within a short time frame makes this tool attractive and indispensable in the field of stem cell research. This is accomplished by harnessing the specific light scattering ability of the cell type, which determines its size and internal complexity. In addition, use of fluorescently conjugated antibodies allows the detection of a specific surface or intracellular antigen present at that particular stage. Fluorescent Activated Cell Sorting (FACS) is used to separate a subset of cells from a heterogeneous cell population based on fluorescent labeling. Here we describe the general principles of flow cytometry and detailed methods for the isolation of HSPCs using flow cytometry as a tool.


Assuntos
Separação Celular/métodos , Citometria de Fluxo/métodos , Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Animais , Antígenos de Superfície/metabolismo , Corantes Fluorescentes/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Células-Tronco/metabolismo
6.
Methods Mol Biol ; 1854: 21-34, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29951740

RESUMO

Autophagy is an important cellular process for maintenance of quality and functionality of cells. This happens through repair and renewal of cellular components like proteins and mitochondria. Reduction in autophagy process in aged hematopoietic stem cells (HSCs) leads to their compromised stemness and self-renewal capacity, and consequently, their applicability in various regenerative therapies also reduces. HSC functions are regulated by their microenvironment, known as "HSC niche," which comprises of mesenchymal stromal cells (MSCs), osteoblasts, endothelial cells, etc. In this niche, the MSCs are known to closely interact with the HSCs, and therefore, they can directly influence the stem cell fate. In our earlier studies, we have demonstrated that young MSCs or aged MSCs rejuvenated by treating them with LY294002, a PI3K inhibitor (rescued aged MSCs), rejuvenate aged HSCs via intercellular transfer of microvesicles (MVs) harboring autophagy-inducing mRNAs.Here, we describe the protocol for induction of autophagy in aged HSCs by incubating them with microvesicles (MVs) collected from young MSCs or rescued aged MSCs. We also describe the protocols for determination of autophagy levels in these HSCs.


Assuntos
Micropartículas Derivadas de Células/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , RNA Mensageiro/genética , Animais , Autofagia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Senescência Celular , Cromonas/farmacologia , Técnicas de Cocultura , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Morfolinas/farmacologia , Nicho de Células-Tronco
7.
Cell Biol Int ; 42(11): 1474-1483, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30136736

RESUMO

Angiogenesis, the process of new blood vessel formation from pre-existing vessels, is essential for growth and development. Development of drugs that can accelerate or decelerate angiogenesis in the context of various diseases requires appropriate preclinical screening. As angiogenesis involves complex cellular and molecular processes, in vivo studies are superior to in vitro investigations. Conventional in vitro, in vivo, and ex ovo models of angiogenesis are time consuming and tedious, and require sophisticated infrastructure for embryo culture. In the present study, we established an in ovo chick embryo yolk sac membrane (YSM) assay for angiogenesis and tested the angiogenic potential of arginine, conditioned medium (CM) from human adipose tissue and placenta-derived mesenchymal stem cells (ADMSCs-CM and PDMSCs-CM), avastin and vitamin C. The obtained results were confirmed with the routinely employed chick embryo Chorioallantoic Membrane (CAM) assay. Both assays revealed the pro-angiogenic nature of arginine, ADMSCs-CM, and PDMSCs-CM, and the anti-angiogenic effect of avastin and vitamin C. This novel in ovo YSM model is simple, reproducible, and highly economic in terms of the time frame and cost incurred. The proposed model is thus a suitable substitute to the CAM model for pilot screening of potential angiogenic and anti-angiogenic agents.


Assuntos
Inibidores da Angiogênese/farmacologia , Bioensaio/métodos , Membrana Corioalantoide/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Saco Vitelino/metabolismo , Animais , Arginina/farmacologia , Embrião de Galinha , Membrana Corioalantoide/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Modelos Biológicos , Projetos Piloto , Reprodutibilidade dos Testes , Saco Vitelino/efeitos dos fármacos
8.
PLoS One ; 13(8): e0202510, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30153276

RESUMO

Cellular secretory products have infinite potential, which is only recently explored for research and therapeutic applications. The present study elaborated on the formation of a unique matrix-entrapped cellular secretome (MCS), a hydrogel-like secretome produced by bone marrow-derived mononuclear cells when cultured on a three-dimensional electrospun nanofiber matrix under specific conditions. These culture conditions support the growth of a mixed population predominantly comprising of endothelial precursor cells (EPCs), along with mesenchymal stromal cells and pericytes. Interestingly, such secretome is not formed in a pure culture of EPCs on the similarly formulated matrix, suggesting that a heterotypic cell-cell interaction is essential for the formation of MCS. In addition, the specific composition of the matrix was found to be a critical necessity for the formation of MCS. Furthermore, the application of the MCS as a substrate promotes the growth of EPCs in culture. It also rescues the diabetes-induced EPC dysfunction as assessed based on the parameters, such as viability, proliferation, colony formation, cellular adhesion, chemotactic migration, and tubule formation. MCS augments the levels of eNOS-specific mRNA (Nos3) and also promotes the restoration of the SDF1/CXCR4 axis in diabetic EPCs. Notably, a topical application of MCS on diabetic wounds leads to an accelerated wound closure. Thus, the current data showed that MCS forms an excellent cell-free biomaterial in the treatment of diabetic wounds and non-healing ulcers.


Assuntos
Diabetes Mellitus Experimental/terapia , Angiopatias Diabéticas/terapia , Células Progenitoras Endoteliais/metabolismo , Matriz Extracelular/química , Nanofibras , Cicatrização , Animais , Proliferação de Células , Sobrevivência Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Células Progenitoras Endoteliais/patologia , Feminino , Masculino , Células-Tronco Mesenquimais , Camundongos , Nanofibras/química , Nanofibras/uso terapêutico , Pericitos/patologia
9.
Stem Cells ; 36(3): 420-433, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29230885

RESUMO

Donor age is one of the major concerns in bone marrow transplantation, as the aged hematopoietic stem cells (HSCs) fail to engraft efficiently. Here, using murine system, we show that a brief interaction of aged HSCs with young mesenchymal stromal cells (MSCs) rejuvenates them and restores their functionality via inter-cellular transfer of microvesicles (MVs) containing autophagy-related mRNAs. Importantly, we show that MSCs gain activated AKT signaling as a function of aging. Activated AKT reduces the levels of autophagy-related mRNAs in their MVs, and partitions miR-17 and miR-34a into their exosomes, which upon transfer into HSCs downregulate their autophagy-inducing mRNAs. Our data identify previously unknown mechanisms operative in the niche-mediated aging of HSCs. Inhibition of AKT in aged MSCs increases the levels of autophagy-related mRNAs in their MVs and reduces the levels of miR-17 and miR-34a in their exosomes. Interestingly, transplantation experiments showed that the rejuvenating power of these "rescued" MVs is even better than that of the young MVs. We demonstrate that such ex vivo rejuvenation of aged HSCs could expand donor cohort and improve transplantation efficacy. Stem Cells 2018;36:420-433.


Assuntos
Envelhecimento/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Micropartículas Derivadas de Células/metabolismo , Exossomos/genética , Exossomos/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo
10.
Stem Cells Dev ; 26(1): 35-48, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27676403

RESUMO

Marrow adipocytes pose a significant problem in post-transplant regeneration of hematopoiesis owing to their negative effects on regeneration of hematopoiesis. However, the precise mechanism operative in this negative regulation is not clear. In this study, we show that marrow adipocytes express neuropilin-1 (NRP1) as a function of differentiation and inhibit regeneration of hematopoiesis by three principal mechanisms: one, by inducing apoptosis in hematopoietic stem/progenitor cells (HSPCs) through the death receptor-mediated pathway; two, by downregulating CXCR4 expression on the HSPCs through ligand-mediated internalization; and three, by secreting copious amounts of transforming growth factor ß1 (TGFß1), a known inhibitor of hematopoiesis. Silencing of NRP1 in these adipocytes rescued the apoptosis of cocultured HSPCs and boosted the CXCR4 surface expression on them, showing an active role of NRP1 in these processes. However, such silencing had no effect on TGFß1 secretion and consequent inhibition of hematopoiesis by them, showing that secretion of TGFß1 by adipocytes is independent of NRP1 expression by them. Surprisingly, mesenchymal stromal cells modified with NRP1 supported expansion of HSPCs having enhanced functionality, suggesting that NRP1 exerts a context-dependent effect on hematopoiesis. Our data demonstrate that NRP1 is an important niche component and exerts context-dependent effects on HSPCs. Based on these data, we speculate that antibody- or peptide-mediated blocking of NRP1-HSC interactions coupled with a pharmacological inhibition of TGFß1 signaling may help in combating the negative regulation of post-transplant regeneration of hematopoiesis in a more effective manner.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Neuropilina-1/metabolismo , Nicho de Células-Tronco , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Apoptose , Diferenciação Celular , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Inativação Gênica , Hematopoese , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Receptores CXCR4/metabolismo , Regeneração , Transcriptoma/genética , Fator de Crescimento Transformador beta1/metabolismo
11.
Stem Cell Res ; 17(3): 646-653, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27865162

RESUMO

Pre-transplant myeloablation is associated with marrow adipogenesis, resulting in delayed engraftment of hematopoietic stem cells (HSCs). This is strongly undesirable, especially when the donor HSCs are fewer in numbers or have compromised functionality. The molecular mechanisms behind irradiation-induced marrow adipogenesis have not been extensively investigated. Here we show that bone marrow (BM) cells, especially T-cells and stromal cells, express and secrete copious amounts of BMP4 in response to irradiation, which causes the bone marrow stromal cells to commit to adipocyte lineage, thereby contributing to an increase in bone marrow adipogenesis. We further demonstrate that Simvastatin inhibits the BMP4-mediated adipogenic commitment of marrow stromal cells by inhibiting Ppar-γ expression. Importantly, Simvastatin does not prevent BMP4 secretion by the BM cells, and thus does not interfere with its salutary role in post-transplant hematopoietic regeneration. Our data identify previously unknown mechanisms operative in marrow adipogenesis post-myeloablation. They also reveal the molecular mechanisms behind the advantage of using Simvastatin as a niche-targeting agent to improve HSC engraftment.


Assuntos
Adipogenia/efeitos da radiação , Células da Medula Óssea/efeitos da radiação , Proteína Morfogenética Óssea 4/metabolismo , Adipócitos/citologia , Adipócitos/efeitos da radiação , Adipogenia/fisiologia , Animais , Células da Medula Óssea/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/biossíntese , Taxa Secretória/efeitos dos fármacos , Taxa Secretória/efeitos da radiação , Sinvastatina/farmacologia , Células Estromais/metabolismo , Células Estromais/efeitos da radiação , Linfócitos T/metabolismo , Linfócitos T/efeitos da radiação , Irradiação Corporal Total
12.
Sci Rep ; 5: 15784, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26507009

RESUMO

Mesenchymal stem cells (MSCs) show immunoregulatory properties. Here, we compared MSCs obtained from placenta (P-MSCs) and umbilical cord (C-MSCs) from the same donor, for their immunomodulatory efficacy. P-MSCs and C-MSCs showed similar morphology and phenotypic profile, but different clonogenic ability. Importantly, they showed a significant difference in their immunosuppressive properties as assessed in mixed leukocyte reaction (MLR). The P-MSCs affected the antigen presenting ability of mononuclear cells (MNCs) and dendritic cells (DCs) significantly as compared to C-MSCs resulting in a reduced T-cell proliferation. P-MSC conditioned medium (CM) showed a significant reduction in T cell proliferation as compared to C-MSC CM, thus suggesting that a cell to cell contact is not essential. We found increased levels of IL-10 and TGFß1 and reduction in levels of IFNγ in P-MSC MLRs as compared to C-MSC MLRs. Furthermore, the CD3(+) CD4(+) CD25(+) T regulatory cells were enriched in case of P-MSCs in both, MSC-MNC and MSC-DC co-cultures. This observation was further supported by increased mRNA expression of FoxP3 in P-MSCs. Presently, cord-derived MSCs are being employed in transplantation therapies parallel to the bone marrow-derived MSCs. Our findings suggest that P-MSCs can be a better alternative to C-MSCs, to provide aid in immunological ailments.


Assuntos
Células-Tronco Mesenquimais/imunologia , Placenta/imunologia , Cordão Umbilical/imunologia , Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Sangue Fetal/imunologia , Fatores de Transcrição Forkhead/imunologia , Humanos , Interleucina-10/imunologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Gravidez , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/imunologia
13.
Stem Cells Dev ; 24(20): 2423-36, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26107807

RESUMO

The bone marrow (BM) microenvironment or the hematopoietic stem cell (HSC) niche is normally hypoxic, which maintains HSC quiescence. Paradoxically, transplanted HSCs rapidly proliferate in this niche. Pretransplant myelosuppression results in a substantial rise in oxygen levels in the marrow microenvironment due to reduced cellularity and consequent low oxygen consumption. Therefore, it may be construed that the rapid proliferation of the engrafted HSCs in the BM niche is facilitated by the transiently elevated oxygen tension in this milieu during the "engraftment window." To determine whether oxygen tension dominantly affects the regeneration of hematopoiesis in the BM niche, we created an "oxygen-independent hypoxic niche" by treating BM-derived mesenchymal stromal cells (BMSCs) with a hypoxia-mimetic compound, cobalt chloride (CoCl2) and cocultured them with BM-derived HSC-enriched cells under normoxic conditions (HSCs; CoCl2-cocultures). Cocultures with untreated BMSCs incubated under normoxia (control- cocultures) or hypoxia (1% O2; hypoxic-cocultures) were used as comparators. Biochemical analyses showed that though, both CoCl2 and hypoxia evoked comparable signals in the BMSCs, the regeneration of hematopoiesis in their respective cocultures was radically different. The CoCl2-BMSCs supported robust hematopoiesis, while the hypoxic-BMSCs exerted strong inhibition. The hematopoiesis-supportive ability of CoCl2-BMSCs was abrogated if the CoCl2-cocultures were incubated under hypoxia, demonstrating that the prevalent oxygen tension in the milieu dominantly affects the outcome of the HSC-BM niche interactions. Our data suggest that pharmacologically delaying the reestablishment of hypoxia in the BM may boost post-transplant regeneration of hematopoiesis.


Assuntos
Células da Medula Óssea/citologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Hiperemia/metabolismo , Oxigênio/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Medula Óssea/metabolismo , Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Regeneração/fisiologia
14.
Syst Synth Biol ; 9(1-2): 1-10, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25972984

RESUMO

Transforming growth factor-betas (TGF-ßs) and their family members that include bone morphogenic proteins and activins have been implicated in the regulation of proliferation, hibernation, quiescence and differentiation of hematopoietic stem cells (HSCs). Increasing evidence suggests that the superfamily of TGF-ßs play an integral role in the intercellular cross-talk between the stem cells and their microenvironment as well as within the cells at an intracellular level. Active sites of hematopoiesis, such as fetal liver and bone marrow are known to have abundant presence of TGF-ß indicating their importance in the maintenance and regulation of hematopoiesis. One of the striking features of TGF-ß superfamily is the variety of effects they evoke, contingent on the developing history of the responding cells. In the present review, we discuss the Smad-dependent and Smad-independent TGF-ß signaling pathways in order to understand and underscore their role in the regulation of HSCs.

16.
PLoS One ; 8(7): e69960, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922871

RESUMO

Diabetes mellitus (DM)-induced endothelial progenitor cell (EPC) dysfunction causes impaired wound healing, which can be rescued by delivery of large numbers of 'normal' EPCs onto such wounds. The principal challenges herein are (a) the high number of EPCs required and (b) their sustained delivery onto the wounds. Most of the currently available scaffolds either serve as passive devices for cellular delivery or allow adherence and proliferation, but not both. This clearly indicates that matrices possessing both attributes are 'the need of the day' for efficient healing of diabetic wounds. Therefore, we developed a system that not only allows selective enrichment and expansion of EPCs, but also efficiently delivers them onto the wounds. Murine bone marrow-derived mononuclear cells (MNCs) were seeded onto a PolyCaprolactone-Gelatin (PCG) nano-fiber matrix that offers a combined advantage of strength, biocompatibility wettability; and cultured them in EGM2 to allow EPC growth. The efficacy of the PCG matrix in supporting the EPC growth and delivery was assessed by various in vitro parameters. Its efficacy in diabetic wound healing was assessed by a topical application of the PCG-EPCs onto diabetic wounds. The PCG matrix promoted a high-level attachment of EPCs and enhanced their growth, colony formation, and proliferation without compromising their viability as compared to Poly L-lactic acid (PLLA) and Vitronectin (VN), the matrix and non-matrix controls respectively. The PCG-matrix also allowed a sustained chemotactic migration of EPCs in vitro. The matrix-effected sustained delivery of EPCs onto the diabetic wounds resulted in an enhanced fibrosis-free wound healing as compared to the controls. Our data, thus, highlight the novel therapeutic potential of PCG-EPCs as a combined 'growth and delivery system' to achieve an accelerated fibrosis-free healing of dermal lesions, including diabetic wounds.


Assuntos
Diabetes Mellitus Experimental/patologia , Células Endoteliais/citologia , Gelatina/farmacologia , Poliésteres/farmacologia , Células-Tronco/citologia , Alicerces Teciduais/química , Cicatrização/efeitos dos fármacos , Animais , Materiais Biocompatíveis/farmacologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Modelos Animais de Doenças , Fibrose , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Adesões Focais/ultraestrutura , Masculino , Teste de Materiais , Camundongos , Transplante de Células-Tronco , Sus scrofa
17.
PLoS One ; 7(11): e50153, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23185562

RESUMO

HT1080 - a human fibrosarcoma-derived cell line - forms aggressive angiogenic tumours in immuno-compromised mice. In spite of its extensive use as a model of tumour angiogenesis, the molecular event(s) initiating the angiogenic program in these cells are not known. Since hypoxia stimulates tumour angiogenesis, we examined the hypoxia-induced events evoked in these cells. In contrast to cells grown under normoxic conditions, hypoxia-primed (1% O(2)) HT1080 cells formed robust tubules on growth factor-reduced matrigel and formed significantly larger tumours in xenograft models in a chetomin-sensitive manner, indicating the role of HIF-1α-mediated transcription in these processes. Immuno-histochemical analyses of tumours formed by GFP-expressing HT1080 cells clearly showed that the tumour cells themselves expressed various angiogenic markers including Neuropilin-1 (NRP-1) and formed functional vessels containing red blood cells, thereby unambiguously demonstrating the vasculogenic mimicry of HT1080 cells in vivo. Experiments performed with the HT1080 cells stably transfected with plasmid constructs expressing shNRP-1 or full-length NRP-1 clearly established that the HIF1α-mediated up-regulation of NRP-1 played a deterministic role in the process. Hypoxia-exposure resulted in an up-regulation of c-Myc and OCT3/4 and a down-regulation of KLF4 mRNAs, suggesting their involvement in the tumour formation and angiogenesis. However, silencing of NRP-1 alone, though not affecting proliferation in culture, was sufficient to abrogate the tumour formation completely; clearly establishing that the hypoxia-mediated HIF-1α-dependent up-regulation of NRP-1 is a critical molecular event involved in the vasculogenic mimicry and tumor formation by HT1080 cells in vivo.


Assuntos
Fibrossarcoma/irrigação sanguínea , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias de Tecido Fibroso/irrigação sanguínea , Neuropilina-1/genética , Oxigênio/farmacologia , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Dissulfetos/farmacologia , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Inativação Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Alcaloides Indólicos/farmacologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neoplasias de Tecido Fibroso/genética , Neoplasias de Tecido Fibroso/metabolismo , Neovascularização Patológica , Neuropilina-1/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Plasmídeos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cytotherapy ; 14(3): 366-80, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22250991

RESUMO

BACKGROUND AIMS: Previous data have shown that the addition of docosahexanoic acid (DHA)/arachidonic acid (AA) has a beneficial effect on cytokine-mediated in vitro generation of megakaryocytes (MK) from umbilical cord blood (UCB).Cryopreservation forms an inherent part of UCB banking and MK progenitors are known to be very sensitive to the stresses of freezing. It is therefore imperative to generate functional cells from cryopreserved cells, and the generated cells need to be cryopreserved until used. In the present study, cryopreservation of ex vivo-expanded MK as well as MK generation from cryopreserved UCB samples was investigated. METHODS: MK generated with or without DHA/AA were cryopreserved in freezing medium containing 10% dimethyl sulfoxide (DMSO). Freezing efficacy was tested by quantitating MK after revival. Cryopreserved CD34(+) cells were cultured with stem cell factor (SCF) and thrombopoietin (TPO), in the presence and absence of DHA/AA for 10 days, and then quantitated for MK. Results. We observed a 1.5-3-fold increase in MK numbers, their progenitor content and their expression of phenotypic markers and MK-related transcription factors. DHA/AA sets showed a 2-5-fold improved engraftment in NOD/SCID mice. These data showed that the beneficial effect of DHA/AA obtained during MK expansion was not altered after freezing stress. The enhancement in MK generation obtained from fresh cord blood (CB) cells was reproduced with comparable efficiency when we used cryopreserved CB samples. CONCLUSIONS: Taken together, our data suggest that in vitro-generated DHA/AA MK survive cryoinjuries in a functionally better state. DHA/AA support a more efficient generation of MK from cryopreserved UCB.


Assuntos
Ácido Araquidônico/farmacologia , Criopreservação/métodos , Crioprotetores/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Sangue Fetal/efeitos dos fármacos , Megacariócitos/citologia , Animais , Antígenos CD34/química , Apoptose , Preservação de Sangue/métodos , Contagem de Células , Técnicas de Cultura de Células/métodos , Células Cultivadas , Dimetil Sulfóxido/farmacologia , Sangue Fetal/citologia , Congelamento , Humanos , Camundongos , Camundongos SCID , Trombopoetina/química , Fatores de Transcrição/química , Túnica Média/química
19.
PLoS One ; 7(1): e29383, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22235291

RESUMO

BACKGROUND: Expansion of hematopoietic stem/progenitor cells (HSPCs) is a well-known strategy employed to facilitate the transplantation outcome. We have previously shown that the prevention of apoptosis by the inhibition of cysteine proteases, caspase and calpain played an important role in the expansion and engraftment of cord blood (CB) derived HSPCs. We hypothesize that these protease inhibitors might have maneuvered the adhesive and migratory properties of the cells rendering them to be retained in the bone marrow for sustained engraftment. The current study was aimed to investigate the mechanism of the homing responses of CB cells during expansion. METHODOLOGY/PRINCIPAL FINDINGS: CB derived CD34(+) cells were expanded using a combination of growth factors with and without Caspase inhibitor -zVADfmk or Calpain 1 inhibitor- zLLYfmk. The cells were analyzed for the expression of homing-related molecules. In vitro adhesive/migratory interactions and actin polymerization dynamics of HSPCs were assessed. In vivo homing assays were carried out in NOD/SCID mice to corroborate these observations. We observed that the presence of zVADfmk or zLLYfmk (inhibitors) caused the functional up regulation of CXCR4, integrins, and adhesion molecules, reflecting in a higher migration and adhesive interactions in vitro. The enhanced actin polymerization and the RhoGTPase protein expression complemented these observations. Furthermore, in vivo experiments showed a significantly enhanced homing to the bone marrow of NOD/SCID mice. CONCLUSION/SIGNIFICANCE: Our present study reveals another novel aspect of the regulation of caspase and calpain proteases in the biology of HSPCs. The priming of the homing responses of the inhibitor-cultured HSPCs compared to the cytokine-graft suggests that the modulation of these proteases may help in overcoming the major homing defects prevalent in the expansion cultures thereby facilitating the manipulation of cells for transplant procedures.


Assuntos
Calpaína/antagonistas & inibidores , Inibidores de Caspase , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Actinas/química , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Integrina alfa4/metabolismo , Integrina alfa5/metabolismo , Camundongos , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
20.
Haematologica ; 97(5): 651-60, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22058199

RESUMO

BACKGROUND: A culture system that closely recapitulates marrow physiology is essential to study the niche-mediated regulation of hematopoietic stem cell fate at a molecular level. We investigated the key features that play a crucial role in the formation of a functional niche in vitro. DESIGN AND METHODS: Hydrogel-based cultures of human placenta-derived mesenchymal stromal cells were established to recapitulate the fibrous three-dimensional architecture of the marrow. Plastic-adherent mesenchymal stromal cells were used as controls. Human bone marrow-derived CD34(+) cells were co-cultured with them. The output hematopoietic cells were characterized by various stem cell-specific phenotypic and functional parameters. RESULTS: The hydrogel-cultures harbored a large pool of primitive hematopoietic stem cells with superior phenotypic and functional attributes. Most importantly, like the situation in vivo, a significant fraction of these cells remained quiescent in the face of a robust multi-lineage hematopoiesis. The retention of a high percentage of primitive stem cells by the hydrogel-cultures was attributed to the presence of CXCR4-SDF1α axis and integrin beta1-mediated adhesive interactions. The hydrogel-grown mesenchymal stromal cells expressed high levels of several molecules that are known to support the maintenance of hematopoietic stem cells. Yet another physiologically relevant property exhibited by the hydrogel cultures was the formation of hypoxia-gradient. Destruction of hypoxia-gradient by incubating these cultures in a hypoxia chamber destroyed their specialized niche properties. CONCLUSIONS: Our data show that hydrogel-based cultures of mesenchymal stromal cells form a functional in vitro niche by mimicking key features of marrow physiology.


Assuntos
Biomarcadores/metabolismo , Medula Óssea/fisiologia , Técnicas de Cultura de Células , Células-Tronco Hematopoéticas/fisiologia , Hidrogel de Polietilenoglicol-Dimetacrilato , Células-Tronco Mesenquimais/fisiologia , Nicho de Células-Tronco , Animais , Antígenos CD34/metabolismo , Western Blotting , Ciclo Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Hematopoese , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise de Sequência com Séries de Oligonucleotídeos , Placenta/citologia , Placenta/metabolismo , Gravidez , Receptores CXCR4/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA