Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Front Endocrinol (Lausanne) ; 12: 635405, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025576

RESUMO

Background: Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the ß-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. Methods: Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. Results: CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. Conclusions: Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Dipeptidil Peptidase 4/biossíntese , Integrina alfa1/biossíntese , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Animais , Peptídeo C/biossíntese , Diferenciação Celular , Separação Celular , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microesferas
2.
Stem Cell Res Ther ; 9(1): 152, 2018 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-29871694

RESUMO

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a motor neuron (MN) disease characterized by the loss of MNs in the central nervous system. As MNs die, patients progressively lose their ability to control voluntary movements, become paralyzed and eventually die from respiratory/deglutition failure. Despite the selective MN death in ALS, there is growing evidence that malfunctional astrocytes play a crucial role in disease progression. Thus, transplantation of healthy astrocytes may compensate for the diseased astrocytes. METHODS: We developed a good manufacturing practice-grade protocol for generation of astrocytes from human embryonic stem cells (hESCs). The first stage of our protocol is derivation of astrocyte progenitor cells (APCs) from hESCs. These APCs can be expanded in large quantities and stored frozen as cell banks. Further differentiation of the APCs yields an enriched population of astrocytes with more than 90% GFAP expression (hES-AS). hES-AS were injected intrathecally into hSOD1G93A transgenic mice and rats to evaluate their therapeutic potential. The safety and biodistribution of hES-AS were evaluated in a 9-month study conducted in immunodeficient NSG mice under good laboratory practice conditions. RESULTS: In vitro, hES-AS possess the activities of functional healthy astrocytes, including glutamate uptake, promotion of axon outgrowth and protection of MNs from oxidative stress. A secretome analysis shows that these hES-AS also secrete several inhibitors of metalloproteases as well as a variety of neuroprotective factors (e.g. TIMP-1, TIMP-2, OPN, MIF and Midkine). Intrathecal injections of the hES-AS into transgenic hSOD1G93A mice and rats significantly delayed disease onset and improved motor performance compared to sham-injected animals. A safety study in immunodeficient mice showed that intrathecal transplantation of hES-AS is safe. Transplanted hES-AS attached to the meninges along the neuroaxis and survived for the entire duration of the study without formation of tumors or teratomas. Cell-injected mice gained similar body weight to the sham-injected group and did not exhibit clinical signs that could be related to the treatment. No differences from the vehicle control were observed in hematological parameters or blood chemistry. CONCLUSION: Our findings demonstrate the safety and potential therapeutic benefits of intrathecal injection of hES-AS for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Astrócitos/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Injeções Espinhais/métodos , Superóxido Dismutase-1/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Ratos , Superóxido Dismutase-1/metabolismo
3.
J Tissue Eng Regen Med ; 9(9): 977-87, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23365073

RESUMO

Vascularization of injured tissues or artificial grafts is a major challenge in tissue engineering, stimulating a continued search for alternative sources for vasculogenic cells and the development of therapeutic strategies. Human pluripotent stem cells (hPSCs), either embryonic or induced, offer a plentiful platform for the derivation of large numbers of vasculogenic cells, as required for clinical transplantations. Various protocols for generation of vasculogenic smooth muscle cells (SMCs) from hPSCs have been described with considerably different SMC derivatives. In addition, we recently identified hPSC-derived pericytes, which are similar to their physiological counterparts, exhibiting unique features of blood vessel-residing perivascular cells, as well as multipotent mesenchymal precursors with therapeutic angiogenic potential. In this review we refer to methodologies for the development of a variety of perivascular cells from hPSCs with respect to developmental induction, differentiation capabilities, potency and their dual function as mesenchymal precursors. The therapeutic effect of hPSC-derived perivascular cells in experimental models of tissue engineering and regenerative medicine are described and compared to those of their native physiological counterparts.


Assuntos
Vasos Sanguíneos/citologia , Transplante de Células , Células-Tronco Pluripotentes/citologia , Animais , Regeneração Óssea , Condrogênese , Humanos , Distrofia Muscular Animal/terapia , Infarto do Miocárdio/terapia , Neovascularização Fisiológica , Pele/citologia
4.
Can J Cardiol ; 30(11): 1279-87, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25442431

RESUMO

Human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) have the capacity to differentiate into any specialized cell type, including cardiomyocytes. Therefore, hESC-derived and hiPSC-derived cardiomyocytes (hESC-CMs and hiPSC-CMs, respectively) offer great potential for cardiac regenerative medicine. Unlike some organs, the heart has a limited ability to regenerate, and dysfunction resulting from significant cardiomyocyte loss under pathophysiological conditions, such as myocardial infarction (MI), can lead to heart failure. Unfortunately, for patients with end-stage heart failure, heart transplantation remains the main alternative, and it is insufficient, mainly because of the limited availability of donor organs. Although left ventricular assist devices are progressively entering clinical practice as a bridge to transplantation and even as an optional therapy, cell replacement therapy presents a plausible alternative to donor organ transplantation. During the past decade, multiple candidate cells were proposed for cardiac regeneration, and their mechanisms of action in the myocardium have been explored. The purpose of this article is to critically review the comprehensive research involving the use of hESCs and hiPSCs in MI models and to discuss current controversies, unresolved issues, challenges, and future directions.


Assuntos
Células-Tronco Embrionárias/transplante , Cardiopatias/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Miócitos Cardíacos/patologia , Transplante de Células-Tronco/métodos , Diferenciação Celular , Cardiopatias/patologia , Humanos
5.
Tissue Eng Part A ; 20(19-20): 2756-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25185111

RESUMO

Extracellular matrix (ECM) has been utilized as a biological scaffold for tissue engineering applications in a variety of body systems, due to its bioactivity and biocompatibility. In the current study we developed a modified protocol for the efficient and reproducible derivation of mesenchymal progenitor cells (MPCs) from human embryonic stem cells as well as human induced pluripotent stem cells (hiPSCs) originating from hair follicle keratinocytes (HFKTs). ECM was produced from these MPCs and characterized in comparison to adipose mesenchymal stem cell ECM, demonstrating robust ECM generation by the excised HFKT-iPSC-MPCs. Exploiting the advantages of electrospinning we generated two types of electrospun biodegradable nanofiber layers (NFLs), fabricated from polycaprolactone (PCL) and poly(lactic-co-glycolic acid) (PLGA), which provide mechanical support for cell seeding and ECM generation. Elucidating the optimized decellularization treatment we were able to generate an available "off-the-shelf" implantable product (NFL-ECM). Using rat subcutaneous transplantation model we demonstrate that this stem-cell-derived construct is biocompatible and biodegradable and holds great potential for tissue regeneration applications.


Assuntos
Matriz Extracelular/química , Células-Tronco Pluripotentes Induzidas/metabolismo , Ácido Láctico/química , Nanofibras/química , Poliésteres/química , Ácido Poliglicólico/química , Medicina Regenerativa , Animais , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Folículo Piloso/citologia , Folículo Piloso/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Queratinócitos/citologia , Queratinócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos
6.
Stem Cells ; 32(12): 3137-49, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25186014

RESUMO

Human embryonic stem cells (hESCs) represent a potential source of transplantable cells for regenerative medicine, but development of teratoma even in syngenic recipients represents a critical obstacle to safe stem cell-based therapies. We hypothesized that hESCs escape the immune surveillance by regulating the environmental immune system. Using cocultures of hESCs with allogenic peripheral blood mononuclear cells, we demonstrated that hESCs prevent proliferation and activation of human CD4+ T lymphocytes, an effect dependent upon monocytes. Altered expression of key signaling molecules responsible for the crosstalk of monocytes with T cells was detected in the presence of hESCs. Analyzing the mechanism of action, we demonstrated that hESCs were able to downregulate intracellular glutathione levels in both monocytes and CD4+ cells by suppressing glutamate cysteine ligase expression and to alter MHCII and CD80 expression in monocytes. These effects were achieved at least partially via TGF-beta signaling, and both monocyte phenotype and GCLC expression were affected by Caspase-3 proteolytic activity. Altogether, our results demonstrate a novel immune-suppressive mechanism used by hESCs.


Assuntos
Diferenciação Celular/fisiologia , Células Dendríticas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Ativação Linfocitária/imunologia , Transdução de Sinais , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Antígenos CD/imunologia , Diferenciação Celular/imunologia , Técnicas de Cocultura/métodos , Humanos , Monócitos/citologia , Transdução de Sinais/fisiologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta/imunologia
7.
Stem Cells Dev ; 23(5): 443-56, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24192045

RESUMO

Adenosine-to-inosine (A-to-I) RNA editing is a post-transcriptional, site-specific modification process that is catalyzed by Adenosine Deaminase Acting on RNA (ADAR) gene family members. Since ADARs act on double-stranded RNA, most A-to-I editing occurs within repetitive elements, particularly Alu elements, as the result of the inherent property of these sequences to fold and form double strands. ADAR1-mediated A-to-I RNA editing was recently implicated in the regulation of human embryonic stem cells (hESCs). Spontaneous and neuronal differentiation of hESC was shown to result in a decrease in A-to-I editing levels. Knockdown of ADAR1 in hESCs results in an elevation of the expression of differentiation-related genes. In addition, we found that hESCs over-expressing ADAR1 could not be generated. The current study shows that the editing levels of induced pluripotent stem cells (iPSCs) change throughout reprogramming, from a source cell level to a level similar to that of hESCs. Up- or down-regulation of the ADAR1 level in human foreskin fibroblast (HFF) cells before induction of reprogramming results in varied reprogramming efficiencies. Furthermore, HFF-iPSC early clones derived from source cells in which the ADAR1 level was down-regulated lose their iPSC properties shortly after iPSC colony formation and instead exhibit characteristics of cancer cells. Taken together, our results imply a role for ADAR1 in the regulation of pluripotency induction as well as in the maintenance of early iPSC properties.


Assuntos
Adenosina Desaminase/biossíntese , Diferenciação Celular/genética , Células-Tronco Embrionárias , Células-Tronco Pluripotentes Induzidas , Adenosina Desaminase/genética , Fibroblastos , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas de Ligação a RNA
8.
Am J Obstet Gynecol ; 208(3): 213.e1-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23433325

RESUMO

OBJECTIVE: Maternal infection or inflammation may induce fetal inflammatory responses associated with fetal injury and cerebral palsy. We sought to assess the inflammation-associated neuroprotective potential of prophylactic N-acetyl-cysteine (NAC). We examined the effect of NAC on prevention of maternal lipopolysaccharide (LPS)-induced neonatal brain injury using magnetic resonance imaging. STUDY DESIGN: Pregnant Sprague Dawley dams (n = 5-8) at embryonic day 18 received intraperitoneal injection of LPS or saline at time 0. Animals were randomized to receive 2 intravenous injections of NAC or saline (time -30 and 120 minutes). Pups were delivered spontaneously and allowed to mature until postnatal day 25. Female offspring were examined by magnetic resonance brain imaging and analyzed using voxel-based analysis after spatial normalization. T2 relaxation time was used to assess white matter injury and diffusion tensor imaging for apparent diffusion coefficient (ADC) to assess white and gray matter injury. RESULTS: Offspring of LPS-treated dams exhibited significantly increased T2 levels and increased ADC levels in white and gray matter (eg, hypothalamus, motor cortex, corpus callosum, thalamus, hippocampus), consistent with diffuse cerebral injury. In contrast, offspring of NAC-treated LPS dams demonstrated similar T2 and ADC levels as control in both white and gray matter. CONCLUSION: Maternal NAC treatment significantly reduced evidence of neonatal brain injury associated with maternal LPS. These studies suggest that maternal NAC therapy may be effective in human deliveries associated with maternal/fetal inflammation.


Assuntos
Acetilcisteína/farmacologia , Animais Recém-Nascidos , Lesões Encefálicas/prevenção & controle , Encéfalo/efeitos dos fármacos , Transmissão Vertical de Doenças Infecciosas , Inflamação/prevenção & controle , Prenhez , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Animais , Encéfalo/patologia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/patologia , Feminino , Inflamação/induzido quimicamente , Inflamação/patologia , Lipopolissacarídeos , Imageamento por Ressonância Magnética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Sprague-Dawley
9.
PLoS One ; 7(7): e41576, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22859999

RESUMO

Post-transcriptional events play an important role in human development. The question arises as to whether Adenosine to Inosine RNA editing, catalyzed by the ADAR (Adenosine Deaminase acting on RNA) enzymes, differs in human embryogenesis and in adulthood. We tested the editing of various target genes in coding (FLNA, BLCAP, CYFIP2) and non-coding sequences at their Alu elements (BRCA1, CARD11, RBBP9, MDM4, FNACC), as well as the transcriptional levels of the ADAR1 enzymes. This analysis was performed on five fetal and adult human tissues: brain, heart, liver, kidney, and spleen, as well as on human embryonic stem cells (hESCs), which represent the blastocyst stage in early human development. Our results show substantially greater editing activity for most adult tissue samples relative to fetal ones, in six of the eight genes tested. To test the effect of reduced A-to-I RNA editing activity in early human development we used human embryonic stem cells (hESCs) as a model and tried to generate hESC clones that overexpress the ADAR1-p110 isoform. We were unable to achieve overexpression of ADAR1-p110 by either transfection or lentiviral infection, though we easily generated hESC clones that expressed the GFP transgene and overexpressed ADAR1-p110 in 293T cells and in primary human foreskin fibroblast (HFF) cells. Moreover, in contrast to the expected overexpression of ADAR1-p110 protein following its introduction into hESCs, the expression levels of this protein decreased dramatically 24-48 hr post infection. Similar results were obtained when we tried to overexpress ADAR1-p110 in pluripotent embryonal carcinoma cells. This suggests that ADAR1 protein is substantially regulated in undifferentiated pluripotent hESCs. Overall, our data suggest that A-to-I RNA editing plays a critical role during early human development.


Assuntos
Desenvolvimento Embrionário , Edição de RNA , RNA Mensageiro/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina/genética , Adenosina/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Elementos Alu , Proteína BRCA1/genética , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteínas Contráteis/genética , Células-Tronco de Carcinoma Embrionário , Células-Tronco Embrionárias , Proteína do Grupo de Complementação C da Anemia de Fanconi/genética , Filaminas , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Guanilato Ciclase/genética , Humanos , Inosina/genética , Inosina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas dos Microfilamentos/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA
10.
J Cell Sci ; 125(Pt 19): 4640-50, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22767507

RESUMO

The mechanisms underlying the immunomodulatory effects of mesenchymal stem cells (MSCs) have been investigated under extreme conditions of strong T cell activation, which induces the rapid death of activated lymphocytes. The objective of this study was to investigate these mechanisms in the absence of additional polyclonal activation. In co-cultures of peripheral mononuclear blood cells with human MSCs (hereafter referred to as hMSCs), we observed a striking decrease in the level of CD8 expression on CD8+ cells, together with decreased expression of CD28 and CD44, and impaired production of IFN-gamma and Granzyme B. This effect was specific to hMSCs, because it was not observed with several other cell lines. Downregulation of CD8 expression required CD14+ monocytes to be in direct contact with the CD8+ cells, whereas the effects of hMSCs on the CD14+ cells were essentially mediated by soluble factors. The CD14+ monocytes exhibited a tolerogenic pattern when co-cultured with hMSCs, with a clear decrease in CD80 and CD86 co-stimulatory molecules, and an increase in the inhibitory receptors ILT-3 and ILT-4. CD8+ cells that were preconditioned by MSCs had similar effects on monocytes and were able to inhibit lymphocyte proliferation. Injection of hMSCs in humanized NSG mice showed similar trends, in particular decreased levels of CD44 and CD28 in human immune cells. Our study demonstrates a new immunomodulation mechanism of action of hMSCs through the modulation of CD8+ cells towards a non-cytotoxic and/or suppressive phenotype. This mechanism of action has to be taken into account in clinical trials, where it should be beneficial in grafts and autoimmune diseases, but potentially detrimental in malignant diseases.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica/imunologia , Células-Tronco Mesenquimais/citologia , Monócitos/citologia , Monócitos/imunologia , Adulto , Animais , Biomarcadores/metabolismo , Antígenos CD8/metabolismo , Moléculas de Adesão Celular/metabolismo , Regulação para Baixo , Feminino , Humanos , Imunização , Receptores de Lipopolissacarídeos/metabolismo , Ativação Linfocitária/genética , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Transcrição Gênica
11.
Arch Gynecol Obstet ; 286(4): 983-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22648447

RESUMO

PURPOSE: To evaluate the learning curve of senior urogynecologic surgeons performing laparoscopic sacral colpopexy (LSCP) and to assess outcomes and complications of LSCP. METHODS: We conducted a retrospective study of 47 consecutive women who underwent LSCP for pelvic organ prolapse repair between March 2009 and December 2010 at one tertiary medical center. Preoperative, intraoperative, postoperative, and demographic data were retrieved from patients' electronic charts. Pelvic organ support was assessed objectively using the Pelvic Organ Prolapse Quantification scale (POP-Q). Anatomic failure was determined as POP-Q stage ≥ II. RESULTS: The mean age of patients was 58 years (range 35-73 years). Seven (15 %) who opted to retain their uterus underwent sacrohysteropexies. The median POP-Q was III (II-IV). Of the 47 operations, 96 % (45) were completed by laparoscopy. The duration of surgery decreased as experience of the surgical team increased, from a mean of 196 ± 62 min for the first 15 cases to 162 ± 30 min for the subsequent 30. Four patients (9 %) presented with recurrence of prolapse; three (7 %) had de novo stress urinary incontinence; two sustained a cystotomy during adhesiolysis, and one had a port-site hernia. CONCLUSIONS: LSCP is a safe and effective treatment for pelvic organ prolapse, with very few complications. Following the first 15 cases of one surgical team, operative time decreased considerably.


Assuntos
Procedimentos Cirúrgicos em Ginecologia , Prolapso de Órgão Pélvico/cirurgia , Adulto , Idoso , Feminino , Procedimentos Cirúrgicos em Ginecologia/efeitos adversos , Humanos , Laparoscopia , Curva de Aprendizado , Pessoa de Meia-Idade , Estudos Retrospectivos , Resultado do Tratamento
12.
Tissue Eng Part A ; 18(21-22): 2290-302, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22731654

RESUMO

Human mesenchymal stem cells (hMSCs) can be derived from various adult and fetal tissues. However, the quality of tissues for the isolation of adult and fetal hMSCs is donor dependent with a nonreproducible yield. In addition, tissue engineering and cell therapy require large-scale production of a pure population of lineage-restricted stem cells that can be easily induced to differentiate into a specific cell type. Therefore, human embryonic stem cells (hESCs) can provide an alternative, plentiful source for generation of reproducible hMSCs. We have developed efficient differentiation protocols for derivation of hMSCs from hESCs, including coculture with murine OP9 stromal cells and feeder layer-free system. Our protocols have resulted in the generation of up to 49% of hMSCs, which expressed CD105, CD90, CD29, and CD44. The hMSCs exhibited high adipogenic, chondrocytic, and osteogenic differentiation in vitro. The latter correlated with osteocalcin secretion and vascular endothelial growth factor (VEGF) production by the differentiating hMSCs. hMSC-derived osteoblasts further differentiated and formed ectopic bone in vivo, and induced the formation of blood vessels in Matrigel implants. Our protocol enables generation of a purified population of hESC-derived MSCs, with the potential of differentiating into several mesodermal lineages, and particularly into vasculogenesis-inducing osteoblasts, which can contribute to the development of bone repair protocols.


Assuntos
Osso e Ossos/irrigação sanguínea , Coristoma/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Separação Celular , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Técnicas de Cocultura , Colágeno/farmacologia , Combinação de Medicamentos , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Humanos , Cinética , Laminina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Neovascularização Fisiológica/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , Proteoglicanas/farmacologia , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Stem Cell Rev Rep ; 8(3): 792-802, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22592939

RESUMO

New sources of beta cells are needed in order to develop cell therapies for patients with diabetes. An alternative to forced expansion of post-mitotic beta cells is the induction of differentiation of stem-cell derived progenitor cells that have a natural self-expansion capacity into insulin-producing cells. In order to learn more about these progenitor cells at different stages along the differentiation process in which they become progressively more committed to the final beta cell fate, we took the approach of identifying, isolating and characterizing stage specific progenitor cells. We generated human embryonic stem cell (HESC) clones harboring BAC GFP reporter constructs of SOX17, a definitive endoderm marker, and PDX1, a pancreatic marker, and identified subpopulations of GFP expressing cells. Using this approach, we isolated a highly enriched population of pancreatic progenitor cells from hESCs and examined their gene expression with an emphasis on the expression of stage-specific cell surface markers. We were able to identify novel molecules that are involved in the pancreatic differentiation process, as well as stage-specific cell markers that may serve to define (alone or in combination with other markers) a specific pancreatic progenitor cell. These findings may help in optimizing conditions for ultimately generating and isolating beta cells for transplantation therapy.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Proteínas de Membrana/metabolismo , Pâncreas/citologia , Proteínas de Arabidopsis/biossíntese , Proteínas de Arabidopsis/genética , Biomarcadores/metabolismo , Carbono-Nitrogênio Liases , Separação Celular , Células Cultivadas , Cromossomos Artificiais Bacterianos/genética , Diabetes Mellitus Tipo 1/terapia , Células-Tronco Embrionárias/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Células Secretoras de Insulina/transplante , Transferases de Grupos Nitrogenados/biossíntese , Transferases de Grupos Nitrogenados/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Fatores de Transcrição SOXF/biossíntese , Fatores de Transcrição SOXF/genética , Transcriptoma , Transgenes
14.
Stem Cells ; 30(6): 1097-108, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22415928

RESUMO

While experimentally induced arrest of human embryonic stem cells (hESCs) in G1 has been shown to stimulate differentiation, it remains unclear whether the unperturbed G1 phase in hESCs is causally related to differentiation. Here, we use centrifugal elutriation to isolate and investigate differentiation propensities of hESCs in different phases of their cell cycle. We found that isolated G1 cells exhibit higher differentiation propensity compared with S and G2 cells, and they differentiate at low cell densities even under self-renewing conditions. This differentiation of G1 cells was partially prevented in dense cultures of these cells and completely abrogated in coculture with S and G2 cells. However, coculturing without cell-to-cell contact did not rescue the differentiation of G1 cells. Finally, we show that the subset of G1 hESCs with reduced phosphorylation of retinoblastoma has the highest propensity to differentiate and that the differentiation is preceded by cell cycle arrest. These results provide direct evidence for increased propensity of hESCs to differentiate in G1 and suggest a role for neighboring cells in preventing differentiation of hESCs as they pass through a differentiation sensitive, G1 phase.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteína do Retinoblastoma/metabolismo , Técnicas de Cultura de Células , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Diferenciação Celular/fisiologia , Fase G1 , Humanos , Fosforilação
15.
Stem Cells ; 30(5): 898-909, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22367714

RESUMO

Approximately 6 million people worldwide are suffering from severe visual impairments or blindness due to corneal diseases. Corneal allogeneic transplantation is often required to restore vision; however, shortage in corneal grafts and immunorejections remain major challenges. The molecular basis of corneal diseases is poorly understood largely due to lack of appropriate cellular models. Here, we described a robust differentiation of human-induced pluripotent stem cells (hiPSCs) derived from hair follicles or skin fibroblasts into corneal epithelial-like cells. We found that BMP4, coupled with corneal fibroblast-derived conditioned medium and collagen IV allowed efficient corneal epithelial commitment of hiPSCs in a manner that recapitulated corneal epithelial lineage development with high purity. Organotypic reconstitution assays suggested the ability of these cells to stratify into a corneal-like epithelium. This model allowed us identifying miR-450b-5p as a molecular switch of Pax6, a major regulator of eye development. miR-450b-5p and Pax6 were reciprocally distributed at the presumptive epidermis and ocular surface, respectively. miR-450b-5p inhibited Pax6 expression and corneal epithelial fate in vitro, altogether, suggesting that by repressing Pax6, miR-450b-5p triggers epidermal specification of the ectoderm, while its absence allows ocular epithelial development. Additionally, miR-184 was detectable in early eye development and corneal epithelial differentiation of hiPSCs. The knockdown of miR-184 resulted in a decrease in Pax6 and K3, in line with recent findings showing that a point mutation in miR-184 leads to corneal dystrophy. Altogether, these data indicate that hiPSCs are valuable for modeling corneal development and may pave the way for future cell-based therapy.


Assuntos
Linhagem da Célula/fisiologia , Córnea/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , MicroRNAs/biossíntese , Modelos Biológicos , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular/fisiologia , Córnea/citologia , Proteínas do Olho/biossíntese , Proteínas do Olho/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Camundongos , MicroRNAs/genética , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição Box Pareados/genética , Células-Tronco Pluripotentes/citologia , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética
16.
Circulation ; 125(1): 87-99, 2012 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-22095829

RESUMO

BACKGROUND: Pericytes represent a unique subtype of microvessel-residing perivascular cells with diverse angiogenic functions and multilineage developmental features of mesenchymal stem cells. Although various protocols for derivation of endothelial and/or smooth muscle cells from human pluripotent stem cells (hPSC, either embryonic or induced) have been described, the emergence of pericytes in the course of hPSC maturation has not yet been elucidated. METHODS AND RESULTS: We found that during hPSC development, spontaneously differentiating embryoid bodies give rise to CD105(+)CD90(+)CD73(+)CD31(-) multipotent clonogenic mesodermal precursors, which can be isolated and efficiently expanded. Isolated and propagated cells expressed characteristic pericytic markers, including CD146, NG2, and platelet-derived growth factor receptor ß, but not the smooth muscle cell marker α-smooth muscle actin. Coimplantation of hPSC-derived endothelial cells with pericytes resulted in functional and rapid anastomosis to the murine vasculature. Administration of pericytes into immunodeficient mice with limb ischemia promoted significant vascular and muscle regeneration. At day 21 after transplantation, recruited hPSC pericytes were found incorporated into recovered muscle and vasculature. CONCLUSIONS: Derivation of vasculogenic and multipotent pericytes from hPSC can be used for the development of vasculogenic models using multiple vasculogenic cell types for basic research and drug screening and can contribute to angiogenic regenerative medicine.


Assuntos
Extremidades/irrigação sanguínea , Isquemia/cirurgia , Células-Tronco Multipotentes/transplante , Pericitos/transplante , Células-Tronco Pluripotentes/transplante , Recuperação de Função Fisiológica/fisiologia , Animais , Células Endoteliais/transplante , Extremidades/cirurgia , Humanos , Isquemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID
17.
Cell Stem Cell ; 9(4): 291-2, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21982227

RESUMO

In a recent study in Nature Biotechnology, Tang et al. (2011) describe a new marker of pluripotency, stage-specific embryonic antigen-5 (SSEA-5), and show that this oligosaccharide, together with two other surface antigens, can be used to remove all tumor-initiating cells from prospective cell transplants.

18.
Hum Reprod ; 26(10): 2874-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21784738

RESUMO

BACKGROUND: It is now well established that a GnRH agonist (GnRHa) ovulation trigger completely prevents ovarian hyperstimulation syndrome. However, early studies, using conventional luteal support, showed inferior clinical results following a GnRHa trigger compared with a conventional hCG trigger in normal responder IVF patients. We here present a novel approach for luteal support after a GnRHa trigger. METHODS Normal responder patients who failed at least one previous IVF attempt, during which a conventional hCG trigger was used, were consecutively enrolled in the study. A GnRH antagonist-based ovarian stimulation protocol was used in combination with a GnRHa trigger (Triptorelin 0.2 mg). The luteal phase was supported with a total of two boluses of 1500 IU hCG: on the day of oocyte retrieval and 4 days later. Neither progesterone nor estradiol was administered for luteal support. RESULTS: The mean age was 33.8 years. The mean (± SD) numbers of oocytes and fertilized oocytes were 6.7 (± 2.5) and 3.6 (± 1.7), respectively. All 15 patients had embryo transfers and 11 patients conceived. On the day of pregnancy test (14 days after retrieval), the mean serum E(2) and progesterone levels were 6607 (± 3789) and 182 (± 50) nmol/l, respectively. Of the pregnancies, seven are ongoing, while four ended as miscarriages. CONCLUSIONS: These preliminary results suggest that two boluses of 1500 IU hCG revert the luteolysis after a GnRHa trigger in the normo-responder patient. Importantly, no additional luteal support is needed. The novel concept combines the potential advantages of a physiological dual trigger (LH and FSH) with a simple, patient friendly, luteal support.


Assuntos
Gonadotropina Coriônica/metabolismo , Corpo Lúteo/metabolismo , Hormônio Liberador de Gonadotropina/agonistas , Ovulação/efeitos dos fármacos , Progesterona/metabolismo , Adulto , Estrogênios/metabolismo , Feminino , Fertilização , Fertilização in vitro/métodos , Humanos , Infertilidade/terapia , Oócitos/citologia , Síndrome de Hiperestimulação Ovariana/tratamento farmacológico , Gravidez , Resultado da Gravidez , Estudos Prospectivos
19.
Nat Protoc ; 6(5): 572-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21527915

RESUMO

Human pluripotent (embryonic or induced) stem cells (hPSCs) have many potential applications, not only for research purposes but also for clinical and industrial uses. While culturing these cells as undifferentiated lines, an adherent cell culture based on supportive layers or matrices is most often used. However, the use of hPSCs for industrial or clinical applications requires a scalable, reproducible and controlled process. Here we present a suspension culture system for undifferentiated hPSCs, based on a serum-free medium supplemented with interleukins and basic fibroblast growth factor, suitable for the mass production of these cells. The described system supports a suspension culture of hPSC lines, in both static and dynamic cultures. Results showed that hPSCs cultured with the described dynamic method maintained all hPSC features after 20 passages, including stable karyotype and pluripotency, and increased in cell numbers by 25-fold in 10 d. Thus, the described suspension method is suitable for large-scale culture of undifferentiated hPSCs.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Pluripotentes/citologia , Humanos , Teratoma/patologia
20.
J Cell Mol Med ; 15(1): 38-51, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20041972

RESUMO

In view of the therapeutic potential of cardiomyocytes derived from induced pluripotent stem (iPS) cells (iPS-derived cardiomyocytes), in the present study we investigated in iPS-derived cardiomyocytes, the functional properties related to [Ca(2+) ](i) handling and contraction, the contribution of the sarcoplasmic reticulum (SR) Ca(2+) release to contraction and the b-adrenergic inotropic responsiveness. The two iPS clones investigated here were generated through infection of human foreskin fibroblasts (HFF) with retroviruses containing the four human genes: OCT4, Sox2, Klf4 and C-Myc. Our major findings showed that iPS-derived cardiomyocytes: (i) express cardiac specific RNA and proteins; (ii) exhibit negative force-frequency relations and mild (compared to adult) post-rest potentiation; (iii) respond to ryanodine and caffeine, albeit less than adult cardiomyocytes, and express the SR-Ca(2+) handling proteins ryanodine receptor and calsequestrin. Hence, this study demonstrates that in our cardiomyocytes clones differentiated from HFF-derived iPS, the functional properties related to excitation-contraction coupling, resemble in part those of adult cardiomyocytes.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/metabolismo , Animais , Cafeína/farmacologia , Cálcio/metabolismo , Calsequestrina/genética , Calsequestrina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/metabolismo , Imunofluorescência , Prepúcio do Pênis/citologia , Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos SCID , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/metabolismo , Rianodina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Fatores de Transcrição SOXB1/genética , Retículo Sarcoplasmático/metabolismo , Teratoma/metabolismo , Teratoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA