Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 42(1): 13-28, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37931173

RESUMO

Insight into the molecular mechanisms governing the development and maintenance of pluripotency is important for understanding early development and the use of stem cells in regenerative medicine. We demonstrate the selective inhibition of mTORC1 signaling is important for developing the inner cell mass (ICM) and the self-renewal of human embryonic stem cells. S6K suppressed the expression and function of pluripotency-related transcription factors (PTFs) OCT4, SOX2, and KLF4 through phosphorylation and ubiquitin proteasome-mediated protein degradation, indicating that S6K inhibition is required for pluripotency. PTFs inhibited mTOR signaling. The phosphorylation of S6 was decreased in PTF-positive cells of the ICM in embryos. Activation of mTORC1 signaling blocked ICM formation and the selective inhibition of S6K by rapamycin increased the ICM size in mouse blastocysts. Thus, selective inhibition of mTORC1 signaling supports the development and maintenance of pluripotency.


Assuntos
Blastocisto , Transdução de Sinais , Humanos , Animais , Camundongos , Sirolimo/farmacologia , Fosforilação , Alvo Mecanístico do Complexo 1 de Rapamicina
2.
Prostate ; 80(13): 1145-1156, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32659025

RESUMO

BACKGROUND: Epithelial stem cells (ESCs) demonstrate a capacity to maintain normal tissues homeostasis and ESCs with a deregulated behavior can contribute to cancer development. The ability to reprogram normal tissue epithelial cells into prostate or mammary stem-like cells holds great promise to help understand cell of origin and lineage plasticity in prostate and breast cancers in addition to understanding normal gland development. We previously showed that an intracellular chemokine, CXCL12γ induced cancer stem cells and neuroendocrine characteristics in both prostate and breast adenocarcinoma cell lines. However, its role in normal prostate or mammary epithelial cell fate and development remains unknown. Therefore, we sought to elucidate the functional role of CXCL12γ in the regulation of ESCs and tissue development. METHODS: Prostate epithelial cells (PNT2) or mammary epithelial cells (MCF10A) with overexpressed CXCL12γ was characterized by quantitative real-time polymerase chain reaction, Western blots, and immunofluorescence for lineage marker expression, and fluorescence activated cell sorting analyses and sphere formation assays to examine stem cell surface phenotype and function. Xenotransplantation animal models were used to evaluate gland or acini formation in vivo. RESULTS: Overexpression of CXCL12γ promotes the reprogramming of cells with a differentiated luminal phenotype to a nonluminal phenotype in both prostate (PNT2) and mammary (MCF10A) epithelial cells. The CXCL12γ-mediated nonluminal type cells results in an increase of epithelial stem-like phenotype including the subpopulation of EPCAMLo /CD49fHi /CD24Lo /CD44Hi cells capable of sphere formation. Critically, overexpression of CXCL12γ promotes the generation of robust gland-like structures from both prostate and mammary epithelial cells in in vivo xenograft animal models. CONCLUSIONS: CXCL12γ supports the reprogramming of epithelial cells into nonluminal cell-derived stem cells, which facilitates gland development.


Assuntos
Quimiocina CXCL12/biossíntese , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Próstata/crescimento & desenvolvimento , Animais , Reprogramação Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Xenoenxertos , Humanos , Masculino , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Camundongos , Próstata/citologia , Próstata/metabolismo , Isoformas de Proteínas
3.
Stem Cells ; 37(6): 766-778, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30786091

RESUMO

Aberrant wound healing presents as inappropriate or insufficient tissue formation. Using a model of musculoskeletal injury, we demonstrate that loss of transforming growth factor-ß activated kinase 1 (TAK1) signaling reduces inappropriate tissue formation (heterotopic ossification) through reduced cellular differentiation. Upon identifying increased proliferation with loss of TAK1 signaling, we considered a regenerative approach to address insufficient tissue production through coordinated inactivation of TAK1 to promote cellular proliferation, followed by reactivation to elicit differentiation and extracellular matrix production. Although the current regenerative medicine paradigm is centered on the effects of drug treatment ("drug on"), the impact of drug withdrawal ("drug off") implicit in these regimens is unknown. Because current TAK1 inhibitors are unable to phenocopy genetic Tak1 loss, we introduce the dual-inducible COmbinational Sequential Inversion ENgineering (COSIEN) mouse model. The COSIEN mouse model, which allows us to study the response to targeted drug treatment ("drug on") and subsequent withdrawal ("drug off") through genetic modification, was used here to inactivate and reactivate Tak1 with the purpose of augmenting tissue regeneration in a calvarial defect model. Our study reveals the importance of both the "drug on" (Cre-mediated inactivation) and "drug off" (Flp-mediated reactivation) states during regenerative therapy using a mouse model with broad utility to study targeted therapies for disease. Stem Cells 2019;37:766-778.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Fraturas Ósseas/genética , MAP Quinase Quinase Quinases/genética , Células-Tronco Mesenquimais/enzimologia , Osteoblastos/enzimologia , Cicatrização/genética , Animais , Regeneração Óssea/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , DNA Nucleotidiltransferases/genética , DNA Nucleotidiltransferases/metabolismo , Feminino , Efeito Fundador , Fraturas Ósseas/tratamento farmacológico , Fraturas Ósseas/enzimologia , Fraturas Ósseas/patologia , Regulação da Expressão Gênica , Integrases/genética , Integrases/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/deficiência , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Crânio/efeitos dos fármacos , Crânio/lesões , Crânio/metabolismo , Cicatrização/efeitos dos fármacos
4.
Orthod Craniofac Res ; 22 Suppl 1: 192-198, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31074151

RESUMO

OBJECTIVES: Skeletal stem cells (SSCs) are characterized by expression of cell surface biomarkers and their ability to differentiate into bone, cartilage and fat. However, the current biomarkers used to identify these cell populations are not cell-type-specific or indicative of the differentiation status of these cells and are therefore unreliable. Our objective was to identify alternative cell surface biomarkers and transcription factors shared between SSCs isolated from the bone marrow (BM) and those derived from pluripotent stem cells (PSC). MATERIALS AND METHODS: Human PSCs were induced into SSCs. FACS and qRT-PCR were used to determine differences in expression of cell surface biomarkers and transcription factors between SSCs derived from PSCs and isolated from BM, in differentiating cells, in cells from early and late passage, and in fibroblasts. RESULTS: A significant reduction in proliferation and capacity of SSCs to differentiate into adipocytes and osteoblasts was observed after 3 passages. Protein and mRNA analysis indicated that commonly used biomarkers remain highly expressed in cells that lost capacity for differentiation. However, integrin α6 (CD49f) and transcription factors GATA6, PRDM16, SIM2 and SOX11 were significantly upregulated in SSCs compared to fibroblasts. In early stages of adipogenic and osteogenic differentiation, the expression of CD49f, GATA6 and SIM2 was reduced in later passage cells, which have limited proliferation and differentiation capabilities. CONCLUSIONS: Our results suggest that CD49f and transcription factors GATA6 and SIM2 identify functional SSCs.


Assuntos
Osteogênese , Células-Tronco , Adipogenia , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Biomarcadores , Diferenciação Celular , Células Cultivadas , Fator de Transcrição GATA6 , Humanos
5.
Genesis ; 56(6-7): e23220, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-30134066

RESUMO

Craniosynostosis is defined as congenital premature fusion of one or more cranial sutures. While the genetic basis for about 30% of cases is known, the causative genes for the diverse presentations of the remainder of cases are unknown. The recently discovered cranial suture stem cell population affords an opportunity to identify early signaling pathways that contribute to craniosynostosis. We previously demonstrated that enhanced BMP signaling in neural crest cells (caA3 mutants) leads to premature cranial suture fusion resulting in midline craniosynostosis. Since enhanced mTOR signaling in neural crest cells leads to craniofacial bone lesions, we investigated the extent to which mTOR signaling is involved in the pathogenesis of BMP-mediated craniosynostosis by affecting the suture stem cell population. Our results demonstrate a loss of suture stem cells in the caA3 mutant mice by the newborn stage. We have found increased activation of mTOR signaling in caA3 mutant mice during embryonic stages, but not at the newborn stage. Our study demonstrated that inhibition of mTOR signaling via rapamycin in a time specific manner partially rescued the loss of the suture stem cell population. This study provides insight into how enhanced BMP signaling regulates suture stem cells via mTOR activation.


Assuntos
Craniossinostoses/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/fisiologia , Suturas Cranianas/embriologia , Craniossinostoses/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/metabolismo , Crânio/embriologia
6.
J Cell Biochem ; 119(10): 8074-8083, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29380900

RESUMO

Therapeutic strategies targeting both cancer cells and associated cells in the tumor microenvironment offer significant promise in cancer therapy. We previously reported that generation 5 (G5) dendrimers conjugated with cyclic-RGD peptides target cells expressing integrin alpha V beta 3. In this study, we report a novel dendrimer conjugate modified to deliver the mammalian target of rapamycin (mTOR) inhibitor, rapamycin. In vitro analyses demonstrated that this drug conjugate, G5-FI-RGD-rapamycin, binds to prostate cancer (PCa) cells and fibroblasts to inhibit mTOR signaling and VEGF expression. In addition, G5-FI-RGD-rapamycin inhibits mTOR signaling in cancer cells more efficiently under proinflammatory conditions compared to free rapamycin. In vivo studies established that G5-FI-RGD-rapamycin significantly inhibits fibroblast-mediated PCa progression and metastasis. Thus, our results suggest the potential of new rapamycin-conjugated multifunctional nanoparticles for PCa therapy.


Assuntos
Dendrímeros/química , Integrina alfaVbeta3/metabolismo , Metástase Neoplásica/tratamento farmacológico , Peptídeos Cíclicos/química , Neoplasias da Próstata/tratamento farmacológico , Sirolimo/química , Sirolimo/uso terapêutico , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Fibroblastos , Citometria de Fluxo , Humanos , Masculino , Camundongos , Células PC-3
7.
Stem Cells ; 34(3): 588-600, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26661329

RESUMO

Although a specific group of transcription factors such as OCT4, SOX2, and NANOG are known to play essential roles in pluripotent stem cell (PSC) self-renewal, pluripotency, and reprogramming, other factors and the key signaling pathways regulating these important properties are not completely understood. Here, we demonstrate that the PSC marker Developmental Pluripotency Associated 5 (DPPA5) plays an important role in human PSC (hPSC) self-renewal and cell reprogramming in feeder-free conditions. Compared to hPSCs grown on mouse embryonic fibroblasts, cells cultured on feeder-free substrates, such as Matrigel, Laminin-511, Vitronectin, or the synthetic polymer poly[2-(methacryloyloxy) ethyl dimethyl-(3-sulfopropyl) ammonium hydroxide], had significantly higher DPPA5 gene expression and protein levels. Overexpression of DPPA5 in hPSCs increased NANOG protein levels via a post-transcriptional mechanism. Coimmunoprecipitation, protein stability assays, and quantitative RT-PCR, demonstrated that DPPA5 directly interacted, stabilized, and enhanced the function of NANOG in hPSCs. Additionally, DPPA5 increased the reprogramming efficiency of human somatic cells to induced pluripotent stem cells (hiPSCs). Our study provides new insight into the function of DPPA5 and NANOG regulation in hPSCs.


Assuntos
Reprogramação Celular/genética , Proteína Homeobox Nanog/genética , Células-Tronco Pluripotentes , Proteínas/genética , Animais , Diferenciação Celular/genética , Meios de Cultura , Células-Tronco Embrionárias , Fibroblastos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Proteína Homeobox Nanog/biossíntese , Transdução de Sinais
8.
Stem Cells ; 34(7): 1753-64, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26930028

RESUMO

Self-renewal of human embryonic stem cells and human induced pluripotent stem cells (hiPSCs)-known as pluripotent stem cells (PSC)-is influenced by culture conditions, including the substrate on which they are grown. However, details of the molecular mechanisms interconnecting the substrate and self-renewal of these cells remain unclear. We describe a signaling pathway in hPSCs linking self-renewal and expression of pluripotency transcription factors to integrin α6ß1 and inactivation of focal adhesion kinase (FAK). Disruption of this pathway results in hPSC differentiation. In hPSCs, α6ß1 is the dominant integrin and FAK is not phosphorylated at Y397, and thus, it is inactive. During differentiation, integrin α6 levels diminish and Y397 FAK is phosphorylated and activated. During reprogramming of fibroblasts into iPSCs, integrin α6 is upregulated and FAK is inactivated. Knockdown of integrin α6 and activation of ß1 integrin lead to FAK phosphorylation and reduction of Nanog, Oct4, and Sox2, suggesting that integrin α6 functions in inactivation of integrin ß1 and FAK signaling and prevention of hPSC differentiation. The N-terminal domain of FAK, where Y397 is localized, is in the nuclei of hPSCs interacting with Oct4 and Sox2, and this immunolocalization is regulated by Oct4. hPSCs remodel the extracellular microenvironment and deposit laminin α5, the primary ligand of integrin α6ß1. Knockdown of laminin α5 resulted in reduction of integrin α6 expression, phosphorylation of FAK and decreased Oct4. In conclusion, hPSCs promote the expression of integrin α6ß1, and nuclear localization and inactivation of FAK to supports stem cell self-renewal. Stem Cells 2016;34:1753-1764.


Assuntos
Autorrenovação Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrina alfa6beta1/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Diferenciação Celular , Núcleo Celular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/química , Adesões Focais/metabolismo , Células HEK293 , Humanos , Laminina/metabolismo , Fosforilação , Ligação Proteica , Domínios Proteicos , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/metabolismo
9.
Nat Mater ; 13(6): 599-604, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24728461

RESUMO

Our understanding of the intrinsic mechanosensitive properties of human pluripotent stem cells (hPSCs), in particular the effects that the physical microenvironment has on their differentiation, remains elusive. Here, we show that neural induction and caudalization of hPSCs can be accelerated by using a synthetic microengineered substrate system consisting of poly(dimethylsiloxane) micropost arrays (PMAs) with tunable mechanical rigidities. The purity and yield of functional motor neurons derived from hPSCs within 23 days of culture using soft PMAs were improved more than fourfold and tenfold, respectively, compared with coverslips or rigid PMAs. Mechanistic studies revealed a multi-targeted mechanotransductive process involving Smad phosphorylation and nucleocytoplasmic shuttling, regulated by rigidity-dependent Hippo/YAP activities and actomyosin cytoskeleton integrity and contractility. Our findings suggest that substrate rigidity is an important biophysical cue influencing neural induction and subtype specification, and that microengineered substrates can thus serve as a promising platform for large-scale culture of hPSCs.


Assuntos
Diferenciação Celular , Mecanotransdução Celular , Neurônios Motores/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Actomiosina/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Citoesqueleto/metabolismo , Via de Sinalização Hippo , Humanos , Neurônios Motores/citologia , Proteínas Nucleares/genética , Fosforilação , Células-Tronco Pluripotentes/citologia , Proteínas Smad/metabolismo , Fatores de Transcrição/genética
10.
Ann Surg ; 259(5): 993-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-23673767

RESUMO

OBJECTIVE: To demonstrate the pro-osteogenic effect of burn injury on heterotopic bone formation using a novel burn ossicle in vivo model. BACKGROUND: Heterotopic ossification (HO), or the abnormal formation of bone in soft tissue, is a troubling sequela of burn and trauma injuries. The exact mechanism by which burn injury influences bone formation is unknown. The aim of this study was to develop a mouse model to study the effect of burn injury on heterotopic bone formation. We hypothesized that burn injury would enhance early vascularization and subsequent bone formation of subcutaneously implanted mesenchymal stem cells. METHODS: Mouse adipose-derived stem cells were harvested from C57/BL6 mice, transfected with a BMP-2 adenovirus, seeded on collagen scaffolds (ossicles), and implanted subcutaneously in the flank region of 8 adult mice. Burn and sham groups were created with exposure of 30% surface area on the dorsum to 60°C water or 30°C water for 18 seconds, respectively (n = 4/group). Heterotopic bone volume was analyzed in vivo by micro-computed tomography for 3 months. Histological analysis of vasculogenesis was performed with platelet endothelial cell adhesion molecule staining. Osteogenic histological analysis was performed by Safranin O, Picrosirius red, and aniline blue staining. Qualitative analysis of heterotopic bone composition was completed with ex vivo Raman spectroscopy. RESULTS: Subcutaneously implanted ossicles formed heterotopic bone. Ossicles from mice with burn injuries developed significantly more bone than sham control mice, analyzed by micro-computed tomography at 1, 2, and 3 months (P < 0.05), and had enhanced early and late endochondral ossification as demonstrated by Safranin O, Picrosirius red, and aniline blue staining. In addition, burn injury enhanced vascularization of the ossicles (P < 0.05). All ossicles demonstrated chemical composition characteristic of bone as demonstrated by Raman spectroscopy. CONCLUSIONS: Burn injury increases the predilection to osteogenic differentiation of ectopically implanted ossicles. Early differences in vascularity correlated with later bone development. Understanding the role of burn injury on heterotopic bone formation is an important first step toward the development of treatment strategies aimed to prevent unwanted and detrimental heterotopic bone formation.


Assuntos
Osso e Ossos/diagnóstico por imagem , Queimaduras/complicações , Ossificação Heterotópica/etiologia , Animais , Osso e Ossos/patologia , Queimaduras/patologia , Diferenciação Celular , Modelos Animais de Doenças , Seguimentos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/patologia , Microtomografia por Raio-X
11.
Stem Cells ; 31(10): 2183-92, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23766271

RESUMO

Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) is an essential transcription factor for adipocyte differentiation. In mesenchymal stem cells, PPARγ has been assumed to play a negative role in osteoblastic differentiation, by working in an adipogenesis dependent manner, due to the reciprocal relationship between osteoblast and adipocyte differentiation. However, the direct role of PPARγ in osteoblast function is not fully understood, due in part to inadequate model systems. Here, we describe an adenoviral-mediated PPARγ knockout system in which suppression of PPARγ in mesenchymal stem cells enhanced osteoblast differentiation and inhibited adipogenesis in vitro. Consistent with this in vitro observation, lipoatrophic A-ZIP/F1 mice, which do not form adipocytes, displayed a phenotype in which both cortical and trabecular bone was significantly increased compared with wild-type mice. We next developed an inducible osteoblast-targeted PPARγ knockout (Osx Cre/flox- PPARγ) mouse to determine the direct role of PPARγ in bone formation. Data from both in vitro cultures of mesenchymal stem cells and in vivo µCT analysis of bones suggest that suppression of PPARγ activity in osteoblasts significantly increased osteoblast differentiation and trabecular number. Endogenous PPARγ in mesenchymal stem cells and osteoblasts strongly inhibited Akt/mammalian target of rapamycin (mTOR)/p70S6k activity and led to decreased osteoblastic differentiation. Therefore, we conclude that PPARγ modulates osteoblast differentiation and bone formation through both direct and indirect mechanisms. The direct mode, as shown here, involves PPARγ regulation of the mTOR pathway, while the indirect pathway is dependent on the regulation of adipogenesis.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Osteoblastos/metabolismo , Osteogênese , PPAR gama/genética , Serina-Treonina Quinases TOR/metabolismo , Adipogenia , Animais , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/fisiologia , Calcificação Fisiológica , Células Cultivadas , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo , Radiografia , Transdução de Sinais
12.
Front Oncol ; 14: 1375498, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38532930

RESUMO

mEAK-7 (mammalian EAK-7 or MTOR-associated protein, eak-7 homolog), is an evolutionarily conserved lysosomal membrane protein that is highly expressed in several cancer cells. Multiple recent studies have identified mEAK-7 as a positive activator of mTOR (mammalian/mechanistic target of rapamycin) signaling via an alternative mTOR complex, implying that mEAK-7 plays an important role in the promotion of cancer proliferation and migration. In addition, structural analyses investigating interactions between mEAK-7 and V-ATPase, a protein complex responsible for regulating pH homeostasis in cellular compartments, have suggested that mEAK-7 may contribute to V-ATPase-mediated mTORC1 activation. The C-terminal α-helix of mEAK-7 binds to the D and B subunits of the V-ATPase, creating a pincer-like grip around its B subunit. This binding undergoes partial disruption during ATP hydrolysis, potentially enabling other proteins such as mTOR to bind to the α-helix of mEAK-7. mEAK-7 also promotes chemoresistance and radiation resistance by sustaining DNA damage-mediated mTOR signaling through interactions with DNA-PKcs (DNA-dependent protein kinase catalytic subunit). Taken together, these findings indicate that mEAK-7 may be a promising therapeutic target against tumors. However, the precise molecular mechanisms and signal transduction pathways of mEAK-7 in cancer remain largely unknown, motivating the need for further investigation. Here, we summarize the current known roles of mEAK-7 in normal physiology and cancer development by reviewing the latest studies and discuss potential future developments of mEAK-7 in targeted cancer therapy.

13.
Exp Cell Res ; 318(19): 2427-37, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22841688

RESUMO

In the bone marrow cavity, hematopoietic stem cells (HSC) have been shown to reside in the endosteal and subendosteal perivascular niches, which play specific roles on HSC maintenance. Although cells with long-term ability to reconstitute full hematopoietic system can be isolated from both niches, several data support a heterogenous distribution regarding the cycling behavior of HSC. Whether this distinct behavior depends upon the role played by the stromal populations which distinctly create these two niches is a question that remains open. In the present report, we used our previously described in vivo assay to demonstrate that endosteal and subendosteal stromal populations are very distinct regarding skeletal lineage differentiation potential. This was further supported by a microarray-based analysis, which also demonstrated that these two stromal populations play distinct, albeit complementary, roles in HSC niche. Both stromal populations were preferentially isolated from the trabecular region and behave distinctly in vitro, as previously reported. Even though these two niches are organized in a very close range, in vivo assays and molecular analyses allowed us to identify endosteal stroma (F-OST) cells as fully committed osteoblasts and subendosteal stroma (F-RET) cells as uncommitted mesenchymal cells mainly represented by perivascular reticular cells expressing high levels of chemokine ligand, CXCL12. Interestingly, a number of cytokines and growth factors including interleukin-6 (IL-6), IL-7, IL-15, Hepatocyte growth factor (HGF) and stem cell factor (SCF) matrix metalloproteases (MMPs) were also found to be differentially expressed by F-OST and F-RET cells. Further microarray analyses indicated important mechanisms used by the two stromal compartments in order to create and coordinate the "quiescent" and "proliferative" niches in which hematopoietic stem cells and progenitors reside.


Assuntos
Medula Óssea/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Células Estromais/fisiologia , Animais , Medula Óssea/metabolismo , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Perfilação da Expressão Gênica/métodos , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/fisiologia , Células Estromais/citologia , Células Estromais/metabolismo
14.
Heliyon ; 9(10): e20655, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37867861

RESUMO

Growing evidence suggests an association between Mycoplasma infections and the development and progression of prostate cancer (PCa). In this study, we report that chronic and persistent M. hyorhinis infection induced robust TNF-α secretion from PCa cells. TNF-α secreted from M. hyorhinis-infected PCa cells subsequently led to activation of the NF-κB pathway. Chronic M. hyorhinis infection induced gene expression of pro-inflammatory cytokines and chemokines in a NF-κB-dependent manner and promoted cell proliferation, migration, and invasion in PCa cells. The elimination of M. hyorhinis in PCa cells significantly blocked TNF-α secretion, gene expression of cytokines and chemokines, migration, and invasion in PCa cells, suggesting M. hyorhinis-induced TNF-α plays an important role to promote malignant transformation of PCa. Furthermore, second mitochondria-derived activator of caspases (SMAC) mimetics potentiated caspase activation and cell death in M. hyorhinis-infected PCa by antagonizing inhibitor of apoptosis proteins (IAPs) activity. Tissue microarray analysis indicated that TNF-α is co-expressed in M. hyorhinis-infected human patient tissues. Findings from this study advance our understanding of the mycoplasma-oncogenesis process and suggest the potential for new approaches for preventions, diagnosis, and therapeutic approaches against prostate cancers.

15.
Ann Clin Transl Neurol ; 10(7): 1239-1253, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37283238

RESUMO

OBJECTIVE: Brain organoids are miniaturized in vitro brain models generated from pluripotent stem cells, which resemble full-sized brain more closely than conventional two-dimensional cell cultures. Although brain organoids mimic the human brain's cell-to-cell network interactions, they generally fail to faithfully recapitulate cell-to-matrix interactions. Here, an engineered framework, called an engineered extracellular matrix (EECM), was developed to provide support and cell-to-matrix interactions to developing brain organoids. METHODS: We generated brain organoids using EECMs comprised of human fibrillar fibronectin supported by a highly porous polymer scaffold. The resultant brain organoids were characterized by immunofluorescence microscopy, transcriptomics, and proteomics of the cerebrospinal fluid (CSF) compartment. RESULTS: The interstitial matrix-mimicking EECM enhanced neurogenesis, glial maturation, and neuronal diversity from human embryonic stem cells versus conventional protein matrix (Matrigel). Additionally, EECMs supported long-term culture, which promoted large-volume organoids containing over 250 µL of CSF. Proteomics analysis of the CSF found it superseded previous brain organoids in protein diversity, as indicated by 280 proteins spanning 500 gene ontology pathways shared with adult CSF. INTERPRETATION: Engineered EECM matrices represent a major advancement in neural engineering as they have the potential to significantly enhance the structural, cellular, and functional diversity that can be achieved in advanced brain models.


Assuntos
Organoides , Células-Tronco Pluripotentes , Adulto , Humanos , Organoides/metabolismo , Matriz Extracelular , Encéfalo , Neurogênese
16.
J Cell Biochem ; 113(1): 220-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898543

RESUMO

The role of erythropoietin (Epo) and Epo/Epo receptor (EpoR) signaling pathways for production of red blood cells are well established. However, little is known about Epo/EpoR signaling in non-hematopoietic cells. Recently, we demonstrated that Epo activates JAK/STAT signaling in hematopoietic stem cells (HSCs), leading to the production of bone morphogenetic protein 2 (BMP2) and bone formation and that Epo also directly activates mesenchymal cells to form osteoblasts in vitro. In this study, we investigated the effects of mTOR signaling on Epo-mediated osteoblastogenesis and osteoclastogenesis. We found that mTOR inhibition by rapamycin blocks Epo-dependent and -independent osteoblastic phenotypes in human bone marrow stromal cells (hBMSCs) and ST2 cells, respectively. Furthermore, we found that rapamycin inhibits Epo-dependent and -independent osteoclastogenesis in mouse bone marrow mononuclear cells and Raw264.7 cells. Finally, we demonstrated that Epo increases NFATc1 expression and decreases cathepsin K expression in an mTOR-independent manner, resulting in an increase of osteoclast numbers and a decrease in resorption activity. Taken together, these results strongly indicate that mTOR signaling plays an important role in Epo-mediated bone homeostasis.


Assuntos
Eritropoetina/metabolismo , Osteogênese/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Catepsina K/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Fatores de Transcrição NFATC/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Células Estromais/metabolismo
17.
Am J Pathol ; 178(6): 2461-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21641374

RESUMO

Fibrotic obliteration of the small airways leading to progressive airflow obstruction, termed bronchiolitis obliterans syndrome (BOS), is the major cause of poor outcomes after lung transplantation. We recently demonstrated that a donor-derived population of multipotent mesenchymal stem cells (MSCs) can be isolated from the bronchoalveolar lavage (BAL) fluid of human lung transplant recipients. Herein, we study the organ specificity of these cells and investigate the role of local mesenchymal progenitors in fibrogenesis after lung transplantation. We demonstrate that human lung allograft-derived MSCs uniquely express embryonic lung mesenchyme-associated transcription factors with a 35,000-fold higher expression of forkhead/winged helix transcription factor forkhead box (FOXF1) noted in lung compared with bone marrow MSCs. Fibrotic differentiation of MSCs isolated from normal lung allografts was noted in the presence of profibrotic mediators associated with BOS, including transforming growth factor-ß and IL-13. MSCs isolated from patients with BOS demonstrated increased expression of α-SMA and collagen I when compared with non-BOS controls, consistent with a stable in vivo fibrotic phenotype. FOXF1 mRNA expression in the BAL cell pellet correlated with the number of MSCs in the BAL fluid, and myofibroblasts present in the fibrotic lesions expressed FOXF1 by in situ hybridization. These data suggest a key role for local tissue-specific, organ-resident, mesenchymal precursors in the fibrogenic processes in human adult lungs.


Assuntos
Transplante de Pulmão , Pulmão/patologia , Células-Tronco Mesenquimais/patologia , Actinas/metabolismo , Biomarcadores/metabolismo , Biópsia , Células da Medula Óssea/patologia , Bronquiolite Obliterante/patologia , Líquido da Lavagem Broncoalveolar , Contagem de Células , Diferenciação Celular , Separação Celular , Colágeno/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Humanos , Pulmão/embriologia , Pulmão/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miofibroblastos/patologia , Especificidade de Órgãos , Fenótipo , Receptores de Interleucina-13/metabolismo , Transplante Homólogo
18.
Blood ; 115(13): 2592-600, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20110425

RESUMO

Because the microenvironment that supports hematopoietic stem cell (HSC) proliferation and differentiation is not fully understood, we adapted a heterotopic bone formation model as a new approach for studying the HSC microenvironment in vivo. Endogenous HSCs homed to tissue-engineered ossicles and individually sorted HSCs from ossicles were able to reconstitute lethally irradiated mice. To further explore this model as a system to study the stem cell niche, ossicles were established with or without anabolic parathyroid hormone (PTH) treatment during the 4-week course of bone development. Histology and micro-computed tomography showed higher bone area-to-total area ratios, thicker cortical bone and trabecular bone, significantly higher bone mineral density and bone volume fraction in PTH-treated groups than in controls. By an in vivo competitive long-term reconstitution assay, HSC frequency in the ossicle marrow was 3 times greater in PTH groups than in controls. When whole bone marrow cells were directly injected into the ossicles after lethal irradiation, the PTH-treated groups showed an enhanced reconstitution rate compared with controls. These findings suggest the residence of HSCs in heterotopic bone marrow and support the future use of this ossicle model in elucidating the composition and regulation of the HSC niche.


Assuntos
Células-Tronco Hematopoéticas/citologia , Modelos Animais , Ossificação Heterotópica , Adipócitos/fisiologia , Animais , Biomarcadores , Medula Óssea/ultraestrutura , Contagem de Células , Linhagem da Célula , Células Cultivadas/transplante , Transplante de Células-Tronco Hematopoéticas/métodos , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/farmacologia , Quimera por Radiação , Células Estromais/fisiologia , Tela Subcutânea , Alicerces Teciduais
19.
Proc Natl Acad Sci U S A ; 106(32): 13219-24, 2009 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-19628695

RESUMO

Gap junction intercellular communication (GJIC) is ubiquitous in the majority of vertebrate cells and is required for the proper development of most tissues. The loss of gap junction-mediated cell-to-cell communication leads to compromised development in many tissues and organs. Because cells constantly interact through gap junctions to coordinate tissue functions and homeostasis, we hypothesized that increasing cell-to-cell communication, via genetically engineering cells to overexpress gap junction proteins, could enhance cell differentiation in the interior regions of 3D tissue equivalents, thereby increasing the ability to regenerate larger and more uniform volumes of tissue. To test this hypothesis, we used bone as a model tissue because of the difficulty in achieving spatially uniform bone regeneration in 3D. In bone marrow stromal cells (BMSC), GJIC and osteogenic differentiation were compromised in 3D cultures relative to 2D monolayers and in the core of 3D cultures relative to the surface. Overexpression of connexin 43 (Cx43) via transduction of BMSCs with a lentivirus overcame this problem, enhancing both the magnitude and spatial distribution of GJIC and osteogenic differentiation markers throughout 3D constructs. Transplantation of cells overexpressing Cx43 resulted in an increased volume fraction and spatial uniformity of bone in vivo, relative to nontransduced BMSCs. Increased GJIC also enhanced the effect of a potent osteoinductive agent (BMP-7), suggesting a synergism between the soluble factor and GJIC. These findings present a platform to improve cell-to-cell communication in 3D and to achieve uniformly distributed tissue regeneration in 3D.


Assuntos
Regeneração Óssea , Osso e Ossos/patologia , Conexina 43/metabolismo , Transdução de Sinais , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Osso e Ossos/diagnóstico por imagem , Comunicação Celular , Diferenciação Celular , Conexina 43/genética , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Medicina Regenerativa , Células Estromais/citologia , Células Estromais/metabolismo , Engenharia Tecidual , Transdução Genética , Microtomografia por Raio-X
20.
J Craniofac Surg ; 23(1): 333-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22337437

RESUMO

Gene therapy in the craniofacial region provides a unique tool for delivery of DNA to coordinate protein production in both time and space. The drive to bring this technology to the clinic is derived from the fact that more than 85% of the global population may at one time require repair or replacement of a craniofacial structure. This need ranges from mild tooth decay and tooth loss to temporomandibular joint disorders and large-scale reconstructive surgery. Our ability to insert foreign DNA into a host cell has been developing since the early uses of gene therapy to alter bacterial properties for waste cleanup in the 1980s followed by successful human clinical trials in the 1990s to treat severe combined immunodeficiency. In the past 20 years, the emerging field of craniofacial tissue engineering has adopted these techniques to enhance regeneration of mineralized tissues, salivary gland, and periodontium and to reduce tumor burden of head and neck squamous cell carcinoma. Studies are currently pursuing research on both biomaterial-mediated gene delivery and more clinically efficacious, although potentially more hazardous, viral methods. Although hundreds of gene therapy clinical trials have taken place in the past 20 years, we must still work to ensure an ideal safety profile for each gene and delivery method combination. With adequate genotoxicity testing, we can expect gene therapy to augment protein delivery strategies and potentially allow for tissue-specific targeting, delivery of multiple signals, and increased spatial and temporal control with the goal of natural tissue replacement in the craniofacial complex.


Assuntos
Regeneração Óssea/genética , Ossos Faciais/anatomia & histologia , Terapia Genética/métodos , Crânio/anatomia & histologia , Engenharia Tecidual/métodos , Materiais Biocompatíveis/química , Técnicas de Transferência de Genes , Engenharia Genética/métodos , Vetores Genéticos , Humanos , Engenharia de Proteínas/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA