Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1866(4): 165314, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30412793

RESUMO

Osteoporosis is a silent systemic disease that causes bone deterioration, and affects over 10 million people in the US alone. This study was undertaken to develop a potential stem cell therapy for osteoporosis. We have isolated and expanded human dental pulp-derived stem cells (DPSCs), characterized them, and confirmed their multipotential differentiation abilities. Stem cells often remain quiescent and require activation to differentiate and function. Herein, we show that ferutinin activates DPSCs by modulating the Wnt/ß-catenin signaling pathway and key osteoblast-secreted proteins osteocalcin and collagen 1A1 both mRNA and protein levels. To confirm that ferutinin modulates the Wnt pathway, we inhibited glycogen synthase kinase 3 (GSK3) and found that protein expression patterns were similar to those found in ferutinin-treated DPSCs. To evaluate the role of ferutinin in epigenetic regulation of canonical Wnt signaling, the pathway molecules Wnt3a and Dvl3 were analyzed using chromatin immunoprecipitation (ChIP)-quantitative PCR approaches. We confirmed that active marks of both H3K9 acetylation and H3K4 trimethylation were significantly enhanced in the promoter sites of the WNT3A and DVL3 genes in DPSCs after addition of ferutinin. These data provide evidence that ferutinin activates and promotes osteogenic differentiation of DPSCs, and could be used as an inducer as a potentially effective stem cell therapy for osteoporosis.


Assuntos
Benzoatos/farmacologia , Cicloeptanos/farmacologia , Polpa Dentária/metabolismo , Epigênese Genética/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Sesquiterpenos/farmacologia , Células-Tronco/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Compostos Bicíclicos com Pontes/farmacologia , Polpa Dentária/citologia , Humanos , Células-Tronco/citologia
3.
Cells ; 8(8)2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31426355

RESUMO

Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease, and Krüppel-like factor 2 (KLF2) regulates immune cell activation and function. Herein, we show that in our experiments 50% global deficiency of KLF2 significantly elevated arthritic inflammation and pathogenesis, osteoclastic differentiation, matrix metalloproteinases (MMPs), and inflammatory cytokines in K/BxN serum-induced mice. The severities of RA pathogenesis, as well as the causative and resultant cellular and molecular factors, were further confirmed in monocyte-specific KLF2 deficient mice. In addition, induction of RA resulted in a decreased level of KLF2 in monocytes isolated from both mice and humans along with higher migration of activated monocytes to the RA sites in humans. Mechanistically, overexpression of KLF2 decreased the level of MMP9; conversely, knockdown of KLF2 increased MMP9 in monocytes along with enrichment of active histone marks and histone acetyltransferases on the MMP9 promoter region. These findings define the critical regulatory role of myeloid KLF2 in RA pathogenesis.


Assuntos
Artrite Reumatoide/imunologia , Inflamação/imunologia , Fatores de Transcrição Kruppel-Like/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Monócitos/imunologia , Animais , Artrite Experimental/imunologia , Diferenciação Celular , Células Cultivadas , Citocinas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/metabolismo
4.
Sci Rep ; 9(1): 8415, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182750

RESUMO

Despite advances in diabetic wound care, the significant number of amputations that occur every year demands more effective therapeutics. Herein, we offer an aminated polyethersulfone nanofiber-expanded human umbilical cord blood-derived CD34+ cells (henceforth CD34+ cells) effective therapy, tested in cutaneous wounds developed in streptozotocin-induced diabetic NOD/SCID mice. We show that systemic administration of CD34+ cells homed to the wound site and significantly accelerated wound closure. Wound closure was associated with improved re-epithelialization and increased neovascularization; and with decreased sustained pro-inflammatory activity of NF-κB and its downstream effector molecules TNF-α, IL-1ß, and IL-6 at the wound bed. This finding was further supported by the observation of a decreased number of myeloperoxidase positive neutrophils, and concomitantly increased levels of IL-10. In addition, improved granulation tissue formation was observed along with higher collagen deposition and myofibroblasts and decreased expressions of MMP-1. Mechanistically, CD34+ cells reduced the level of MMP-1 expression by inhibiting recruitment of NF-κB to the MMP-1 promoter site in dermal fibroblasts. In summary, we provide evidence of a novel nanofiber-expanded CD34+ stem cell therapeutic development for treating diabetic wounds by defining their cellular and molecular mechanisms.


Assuntos
Antígenos CD34/metabolismo , Diabetes Mellitus Experimental/patologia , Nanofibras/química , Pele/patologia , Cicatrização , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Derme/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Sangue Fetal/citologia , Fibroblastos/efeitos dos fármacos , Tecido de Granulação/patologia , Humanos , Inflamação/patologia , Metaloproteinases da Matriz/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , NF-kappa B/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Estreptozocina , Fator de Necrose Tumoral alfa/farmacologia , Cicatrização/efeitos dos fármacos
5.
J Cell Mol Med ; 23(2): 1386-1395, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30506878

RESUMO

Krüppel-like factor 2 (KLF2) critically regulates activation and function of monocyte, which plays important pathogenic role in progressive joint destruction in rheumatoid arthritis (RA). It is yet to be established the molecular basis of KLF2-mediated regulation of monocytes in RA pathogenesis. Herein, we show that a class of compound, HDAC inhibitors (HDACi) induced KLF2 expression in monocytes both in vitro and in vivo. KLF2 level was also elevated in tissues, such as bone marrow, spleen and thymus in mice after infusion of HDACi. Importantly, HDACi significantly reduced osteoclastic differentiation of monocytes with the up-regulation of KLF2 and concomitant down-regulation of matrixmetalloproteinases both in the expression level as well as in the protein level. In addition, HDACi reduced K/BxN serum-induced arthritic inflammation and joint destruction in mice in a dose-dependent manner. Finally, co-immunoprecipitation and overexpression studies confirmed that KLF2 directly interacts with HDAC4 molecule in cells. These findings provide mechanistic evidence of KLF2-mediated regulation of K/BxN serum-induced arthritic inflammation.


Assuntos
Artrite Experimental/prevenção & controle , Artrite Reumatoide/prevenção & controle , Inibidores de Histona Desacetilases/farmacologia , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Artrite Experimental/etiologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Artrite Reumatoide/etiologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Diferenciação Celular , Feminino , Histona Desacetilases/química , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Células RAW 264.7
6.
Methods Mol Biol ; 1213: 209-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25173385

RESUMO

To evaluate therapeutic efficacy and to investigate involved molecular mechanisms of cell-based therapy in osteoporosis, the generation of a clinically relevant model is critically important. Herein, we describe detailed methods in generation of an immune-deficient osteoporotic murine model, and application of human umbilical cord blood-derived stem cells to assess their therapeutic efficacy.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Osteoporose/terapia , Transplante de Células-Tronco , Animais , Antígenos CD34/metabolismo , Modelos Animais de Doenças , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Hospedeiro Imunocomprometido , Camundongos , Osteoporose/etiologia
7.
Biomaterials ; 35(30): 8566-75, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25002260

RESUMO

Despite recent advances in cardiovascular medicine, ischemic diseases remain a major cause of morbidity and mortality. Although stem cell-based therapies for the treatment of ischemic diseases show great promise, limited availability of biologically functional stem cells mired the application of stem cell-based therapies. Previously, we reported a PES-nanofiber based ex vivo stem cell expansion technology, which supports expansion of human umbilical cord blood (UCB)-derived CD133(+)/CD34(+) progenitor cells ∼225 fold. Herein, we show that using similar technology and subsequent re-expansion methods, we can achieve ∼5 million-fold yields within 24 days of the initial seeding. Interestingly, stem cell phenotype was preserved during the course of the multiple expansions. The high level of the stem cell homing receptor, CXCR4 was expressed in the primary expansion cells, and was maintained throughout the course of re-expansions. In addition, re-expanded cells preserved their multi-potential differential capabilities in vitro, such as, endothelial and smooth muscle lineages. Moreover, biological functionality of the re-expanded cells was preserved and was confirmed by a murine hind limb ischemia model for revascularization. These cells could also be genetically modified for enhanced vasculogenesis. Immunohistochemical evidences support enhanced expression of angiogenic factors responsible for this enhanced neovascularization. These data further confirms that nanofiber-based ex-vivo expansion technology can generate sufficient numbers of biologically functional stem cells for potential clinical applications.


Assuntos
Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/fisiologia , Nanofibras/química , Neovascularização Fisiológica/efeitos dos fármacos , Polímeros/farmacologia , Células-Tronco/citologia , Sulfonas/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Isquemia/patologia , Isquemia/fisiopatologia , Camundongos SCID , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Nanofibras/ultraestrutura , Fenótipo , Células-Tronco/efeitos dos fármacos , Células-Tronco/ultraestrutura
8.
Biochim Biophys Acta ; 1842(7): 1071-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24726882

RESUMO

Many ovarian cancer cells express stress-related molecule MICA/B on their surface that is recognized by Vγ2Vδ2 T cells through their NKG2D receptor, which is transmitted to downstream stress-signaling pathway. However, it is yet to be established how Vγ2Vδ2 T cell-mediated recognition of MICA/B signal is transmitted to downstream stress-related molecules. Identifying targeted molecules would be critical to develop a better therapy for ovarian cancer cells. It is well established that ATM/ATR signal transduction pathways, which is modulated by DNA damage, replication stress, and oxidative stress play central role in stress signaling pathway regulating cell cycle checkpoint and apoptosis. We investigated whether ATM/ATR and its down stream molecules affect Vγ2Vδ2 T cell-mediated cytotoxicity. Herein, we show that ATM/ATR pathway is modulated in ovarian cancer cells in the presence of Vγ2Vδ2 T cells. Furthermore, downregulation of ATM pathway resulted downregulation of MICA, and reduced Vγ2Vδ2 T cell-mediated cytotoxicity. Alternately, stimulating ATM pathway enhanced expression of MICA, and sensitized ovarian cancer cells for cytotoxic lysis by Vγ2Vδ2 T cells. We further show that combining currently approved chemotherapeutic drugs, which induced ATM signal transduction, along with Vγ2Vδ2 T cells enhanced cytotoxicity of resistant ovarian cancer cells. These findings indicate that ATM/ATR pathway plays an important role in tumor recognition, and drugs promoting ATM signaling pathway might be considered as a combination therapy together with Vγ2Vδ2 T cells for effectively treating resistant ovarian cancer cells.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/imunologia , Neoplasias Ovarianas/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
9.
J Cell Mol Med ; 18(4): 685-97, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24455991

RESUMO

Nanofiber-expanded human umbilical cord blood-derived CD34(+) cell therapy has been shown to have potential applications for peripheral and myocardial ischaemic diseases. However, the efficacies of expanded CD34(+) cell therapy for treating cutaneous wounds and its mechanisms of action have yet to be established. Using an excisional wound model in non-obese diabetic/severe combined immune deficient mice, we show herein that CD34(+) cells accelerate the wound-healing process by enhancing collagen synthesis, and increasing fibroblast cell migration within the wound bed. Concomitantly, reduced levels of matrix metalloproteinase (MMPs) such as MMP1, MMP3, MMP9 and MMP13 were detected in the wound beds of animals treated with CD34(+) cells compared with vehicle-treated controls. CD34(+) cells were found to mediate enhanced migration and proliferation of dermal fibroblast cells in vitro. Moreover, CD34(+) cells secrete collagen in a serum-deprived environment. In mechanistic studies, co-culture of CD34(+) cells with primary skin fibroblasts increased the expression of collagen1A1, a component of type 1 collagen, and decreased the expression of MMP1 in fibroblast cells in the presence of a proteasome inhibitor. Finally, CD34(+) cell-mediated functions were transcriptionally regulated by the c-Jun N-terminal kinases pathway. Collectively, these data provide evidence of therapeutic efficacy and a novel mechanism of nanofiber-expanded CD34(+) cell-mediated accelerated wound healing.


Assuntos
Antígenos CD34/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Sangue Fetal/transplante , Transplante Heterólogo , Animais , Sangue Fetal/citologia , Humanos , Camundongos , Nanofibras/administração & dosagem , Nanofibras/química , Pele/patologia , Técnicas de Fechamento de Ferimentos , Cicatrização
10.
Stem Cell Res ; 12(1): 275-88, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24321844

RESUMO

Nanofiber-expanded human umbilical cord blood-derived CD34+ cell therapy is under consideration for treating peripheral and cardiac ischemia. However, the therapeutic efficacy of nanofiber-expanded human umbilical cord blood-derived (NEHUCB) CD34+ cell therapy for wound healing and its mechanisms are yet to be established. Using an excision wound model in NOD/SCID mice, we show herein that NEHUCB-CD34+ cells home to the wound site and significantly accelerate the wound-healing process compared to vehicle-treated control. Histological analysis reveals that accelerated wound closure is associated with the re-epithelialization and increased angiogenesis. Additionally, NEHUCB-CD34+ cell-therapy decreases expression of pro-inflammatory cytokines, such as TNF-α, IL-1ß, IL-6 and NOS2A in the wound bed, and concomitantly increases expression of IL-10 compared to vehicle-treated control. These findings were recapitulated in vitro using primary dermal fibroblasts and NEHUCB-CD34+ cells. Moreover, NEHUCB-CD34+ cells attenuate NF-κB activation and nuclear translocation in dermal fibroblasts through enhanced secretion of IL-10, which is known to bind to NF-κB and suppress transcriptional activity. Collectively, these data provide novel mechanistic evidence of NEHUCB-CD34+ cell-mediated accelerated wound healing.


Assuntos
Sangue Fetal/citologia , Interleucina-10/metabolismo , Dermatopatias/terapia , Células-Tronco/citologia , Cicatrização , Animais , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Humanos , Interleucina-10/genética , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Nanofibras/química , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Dermatopatias/metabolismo , Dermatopatias/patologia , Transplante de Células-Tronco , Células-Tronco/química , Fatores de Transcrição/metabolismo
11.
Front Biosci (Landmark Ed) ; 18(3): 970-81, 2013 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-23747860

RESUMO

Because of their ability for self-renewal and neural differentiation, stem cells are believed to be ideal for cell replacement therapy in Parkinson's disease (PD). Nanofiber-expanded human umbilical cord hematopoietic stem cells (HUHSCs) are advantageous to other stem cells as they provide a source of unlimited stem cell production for clinical application. In this study, we investigated whether 1. nanofiber-expanded HUHSCs are capable of neural differentiation in vitro, and 2. they could improve dopaminergic neuron morphology in the caudate/putamen (CPu) and substantia nigra pars compacta (SNc) of the MPTP-mouse model of PD. When cultured under neural differentiation conditions, nanofiber-expanded HUHSCs were able to undergo neural differentiation in vitro, as determined by gene and protein expression of neural markers such as MAP2, NeuN, HuC, GFAP and Oligo2. Thirty days after a single intracardioventricular injection of HUHSCs to MPTP-mice there was a significant recovery of tyrosine hydroxylase (TH) immunostaining in CPu. There was an increase in the size and staining density of TH+ cells in SNc, while their number was unchanged.


Assuntos
Dopamina/metabolismo , Células-Tronco Hematopoéticas/citologia , Neurônios/metabolismo , Transtornos Parkinsonianos/terapia , Animais , Sequência de Bases , Primers do DNA , Modelos Animais de Doenças , Camundongos , Reação em Cadeia da Polimerase
12.
PLoS One ; 8(4): e61398, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23620747

RESUMO

Molecular-focused cancer therapies, e.g., molecularly targeted therapy and immunotherapy, so far demonstrate only limited efficacy in cancer patients. We hypothesize that underestimating the role of biophysical factors that impact the delivery of drugs or cytotoxic cells to the target sites (for associated preferential cytotoxicity or cell signaling modulation) may be responsible for the poor clinical outcome. Therefore, instead of focusing exclusively on the investigation of molecular mechanisms in cancer cells, convection-diffusion of cytotoxic molecules and migration of cancer-killing cells within tumor tissue should be taken into account to improve therapeutic effectiveness. To test this hypothesis, we have developed a mathematical model of the interstitial diffusion and uptake of small cytotoxic molecules secreted by T-cells, which is capable of predicting breast cancer growth inhibition as measured both in vitro and in vivo. Our analysis shows that diffusion barriers of cytotoxic molecules conspire with γδ T-cell scarcity in tissue to limit the inhibitory effects of γδ T-cells on cancer cells. This may increase the necessary ratios of γδ T-cells to cancer cells within tissue to unrealistic values for having an intended therapeutic effect, and decrease the effectiveness of the immunotherapeutic treatment.


Assuntos
Apoptose , Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Imunoterapia , Adulto , Animais , Neoplasias da Mama/patologia , Comunicação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Difusão , Feminino , Humanos , Interferon gama/metabolismo , Camundongos , Modelos Biológicos , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Resultado do Tratamento
13.
Int J Cancer ; 133(9): 2133-44, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23595559

RESUMO

Innate immune system has been known to play an important role in inhibiting the malignant transformation, tumor progression and invasion. However, the mechanistic basis remains ambiguous. Despite polyclonality of human γδ T cells, Vγ2Vδ2 T cell subset was shown to recognize and limit the growth of various tumors at various degrees. The differential recognition of the tumor cells by Vγ2Vδ2 T cells are yet to be defined. Our study reveals that γδ T cells limit in vitro growth of most breast tumor cells, such as SkBr7 (HER2+), MCF7 (ER+) and MDA-MB-231 (ER-) by inhibiting their survival and inducing apoptosis, except BrCa-MZ01 (PR+) cells. To investigate detail mechanisms of antineoplastic effects, we found that cell death was associated with the surface expression levels of MICA/B and ICAM1. Molecular signaling analysis demonstrated that inhibition of cell growth by γδ T cells was associated with the lower expression levels of cell survival-related molecules such as AKT, ERK and concomitant upregulation of apoptosis-related molecules, such as PARP, cleaved caspase 3 and tumor suppressor genes PTEN and P53. However, opposite molecular signaling was observed in the resistant cell line after coculture with γδ T cells. In vivo, antineoplastic effects of γδ T cells were also documented, where tumor growth was inhibited due to the downregulation of survival signals, strong induction of apoptotic molecules, disruption of microvasculature and increased infiltration of tumor associated macrophages. These findings reveal that a complex molecular signaling is involved in γδ T cell-mediated antineoplastic effects.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Subpopulações de Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Western Blotting , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Ciclo Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Células Tumorais Cultivadas
14.
PLoS One ; 7(6): e39365, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22724005

RESUMO

BACKGROUND: Osteoporosis is a bone disorder associated with loss of bone mineral density and micro architecture. A balance of osteoblasts and osteoclasts activities maintains bone homeostasis. Increased bone loss due to increased osteoclast and decreased osteoblast activities is considered as an underlying cause of osteoporosis. METHODS AND FINDINGS: The cures for osteoporosis are limited, consequently the potential of CD34+ cell therapies is currently being considered. We developed a nanofiber-based expansion technology to obtain adequate numbers of CD34(+) cells isolated from human umbilical cord blood, for therapeutic applications. Herein, we show that CD34(+) cells could be differentiated into osteoblastic lineage, in vitro. Systemically delivered CD34(+) cells home to the bone marrow and significantly improve bone deposition, bone mineral density and bone micro-architecture in osteoporotic mice. The elevated levels of osteocalcin, IL-10, GM-CSF, and decreased levels of MCP-1 in serum parallel the improvements in bone micro-architecture. Furthermore, CD34(+) cells improved osteoblast activity and concurrently impaired osteoclast differentiation, maturation and functionality. CONCLUSIONS: These findings demonstrate a novel approach utilizing nanofiber-expanded CD34(+) cells as a therapeutic application for the treatment of osteoporosis.


Assuntos
Antígenos CD34 , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Células-Tronco Hematopoéticas/citologia , Osteoblastos/citologia , Osteoclastos/citologia , Osteoporose/terapia , Animais , Antígenos CD34/metabolismo , Medula Óssea/metabolismo , Osso e Ossos/ultraestrutura , Calcificação Fisiológica , Técnicas de Cultura de Células , Diferenciação Celular , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Osteoblastos/metabolismo , Osteocalcina/sangue , Osteoclastos/metabolismo , Osteogênese , Osteoporose/metabolismo
15.
PLoS One ; 6(9): e23348, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21935360

RESUMO

BACKGROUND: Mechanisms of human Vγ2Vδ2 T cell-mediated tumor immunity have yet to be fully elucidated. METHODS AND FINDINGS: At least some tumor cell recognition is mediated by NKG2D-MICA interactions. Herein, by using MTT assay and PI-BrdU co-staining and Western-blot, we show that these Vγ2Vδ2 T cells can limit the proliferation of ovarian tumor cells by down regulation of apoptosis and cell cycle related molecules in tumor cells. Cell-to-cell contact is critical. γδ T cell-resistant, but not susceptible ovarian tumor cells escape γδ T cell-mediated immune recognition by up-regulating pErk1/2, thereby decreasing surface MICA levels. Erk1/2 inhibitor pretreatment or incubation prevents this MICA decrease, while up-regulating key cell cycle related molecules such as CDK2, CDK4 and Cyclin D1, as well as apoptosis related molecules making resistant tumor cells now vulnerable to γδ T cell-mediated lysis. CONCLUSION: These findings demonstrate novel effects of γδT cells on ovarian tumor cells.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/biossíntese , Neoplasias Ovarianas/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Apoptose , Comunicação Celular , Ciclo Celular , Ciclina D1/biossíntese , Quinase 2 Dependente de Ciclina/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Regulação para Baixo , Feminino , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/metabolismo , Linfócitos T/imunologia , Regulação para Cima
16.
Front Biosci (Landmark Ed) ; 15(3): 854-71, 2010 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-20515730

RESUMO

Stem cell therapy is currently considered as an important regime for repairing, replacing or enhancing the biological functions of the damaged tissues. Among adult stem cells, hematopoietic stem cells (HSCs) are commonly used for cure of hematological disorders. However, the number of HSCs obtained from sources like bone marrow, peripheral or umbilical cord blood is not sufficient for routine clinical application. Thus, ex-vivo expansion of HSCs becomes critically important. Ex-vivo culture and expansion of stem cells are challenging, as stem cells differentiate in culture rather than self-renew. Lack of clarity about the factors responsible for quiescence and differentiation of HSCs, investigators struggled to optimize conditions for ex vivo expansion. As we understand better, various strategies can be incorporated to mimic in vivo conditions for successful expansion of stem cells. However, characterization and biological functionality should also be tested for expanded stem cells prior to clinical application. To treat ischemia by enhancing therapeutic angiogenesis and neo-vascularization, the role of genetic modification of HSCs with pro-angiogenic factors is the focus of this review.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Hematopoéticas/citologia , Transgenes/genética , Animais , Técnicas de Cultura de Células , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Transfecção/métodos
17.
Recent Pat Nanotechnol ; 4(2): 125-35, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20420564

RESUMO

Hematopoietic stem cell transplantation has been applied as a standard procedure of treatment for hematological disorders like multiple myeloma and leukemia for several decades. Various sources of stem cells like bone marrow, peripheral blood and umbilical cord blood are used for the transplantation. Among these umbilical cord blood is currently preferred due to the primitiveness of the derived stem cells and minimal possibilities of graft versus host disease or development of graft induced tumors. One of the problems for these sources is the procurement of sufficient number of donor stem cells. Inadequate number of cells may lead to delayed recovery and decrease survivability of the patient. Thus to overcome the limitation of stem cell number, development of an ex-vivo expansion technology is critically important. The recent emerging technology using nanofiber in combination with growth factors has made a significant improvement to the field of regenerative medicine and a couple of patents have been filed. In this review, we will focus on factors regulating hematopoietic stem cell self-renewal and expansion emphasizing on nanofiber as a supporting matrix.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Hematopoéticas , Peptídeos e Proteínas de Sinalização Intercelular/química , Nanofibras/química , Nanotecnologia/métodos , Sangue Fetal/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/administração & dosagem , Nanofibras/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...