Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Cytotherapy ; 26(1): 25-35, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37897472

RESUMO

BACKGROUND AIMS: There are currently no effective anti-viral treatments for coronavirus disease 2019 (COVID-19)-hospitalized patients with hypoxemia. Lymphopenia is a biomarker of disease severity usually present in patients who are hospitalized. Approaches to increasing lymphocytes exerting an anti-viral effect must be considered to treat these patients. Following our phase 1 study, we performed a phase 2 randomized multicenter clinical trial in which we evaluated the efficacy of the infusion of allogeneic off-the-shelf CD45RA- memory T cells containing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells from convalescent donors plus the standard of care (SoC) versus just the SoC treatment. METHODS: Eighty-four patients were enrolled in three Spanish centers. The patients were randomized into the infusion of 1 × 106/kg CD45RA- memory T cells or the SoC. We selected four unvaccinated donors based on the expression of interferon gamma SARS-CoV-2-specific response within the CD45RA- memory T cells and the most frequent human leukocyte antigen typing in the Spanish population. RESULTS: We analyzed data from 81 patients. The primary outcome for recovery, defined as the proportion of participants in each group with normalization of fever, oxygen saturation sustained for at least 24 hours and lymphopenia recovery through day 14 or at discharge, was met for the experimental arm. We also observed faster lymphocyte recovery in the experimental group. We did not observe any treatment-related adverse events. CONCLUSIONS: Adoptive cell therapy with off-the-shelf CD45RA- memory T cells containing SAR-CoV-2-specific T cells is safe, effective and accelerates lymphocyte recovery of patients with COVID-19 pneumonia and/or lymphopenia. TRIAL REGISTRATION: NCT04578210.


Assuntos
COVID-19 , Linfopenia , Humanos , SARS-CoV-2 , COVID-19/terapia , Células T de Memória , Resultado do Tratamento , Linfopenia/terapia , Antivirais
2.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38139339

RESUMO

Globally, a leg is amputated approximately every 30 seconds, with an estimated 85 percent of these amputations being attributed to complications arising from diabetic foot ulcers (DFU), as stated by the American Diabetes Association. Peripheral arterial disease (PAD) is a risk factor resulting in DFU and can, either independently or in conjunction with diabetes, lead to recurring, slow-healing ulcers and amputations. According to guidelines amputation is the recommended treatment for patients with no-option critical ischemia of the limb (CTLI). In this article we propose cell therapy as an alternative strategy for those patients. We also suggest the optimal time-frame for an effective therapy, such as implanting autologous mononuclear cells (MNCs), autologous and allogeneic mesenchymal stromal cells (MSC) as these treatments induce neuropathy relief, regeneration of the blood vessels and tissues, with accelerated ulcer healing, with no serious side effects, proving that advanced therapy medicinal product (ATMPs) application is safe and effective and, hence, can significantly prevent limb amputation.


Assuntos
Diabetes Mellitus , Pé Diabético , Doença Arterial Periférica , Doenças do Sistema Nervoso Periférico , Humanos , Pé Diabético/etiologia , Pé Diabético/terapia , Fatores de Risco , Doença Arterial Periférica/terapia , Doença Arterial Periférica/complicações , Doenças do Sistema Nervoso Periférico/complicações , Amputação Cirúrgica , Terapia Baseada em Transplante de Células e Tecidos , Isquemia/terapia , Isquemia/complicações
3.
Immunology ; 165(2): 234-249, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34775592

RESUMO

COVID-19 disease is the manifestation of syndrome coronavirus 2 (SARS-CoV-2) infection, which is causing a worldwide pandemic. This disease can lead to multiple and different symptoms, being lymphopenia associated with severity one of the most persistent. Natural killer cells (NK cells) are part of the innate immune system, being fighting against virus-infected cells one of their key roles. In this study, we determined the phenotype of NK cells after COVID-19 and the main characteristic of SARS-CoV-2-specific-like NK population in the blood of convalescent donors. CD57+ NKG2C+ phenotype in SARS-CoV-2 convalescent donors indicates the presence of 'memory'/activated NK cells as it has been shown for cytomegalovirus infections. Although the existence of this population is donor dependent, its expression may be crucial for the specific response against SARS-CoV-2, so that, it gives us a tool for selecting the best donors to produce off-the-shelf living drug for cell therapy to treat COVID-19 patients under the RELEASE clinical trial (NCT04578210).


Assuntos
Transferência Adotiva , Doadores de Sangue , COVID-19/imunologia , Convalescença , Memória Imunológica , Células Matadoras Naturais/imunologia , SARS-CoV-2/imunologia , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
4.
Int J Mol Sci ; 23(11)2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35682603

RESUMO

Mesothelial cells form the mesothelium, a simple epithelium lining the walls of serous cavities and the surface of visceral organs. Although mesothelial cells are phenotypically well characterized, their immunoregulatory properties remain largely unknown, with only two studies reporting their capacity to inhibit T cells through TGF-ß and their consumption of L-arginine by arginase-1. Whether human mesothelial cells can suppress other immune cells and possess additional leukosuppressive mechanisms, remain to be addressed to better delineate their therapeutic potential for cell therapy. Herein, we generated secretomes from omental mesothelial cells (OMC) and assess their capacity to inhibit lymphocytes proliferation, suppress activated T and B cells, as well as to modify macrophage activation markers. The secretome from mesenchymal stromal cells (MSC) served as a control of immuno-suppression. Although OMC and MSC were phenotypically divergent, their cytokine secretion patterns as well as expression of inflammatory and immunomodulary genes were similar. As such, OMC- and MSC-derived secretomes (OMC-S and MSC-S) both polarized RAW 264.7 macrophages towards a M2-like anti-inflammatory phenotype and suppressed mouse and human lymphocytes proliferation. OMC-S displayed a strong ability to suppress mouse- and human-activated CD19+/CD25+ B cells as compared to MSC-S. The lymphosuppressive activity of the OMC-S could be significantly counteracted either by SB-431542, an inhibitor of TGFß and activin signaling pathways, or with a monoclonal antibody against the TGFß1, ß2, and ß3 isoforms. A strong blockade of the OMC-S-mediated lymphosuppressive activity was achieved using L-NMMA, a specific inhibitor of nitric oxide synthase (NOS). Taken together, our results suggest that OMC are potent immunomodulators.


Assuntos
Imunomodulação , Células-Tronco Mesenquimais , Animais , Humanos , Ativação Linfocitária , Ativação de Macrófagos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Linfócitos T
5.
Int J Mol Sci ; 23(7)2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35409280

RESUMO

The transcription factor, early growth response-1 (EGR-1), is involved in the regulation of cell differentiation, proliferation, and apoptosis in response to different stimuli. EGR-1 is described to be involved in pancreatic endoderm differentiation, but the regulatory mechanisms controlling its action are not fully elucidated. Our previous investigation reported that exposure of mouse embryonic stem cells (mESCs) to the chemical nitric oxide (NO) donor diethylenetriamine nitric oxide adduct (DETA-NO) induces the expression of early differentiation genes such as pancreatic and duodenal homeobox 1 (Pdx1). We have also evidenced that Pdx1 expression is associated with the release of polycomb repressive complex 2 (PRC2) and P300 from the Pdx1 promoter; these events were accompanied by epigenetic changes to histones and site-specific changes in the DNA methylation. Here, we investigate the role of EGR-1 on Pdx1 regulation in mESCs. This study reveals that EGR-1 plays a negative role in Pdx1 expression and shows that the binding capacity of EGR-1 to the Pdx1 promoter depends on the methylation level of its DNA binding site and its acetylation state. These results suggest that targeting EGR-1 at early differentiation stages might be relevant for directing pluripotent cells into Pdx1-dependent cell lineages.


Assuntos
Endoderma , Células-Tronco Embrionárias Murinas , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias , Endoderma/metabolismo , Camundongos , Óxido Nítrico/metabolismo
6.
Diabetologia ; 62(9): 1667-1680, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31250031

RESUMO

AIMS/HYPOTHESIS: Bisphenol-A (BPA) is a widespread endocrine-disrupting chemical that has been associated with type 2 diabetes development. Low doses of BPA modify pancreatic beta cell function and induce insulin resistance; some of these effects are mediated via activation of oestrogen receptors α (ERα) and ß (ERß). Here we investigated whether low doses of BPA regulate the expression and function of ion channel subunits involved in beta cell function. METHODS: Microarray gene profiling of isolated islets from vehicle- and BPA-treated (100 µg/kg per day for 4 days) mice was performed using Affymetrix GeneChip Mouse Genome 430.2 Array. Expression level analysis was performed using the normalisation method based on the processing algorithm 'robust multi-array average'. Whole islets or dispersed islets from C57BL/6J or oestrogen receptor ß (ERß) knockout (Erß-/-) mice were treated with vehicle or BPA (1 nmol/l) for 48 h. Whole-cell patch-clamp recordings were used to measure Na+ and K+ currents. mRNA expression was evaluated by quantitative real-time PCR. RESULTS: Microarray analysis showed that BPA modulated the expression of 1440 probe sets (1192 upregulated and 248 downregulated genes). Of these, more than 50 genes, including Scn9a, Kcnb2, Kcnma1 and Kcnip1, encoded important Na+ and K+ channel subunits. These findings were confirmed by quantitative RT-PCR in islets from C57BL/6J BPA-treated mice or whole islets treated ex vivo. Electrophysiological measurements showed a decrease in both Na+ and K+ currents in BPA-treated islets. The pharmacological profile indicated that BPA reduced currents mediated by voltage-activated K+ channels (Kv2.1/2.2 channels) and large-conductance Ca2+-activated K+ channels (KCa1.1 channels), which agrees with BPA's effects on gene expression. Beta cells from ERß-/- mice did not present BPA-induced changes, suggesting that ERß mediates BPA's effects in pancreatic islets. Finally, BPA increased burst duration, reduced the amplitude of the action potential and enlarged the action potential half-width, leading to alteration in beta cell electrical activity. CONCLUSIONS/INTERPRETATION: Our data suggest that BPA modulates the expression and function of Na+ and K+ channels via ERß in mouse pancreatic islets. Furthermore, BPA alters beta cell electrical activity. Altogether, these BPA-induced changes in beta cells might play a role in the diabetogenic action of BPA described in animal models.


Assuntos
Compostos Benzidrílicos/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Receptor beta de Estrogênio/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Fenóis/farmacologia , Animais , Receptor alfa de Estrogênio/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Potássio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Sódio/metabolismo
7.
Mol Ther ; 26(11): 2696-2709, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30195725

RESUMO

Diabetes is a chronic metabolic disorder that affects 415 million people worldwide. This pathology is often associated with long-term complications, such as critical limb ischemia (CLI), which increases the risk of limb loss and mortality. Mesenchymal stromal cells (MSCs) represent a promising option for the treatment of diabetes complications. Although MSCs are widely used in autologous cell-based therapy, their effects may be influenced by the constant crosstalk between the graft and the host, which could affect the MSC fate potential. In this context, we previously reported that MSCs derived from diabetic patients with CLI have a defective phenotype that manifests as reduced fibrinolytic activity, thereby enhancing the thrombotic risk and compromising patient safety. Here, we found that MSCs derived from diabetic patients with CLI not only exhibit a prothrombotic profile but also have altered multi-differentiation potential, reduced proliferation, and inhibited migration and homing to sites of inflammation. We further demonstrated that this aberrant cell phenotype is reversed by the platelet-derived growth factor (PDGF) BB, indicating that PDGF signaling is a key regulator of MSC functionality. These findings provide an attractive approach to improve the therapeutic efficacy of MSCs in autologous therapy for diabetic patients.


Assuntos
Diabetes Mellitus/genética , Inflamação/genética , Células-Tronco Mesenquimais/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Complicações do Diabetes/genética , Complicações do Diabetes/patologia , Complicações do Diabetes/terapia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Diabetes Mellitus/terapia , Humanos , Inflamação/patologia , Inflamação/terapia , Camundongos , Camundongos SCID , Osteogênese/genética , Fenótipo , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Transdução de Sinais , Cicatrização/genética
8.
Differentiation ; 92(5): 249-256, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27181524

RESUMO

Efficient induction of defined lineages in pluripotent stem cells constitutes the determinant step for the generation of therapeutically relevant replacement cells to potentially treat a wide range of diseases, including diabetes. Pancreatic differentiation has remained an important challenge in large part because of the need to differentiate uncommitted pluripotent stem cells into highly specialized hormone-secreting cells, which has been shown to require a developmentally informed step-by-step induction procedure. Here, in the framework of using induced pluripotent stem cells (iPSCs) to generate pancreatic cells for pancreatic diseases, we have generated and characterized iPSCs from Pdx1-GFP transgenic mice. The use of a GFP reporter knocked into the endogenous Pdx1 promoter allowed us to monitor pancreatic induction based on the expression of Pdx1, a pancreatic master transcription factor, and to isolate a pure Pdx1-GFP+ population for downstream applications. Differentiated cultures timely expressed markers specific to each stage and end-stage progenies acquired a rather immature beta-cell phenotype, characterized by polyhormonal expression even among cells highly expressing the Pdx1-GFP reporter. Our findings highlight the utility of employing a fluorescent protein reporter under the control of a master developmental gene in order to devise novel differentiation protocols for relevant cell types for degenerative diseases such as pancreatic beta cells for diabetes.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/citologia , Pâncreas/crescimento & desenvolvimento , Transativadores/genética , Animais , Células-Tronco Embrionárias/citologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Pâncreas/citologia , Regiões Promotoras Genéticas
9.
Int J Mol Sci ; 18(6)2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28574454

RESUMO

Consistent evidence from both experimental and human studies indicates that Type 2 diabetes mellitus (T2DM) is a complex disease resulting from the interaction of genetic, epigenetic, environmental, and lifestyle factors. Nutrients and dietary patterns are important environmental factors to consider in the prevention, development and treatment of this disease. Nutritional genomics focuses on the interaction between bioactive food components and the genome and includes studies of nutrigenetics, nutrigenomics and epigenetic modifications caused by nutrients. There is evidence supporting the existence of nutrient-gene and T2DM interactions coming from animal studies and family-based intervention studies. Moreover, many case-control, cohort, cross-sectional cohort studies and clinical trials have identified relationships between individual genetic load, diet and T2DM. Some of these studies were on a large scale. In addition, studies with animal models and human observational studies, in different countries over periods of time, support a causative relationship between adverse nutritional conditions during in utero development, persistent epigenetic changes and T2DM. This review provides comprehensive information on the current state of nutrient-gene interactions and their role in T2DM pathogenesis, the relationship between individual genetic load and diet, and the importance of epigenetic factors in influencing gene expression and defining the individual risk of T2DM.


Assuntos
Diabetes Mellitus Tipo 2/genética , Dieta , Regulação da Expressão Gênica , Nutrigenômica , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Dieta/efeitos adversos , Epigênese Genética , Predisposição Genética para Doença , Variação Genética , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Nutrigenômica/métodos
10.
J Cell Biochem ; 117(9): 2078-88, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26853909

RESUMO

Nitric oxide (NO) delays mouse embryonic stem cell (mESC) differentiation by regulating genes linked to pluripotency and differentiation. Nevertheless, no profound study has been conducted on cell differentiation regulation by this molecule through signaling on essential biological functions. We sought to demonstrate that NO positively regulates the pluripotency transcriptional core, enforcing changes in the chromatin structure, in addition to regulating cell proliferation, and signaling pathways with key roles in stemness. Culturing mESCs with 2 µM of the NO donor diethylenetriamine/NO (DETA/NO) in the absence of leukemia inhibitory factor (LIF) induced significant changes in the expression of 16 genes of the pluripotency transcriptional core. Furthermore, treatment with DETA/NO resulted in a high occupancy of activating H3K4me3 at the Oct4 and Nanog promoters and repressive H3K9me3 and H3k27me3 at the Brachyury promoter. Additionally, the activation of signaling pathways involved in pluripotency, such as Gsk3-ß/ß-catenin, was observed, in addition to activation of PI3 K/Akt, which is consistent with the protection of mESCs from cell death. Finally, a decrease in cell proliferation coincides with cell cycle arrest in G2/M. Our results provide novel insights into NO-mediated gene regulation and cell proliferation and suggest that NO is necessary but not sufficient for the maintenance of pluripotency and the prevention of cell differentiation. J. Cell. Biochem. 117: 2078-2088, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Expressão Gênica/fisiologia , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Células-Tronco Embrionárias Murinas/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triazenos/farmacologia
11.
Eur J Immunol ; 45(8): 2324-34, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25991034

RESUMO

NK cells play a key role in innate elimination of virally infected or neoplastic cells but they can be circumvented by immunoevasive mechanisms enabling viral spread or tumor progression. Engagement of the NKG2D activating receptor with soluble forms of its ligand is one such mechanism of inducing NK cell hyporesponsiveness. Interestingly, this immunoevasive strategy among others is described at the maternal-fetal interface where tolerance of the semi-allogeneic fetus is required to allow successful human pregnancy. Understanding of maternal-fetal tolerance is increasing but mechanisms preventing alloreactivity of fetal immune cells against the maternal host are less well understood. The study of umbilical cord blood has enabled insight of the fetal immune system, which appears immature and inert. We have found that soluble NKG2D ligands (sNKG2DLs) are present in cord blood plasma (CBP) and associate with adult NK cell hyporesponsiveness demonstrated by reduced CD107a expression and secretion of IFN-γ upon stimulation. The capacity of NK cells to kill K562 cells or proliferate was also reduced by incubation with CBP; however, physical removal of sNKG2DL from CBP restored K562 lytic function and NKG2D expression. Therefore, our results strongly suggest sNKG2DLs are expressed in CBP as a mechanism of fetal-maternal tolerance in human pregnancy.


Assuntos
Sangue Fetal , Imunidade Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Células Matadoras Naturais , Troca Materno-Fetal/fisiologia , Adulto , Feminino , Sangue Fetal/imunologia , Sangue Fetal/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Células K562 , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/biossíntese , Proteína 1 de Membrana Associada ao Lisossomo/imunologia , Masculino , Subfamília K de Receptores Semelhantes a Lectina de Células NK/sangue , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Gravidez
12.
J Membr Biol ; 248(4): 671-82, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25666166

RESUMO

Mouse embryonic stem cells (mESCs) are capable of both self-renewal and multilineage differentiation; thus, they can be expanded in vivo or in vitro and differentiated to produce different cell types. Despite their biological and medical interest, many physiological properties of undifferentiated mESCs, such as ion channel function, are not fully understood. Ion channels are thought to be involved in cell proliferation and differentiation. The aim of this study was to characterize functional ion channels in cultured undifferentiated mESCs and their role in cell proliferation. L-type voltage-activated Ca(2+) channels sensitive to nifedipine and small-conductance Ca(2+)-activated K(+) (SK) channels sensitive to apamin were identified. Ca(2+)-activated K(+) currents were blocked by millimolar concentrations of tetraethylammonium. The effects of Ca(2+) channel and Ca(2+)-activated K(+) channel blockers on the proliferation of undifferentiated mESCs were investigated by bromodeoxyuridine (BrdU) incorporation. Dihydropyridine derivatives, such as nifedipine, inhibited cell growth and BrdU incorporation into the cells, whereas apamin, which selectively blocks SK channels, had no effect on cell growth. These results demonstrate that functional voltage-operated Ca(2+) channels and Ca(2+)-activated K(+) channels are present in undifferentiated mESCs. Moreover, voltage-gated L-type Ca(2+) channels, but not SK channels, might be necessary for proliferation of undifferentiated mESCs.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proliferação de Células/fisiologia , Células-Tronco Embrionárias Murinas/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Tetraetilamônio/farmacologia
13.
Hepatology ; 59(6): 2358-70, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24415412

RESUMO

UNLABELLED: The zinc finger transcription factor GATA4 controls specification and differentiation of multiple cell types during embryonic development. In mouse embryonic liver, Gata4 is expressed in the endodermal hepatic bud and in the adjacent mesenchyme of the septum transversum. Previous studies have shown that Gata4 inactivation impairs liver formation. However, whether these defects are caused by loss of Gata4 in the hepatic endoderm or in the septum transversum mesenchyme remains to be determined. In this study, we have investigated the role of mesenchymal GATA4 activity in liver formation. We have conditionally inactivated Gata4 in the septum transversum mesenchyme and its derivatives by using Cre/loxP technology. We have generated a mouse transgenic Cre line, in which expression of Cre recombinase is controlled by a previously identified distal Gata4 enhancer. Conditional inactivation of Gata4 in hepatic mesenchymal cells led to embryonic lethality around mouse embryonic stage 13.5, likely as a consequence of fetal anemia. Gata4 knockout fetal livers exhibited reduced size, advanced fibrosis, accumulation of extracellular matrix components and hepatic stellate cell (HSC) activation. Haploinsufficiency of Gata4 accelerated CCl4 -induced liver fibrosis in adult mice. Moreover, Gata4 expression was dramatically reduced in advanced hepatic fibrosis and cirrhosis in humans. CONCLUSIONS: Our data demonstrate that mesenchymal GATA4 activity regulates HSC activation and inhibits the liver fibrogenic process.


Assuntos
Regulação para Baixo , Fator de Transcrição GATA4/fisiologia , Cirrose Hepática/metabolismo , Fígado/embriologia , Animais , Intoxicação por Tetracloreto de Carbono/complicações , Linhagem Celular , Matriz Extracelular/metabolismo , Células Estreladas do Fígado/fisiologia , Humanos , Integrases , Cirrose Hepática/etiologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo
14.
Cell Mol Life Sci ; 71(13): 2383-402, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24221136

RESUMO

The field of pancreas development has markedly expanded over the last decade, significantly advancing our understanding of the molecular mechanisms that control pancreas organogenesis. This growth has been fueled, in part, by the need to generate new therapeutic approaches for the treatment of diabetes. The creation of sophisticated genetic tools in mice has been instrumental in this progress. Genetic manipulation involving activation or inactivation of genes within specific cell types has allowed the identification of many transcription factors (TFs) that play critical roles in the organogenesis of the pancreas. Interestingly, many of these TFs act at multiple stages of pancreatic development, and adult organ function or repair. Interaction with other TFs, extrinsic signals, and epigenetic regulation are among the mechanisms by which TFs may play context-dependent roles during pancreas organogenesis. Many of the pancreatic TFs directly regulate each other and their own expression. These combinatorial interactions generate very specific gene regulatory networks that can define the different cell lineages and types in the developing pancreas. Here, we review recent progress made in understanding the role of pancreatic TFs in mouse pancreas formation. We also summarize our current knowledge of human pancreas development and discuss developmental pancreatic TFs that have been associated with human pancreatic diseases.


Assuntos
Diabetes Mellitus/genética , Organogênese/genética , Pâncreas/crescimento & desenvolvimento , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Diabetes Mellitus/terapia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Mamíferos , Camundongos
15.
J Membr Biol ; 246(3): 215-30, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23188062

RESUMO

Cell-attached and inside-out patch clamp recording was used to compare the functional expression of membrane ion channels in mouse and human embryonic stem cells (ESCs). Both ESCs express mechanosensitive Ca²âº permeant cation channels (MscCa) and large conductance (200 pS) Ca²âº-sensitive K⁺ (BK(Ca²âº)) channels but with markedly different patch densities. MscCa is expressed at higher density in mESCs compared with hESCs (70 % vs. 3 % of patches), whereas the BK(Ca²âº) channel is more highly expressed in hESCs compared with mESCs (~50 % vs. 1 % of patches). ESCs of both species express a smaller conductance (25 pS) nonselective cation channel that is activated upon inside-out patch formation but is neither mechanosensitive nor strictly Ca²âº-dependent. The finding that mouse and human ESCs express different channels that sense membrane tension and intracellular [Ca²âº] may contribute to their different patterns of growth and differentiation in response to mechanical and chemical cues.


Assuntos
Canais de Cálcio/metabolismo , Células-Tronco Embrionárias/fisiologia , Mecanotransdução Celular/fisiologia , Animais , Humanos , Potenciais da Membrana/fisiologia , Camundongos , Técnicas de Patch-Clamp
16.
Br Med Bull ; 105: 85-105, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23184855

RESUMO

INTRODUCTION: New therapies with genes, tissues and cells have taken the emerging field for the treatment of many diseases. Advances on stem cell therapy research have led to international regulatory agencies to harmonize and regulate the development of new medicines with stem cells. SOURCES OF DATA: European Medicines Agency on September 15, 2012. AREAS OF AGREEMENT: Cell therapy medicinal products should be subjected to the same regulatory principles than any other medicine. AREAS OF CONTROVERSY: Their technical requirements for quality, safety and efficacy must be more specific and stringent than other biologic products and medicines. GROWING POINTS: Cell therapy medicinal products are at the cutting edge of innovation and offer a major hope for various diseases for which there are limited or no therapeutic options. AREAS TIMELY FOR DEVELOPING RESEARCH: The development of cell therapy medicinal products constitutes an alternative therapeutic strategy to conventional clinical therapy, for which no effective cure was previously available.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/estatística & dados numéricos , Doença Crônica/terapia , Qualidade de Produtos para o Consumidor/legislação & jurisprudência , Qualidade de Produtos para o Consumidor/normas , União Europeia , Humanos , Legislação de Medicamentos , Medicina , Saúde Pública/legislação & jurisprudência
17.
Proc Natl Acad Sci U S A ; 107(31): 13736-41, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20631301

RESUMO

The longevity-promoting NAD+-dependent class III histone deacetylase Sirtuin 1 (SIRT1) is involved in stem cell function by controlling cell fate decision and/or by regulating the p53-dependent expression of NANOG. We show that SIRT1 is down-regulated precisely during human embryonic stem cell differentiation at both mRNA and protein levels and that the decrease in Sirt1 mRNA is mediated by a molecular pathway that involves the RNA-binding protein HuR and the arginine methyltransferase coactivator-associated arginine methyltransferase 1 (CARM1). SIRT1 down-regulation leads to reactivation of key developmental genes such as the neuroretinal morphogenesis effectors DLL4, TBX3, and PAX6, which are epigenetically repressed by this histone deacetylase in pluripotent human embryonic stem cells. Our results indicate that SIRT1 is regulated during stem cell differentiation in the context of a yet-unknown epigenetic pathway that controls specific developmental genes in embryonic stem cells.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sirtuína 1/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Linhagem Celular , Guanilato Ciclase/metabolismo , Humanos , Camundongos , Camundongos Knockout , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Estabilidade de RNA , Sirtuína 1/deficiência , Sirtuína 1/genética
18.
Front Immunol ; 14: 1232472, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37767093

RESUMO

An unprecedented global social and economic impact as well as a significant number of fatalities have been brought on by the coronavirus disease 2019 (COVID-19), produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Acute SARS-CoV-2 infection can, in certain situations, cause immunological abnormalities, leading to an anomalous innate and adaptive immune response. While most patients only experience mild symptoms and recover without the need for mechanical ventilation, a substantial percentage of those who are affected develop severe respiratory illness, which can be fatal. The absence of effective therapies when disease progresses to a very severe condition coupled with the incomplete understanding of COVID-19's pathogenesis triggers the need to develop innovative therapeutic approaches for patients at high risk of mortality. As a result, we investigate the potential contribution of promising combinatorial cell therapy to prevent death in critical patients.

19.
Lancet Reg Health Am ; 15: 100347, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36778067

RESUMO

Background: Malaria is one of the biggest impediments to global progress. In Peru, it is still a major public health problem. Measures of health and economic burden due to malaria are relevant considerations for the assessment of current policies. Methods: We used estimates from the Global Burden of Diseases Study 2019 for malaria in Peru, grouped by gender and age, from 1990 to 2019. Results are presented as absolute numbers and age-standardized rates with 95% uncertainty intervals (UI). We collected economic data from the World Bank and The National Institute of Statistics and Informatics of Peru and Loreto to calculate the economic burden of productivity loss (EBPL) using the human capital approach. Economic values were presented in constant dollars, soles, and percentages. Findings: Rates of deaths, years of life lost (YLLs), years lived with disability (YLDs), and disability-adjusted life years (DALYs), as well as the EBPL, were drastically reduced from 1990 to 2019. DALYs had a greater percentage of YLDs in 2019 than in 1990. DALYs rates showed no preference between sexes, but the "< 1 year" age group had the highest DALYs values over the study period. We found that the EBPL due to malaria for Loreto was considerably higher than Peru's in terms of GDP percentage. Interpretation: Our study shows that the fight against malaria in Peru reduced remarkably the impact of the disease since 1990; however, during the last decade the estimates were stable or even increased. Our results help to measure the malaria impact on the health status of the Peruvian population as well as the economic pressure that it exerts, constituting remarkable tools for policymaking aimed at reducing the burden of this disease. Strengthening the malaria elimination program is important to achieve the elimination of the disease in the coming years. Funding: This study was supported by the Universidad Nacional Toribio Rodríguez de Mendoza and FONDECYT: Contrato Nº 09-2019-FONDECYT-BMINC.INV and FONDECYT-BM, Perú (Program INCORPORACIÓN DE INVESTIGADORES E038-2019-01, Registry Number: 64007).

20.
Cryobiology ; 62(2): 145-51, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21315706

RESUMO

Cryobanking skin samples permit preserving a maximum of genetic representation from the population biodiversity. This is a relevant aspect for threatened species, potentially menaced by an epizooty and from which it is difficult to obtain gametes. As a first step for properly cryobanking skin samples of a given species, the optimal conditions of culture and freezing have to be studied by covering a broad range of possibilities. This paper presents, for the first time, a systematic study of such conditions for the Iberian lynx (Lynx pardinus). To that end, we have analyzed twenty different culture conditions and fifteen different freezing solutions for skin explants, as well as three freezing solutions for isolated cells derived from them. The culture conditions included both two different culture strategies and several combinations of nutritional supplements and mitotic agents. For the freezing solutions, we have considered different concentrations of the permeating cryoprotectant dimethyl sulfoxide (Me(2)SO) either alone (5%, 7.5%, 10%, 12.5% and 15% v/v for explants, 10% for isolated cells) or along with the non-permeating cryoprotectant sucrose (0.1 or 0.2M). Our results have been analyzed through several quantitative parameters and show that only thawed explants cryopreserved in Me(2)SO (10%) either alone or with sucrose (0.2M) presented similar properties to those in optimal fresh cultures. In addition, for these freezing conditions, isolated thawed cells also presented high survival rates (90%) and percentages of cellular functionality (85%). These results, focussed on the most endangered felid in the world, could be also useful for other threatened/endangered species.


Assuntos
Criopreservação/métodos , Criopreservação/veterinária , Crioprotetores/farmacologia , Dimetil Sulfóxido/farmacologia , Espécies em Perigo de Extinção , Lynx , Pele/patologia , Sacarose/farmacologia , Animais , Biópsia/veterinária , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Conservação dos Recursos Naturais , Congelamento , Variação Genética , Células Germinativas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA