Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38496665

RESUMO

The cloaca is a transient structure that forms in the terminal hindgut giving rise to the rectum dorsally and the urogenital sinus ventrally. Similarly, human hindgut cultures derived from human pluripotent stem cells generate human colonic organoids (HCOs) which also contain co-developing urothelial tissue. In this study, our goal was to identify pathways involved in cloacal patterning and apply this to human hindgut cultures. RNA-seq data comparing dorsal versus ventral cloaca in e10.5 mice revealed that WNT signaling was elevated in the ventral versus dorsal cloaca. Inhibition of WNT signaling in hindgut cultures biased their differentiation towards a colorectal fate. WNT activation biased differentiation towards a urothelial fate, giving rise to human urothelial organoids (HUOs). HUOs contained cell types present in human urothelial tissue. Based on our results, we propose a mechanism whereby WNT signaling patterns the ventral cloaca, prior to cloacal septation, to give rise to the urogenital sinus.

2.
Nano Lett ; 23(6): 2321-2331, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36893018

RESUMO

Emerging heart-on-a-chip technology is a promising tool to establish in vitro cardiac models for therapeutic testing and disease modeling. However, due to the technical complexity of integrating cell culture chambers, biosensors, and bioreactors into a single entity, a microphysiological system capable of reproducing controlled microenvironmental cues to regulate cell phenotypes, promote iPS-cardiomyocyte maturity, and simultaneously measure the dynamic changes of cardiomyocyte function in situ is not available. This paper reports an ultrathin and flexible bioelectronic array platform in 24-well format for higher-throughput contractility measurement under candidate drug administration or defined microenvironmental conditions. In the array, carbon black (CB)-PDMS flexible strain sensors were embedded for detecting iPSC-CM contractility signals. Carbon fiber electrodes and pneumatic air channels were integrated to provide electrical and mechanical stimulation to improve iPSC-CM maturation. Performed experiments validate that the bioelectronic array accurately reveals the effects of cardiotropic drugs and identifies mechanical/electrical stimulation strategies for promoting iPSC-CM maturation.


Assuntos
Técnicas Biossensoriais , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Técnicas de Cultura de Células , Preparações Farmacêuticas , Diferenciação Celular
3.
J Vis Exp ; (173)2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34309606

RESUMO

Intestinal regional specification describes a process through which unique morphology and function are imparted to defined areas of the developing gastrointestinal (GI) tract. Regional specification in the intestine is driven by multiple developmental pathways, including the bone morphogenetic protein (BMP) pathway. Based on normal regional specification, a method to generate human colonic organoids (HCOs) from human pluripotent stem cells (hPSCs), which include human embryonic stem cells (hES) and induced pluripotent stem cells (iPSCs), was developed. A three-day induction of BMP signaling sufficiently patterns mid/hindgut tube cultures into special AT-rich sequence-binding protein 2 (SATB2)-expressing HCOs containing all of the main epithelial cell types present in human colon as well as co-developing mesenchymal cells. Omission of BMP (or addition of the BMP inhibitor NOGGIN) during this critical patterning period resulted in the formation of human intestinal organoids (HIOs). HIOs and HCOs morphologically and molecularly resemble human developing small intestine and colon, respectively. Despite the utility of HIOs and HCOs for studying human intestinal development, the generation of HIOs and HCOs is challenging. This paper presents methods for generating, maintaining, and characterizing HIOs and HCOs. In addition, the critical steps in the protocol and troubleshooting recommendations are provided.


Assuntos
Organoides , Células-Tronco Pluripotentes , Diferenciação Celular , Colo , Endoderma , Humanos , Intestinos
4.
Methods Cell Biol ; 159: 201-227, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32586443

RESUMO

Advances in human pluripotent stem cell (hPSC) biology now allow the generation of organoids that resemble different regions of the gastrointestinal tract. Generation of region-specific organoids has been facilitated by developmental biology studies carried out in model organisms such as mouse, frog and chick. By mimicking embryonic development, hPSC-derived human colonic organoids (HCOs) can be generated through a stepwise differentiation, first into definitive endoderm (DE), then into mid/hindgut spheroids which are then patterned into posterior gut tissue which gives rise to HCOs following prolonged in vitro culture. HCOs undergo transitions similar to those observed in the developing colon of model organisms and human embryos. HCOs develop into tissue that resembles fetal colon on the basis of morphology, gene expression and presence of differentiated cell types. Generation of HCOs without the proper training or expertise can be a daunting task. Here, we describe a detailed protocol for differentiating hPSCs into HCOs, we include suggestions for troubleshooting these differentiations, and we discuss experimental design considerations. We have also highlighted the key advantages and limitations of the system.


Assuntos
Técnicas de Cultura de Células/métodos , Colo/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular , Endoderma/citologia , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Humanos , Organoides/metabolismo , Células-Tronco Pluripotentes/metabolismo , Esferoides Celulares/citologia
5.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-32023984

RESUMO

The tumor microenvironment has been recently recognized as a critical contributor to cancer progression and anticancer therapy-resistance. Cyclophosphamide (CTX) is a cytotoxic agent commonly used in clinics for the treatment of cancer. Previous reports demonstrated that CTX given at low continuous doses, known as metronomic schedule, mainly targets endothelial cells and circulating Tregs with unknown mechanisms. Here, we investigated the antitumor activity of two different metronomic schedules of CTX along with their corresponding MTD regimen and further explored their effect on immune function and tumor microenvironment. Toxicity evaluation was monitored by overall survival rate, weight loss, and histopathological analysis. A nude mouse model of Lewis lung cancer was established to assess the anti-metastatic effects of CTX in vivo. CD4+, CD8+, and CD4+CD25+FoxP3 T cells were selected by flow cytometry analysis. Low and continuous administration of CTX was able to restore immune function via increase of CD4+/CD8+ T cells and depletion of T regulatory cells, not only in circulatory and splenic compartments, but also at the tumor site. Low-dose CTX also reduced myofibroblasts, accompanied with an increased level of E-cadherin and low N-cadherin, both in the primary tumor and lung through the TGF-ß pathway by the downregulated expression of TGF-ß receptor 2. Our data may indicate that several other molecular mechanisms of CTX for tumor may be involved in metronomic chemotherapy, besides targeting angiogenesis and regulatory T cells.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Ciclofosfamida/administração & dosagem , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Administração Metronômica , Animais , Antineoplásicos Alquilantes/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclofosfamida/farmacologia , Regulação para Baixo , Camundongos , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
6.
Curr Mol Pharmacol ; 12(3): 215-229, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30767757

RESUMO

BACKGROUND: Epigenetic alterations comprise key regulatory events that dynamically alter gene expression and their deregulation is commonly linked to the pathogenesis of various diseases, including cancer. Unlike DNA mutations, epigenetic alterations involve modifications to proteins and nucleic acids that regulate chromatin structure without affecting the underlying DNA sequence, altering the accessibility of the transcriptional machinery to the DNA, thus modulating gene expression. In cancer cells, this often involves the silencing of tumor suppressor genes or the increased expression of genes involved in oncogenesis. Advances in laboratory medicine have made it possible to map critical epigenetic events, including histone modifications and DNA methylation, on a genome-wide scale. Like the identification of genetic mutations, mapping of changes to the epigenetic landscape has increased our understanding of cancer progression. However, in contrast to irreversible genetic mutations, epigenetic modifications are flexible and dynamic, thereby making them promising therapeutic targets. Ongoing studies are evaluating the use of epigenetic drugs in chemotherapy sensitization and immune system modulation. With the preclinical success of drugs that modify epigenetics, along with the FDA approval of epigenetic drugs including the DNA methyltransferase 1 (DNMT1) inhibitor 5-azacitidine and the histone deacetylase (HDAC) inhibitor vorinostat, there has been a rise in the number of drugs that target epigenetic modulators over recent years. CONCLUSION: We provide an overview of epigenetic modulations, particularly those involved in cancer, and discuss the recent advances in drug development that target these chromatin-modifying events, primarily focusing on novel strategies to regulate the epigenome.


Assuntos
Antineoplásicos/uso terapêutico , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Neoplasias/genética
7.
Front Med (Lausanne) ; 6: 297, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31956653

RESUMO

In recent years, advances in human pluripotent stem cell (hPSC) biology have enabled the generation of gastrointestinal (GI) organoids which recapitulate aspects of normal organ development. HPSC derived gastrointestinal organoids are comprised of epithelium and mesenchyme and have a remarkable ability to self-organize and recapitulate early stages of human intestinal development. Furthermore, hPSC derived organoids can be transplanted into immunocompromised mice which allows further maturation of both the epithelium and mesenchyme. In this review, we will briefly summarize work from model systems which has elucidated mechanisms of GI patterning and how these insights have been used to guide the differentiation of hPSCs into organoids resembling small intestine and colon. We will succinctly discuss how developmental principles have been used to promote maturation of human intestinal organoids (HIOs) in vitro as well as to introduce an enteric nervous system into HIOs. We will then concisely review how organoids have been used to study human pathogens, how new genetic and bioengineering tools are being applied to organoid research, and how this integration has allowed researchers to elucidate mechanisms of human development and disease. Finally, we will briefly discuss remaining challenges in the field and how they can be addressed. HPSC derived organoids are promising new model systems which hold the potential of unlocking unknown mechanisms of human gastrointestinal development and disease.

8.
Front Pharmacol ; 9: 788, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30079021

RESUMO

Targeting tumor microenvironment (TME) is crucial in order to overcome the anti-cancer therapy resistance. In this study, we report the antitumor activity of a newly synthesized ß-carboline derivative "B-9-3." Here, this small molecule showed a promising antitumor activity in vivo along with an enhanced immune response as reflected by a reduction of regulatory T cells and increased CD4+/CD8+ T cells. Further, B-9-3 decreased the number of myofibroblasts not only in the tumor but also in the lung suggesting an anti-metastatic action. The reduction of myofibroblasts was associated with lower expression of epithelial-to-mesenchymal transition markers and a decrease of phosphorylated SMAD2/3 complex indicating the implication of TGF-ß signaling pathway in B-9-3's effect. The blockade of myofibroblasts induction by B-9-3 was also verified in vitro in human fibroblasts treated with TGF-ß. To elucidate the mechanism of B-9-3's action on TGF-ß pathway, first, we investigated the molecular interaction between B-9-3 and TGF-ß receptors using docking method. Data showed a weak interaction of B-9-3 with the ATP-binding pocket of TGFßRI but a strong one with a ternary complex formed of extracellular domains of TGFßRI, TGFßRII, and TGF-ß. In addition, the role of TGFßRI and TGFßRII in B-9-3's activity was explored in vitro. B-9-3 did not decrease any of the two receptors' protein level and only reduced phosphorylated SMAD2/3 suggesting that its effect was more probably due to its interaction with the ternary complex rather than decreasing the expression of TGF-ß receptors or interfering with their ATP-binding domains. B-9-3 is a small active molecule which acts on the TGF-ß signaling pathway and improves the TME to inhibit the proliferation and the metastasis of the tumor with the potential for clinical application.

9.
Oncotarget ; 8(63): 106807-106819, 2017 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29290990

RESUMO

The Eph receptor tyrosine kinase family member EphA2 plays a pivotal role in modulating cytoskeletal dynamics to control cancer cell motility and invasion. EphA2 is frequently upregulated in diverse solid tumors and has emerged as a viable druggable target. We previously reported that extracellular Hsp90 (eHsp90), a known pro-motility and invasive factor, collaborates with EphA2 to regulate tumor invasion in the absence of its cognate ephrin ligand. Here, we aimed to further define the molecular and functional relationship between EphA2 and eHsp90. Ligand dependent ephrin A1 signaling promotes RhoA activation and altered cell morphology to favor transient cell rounding, retraction, and diminished adhesion. Exposure of EphA2-expressing cancer cells to ligand herein revealed a unique role for eHsp90 as an effector of cytoskeletal remodeling. Notably, blockade of eHsp90 via either neutralizing antibodies or administration of cell-impermeable Hsp90-targeted small molecules significantly attenuated ligand dependent cell rounding in diverse tumor types. Although eHsp90 blockade did not appear to influence receptor internalization, downstream signaling events were augmented. In particular, eHsp90 activated a Src-RhoA axis to enhance ligand dependent cell rounding, retraction, and ECM detachment. Moreover, eHsp90 signaling via this axis stimulated activation of the myosin pathway, culminating in formation of an EphA2-myosin complex. Inhibition of either eHsp90 or Src was sufficient to impair ephrin A1 mediated Rho activation, activation of myosin intermediates, and EphA2-myosin complex formation. Collectively, our data support a paradigm whereby eHsp90 and EphA2 exhibit molecular crosstalk and functional cooperation within a ligand dependent context to orchestrate cytoskeletal events controlling cell morphology and attachment.

10.
Eur J Pharmacol ; 766: 37-45, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26415979

RESUMO

The recruitment of monocytes to the active endothelial cells is an early step in the formation of atherosclerotic lesions; therefore, the inhibition of monocyte-endothelial cells interactions may serve as a potential therapeutic strategy for atherosclerosis. Recent studies suggest that ß-elemene can protect against atherosclerosis in vivo and vitro; however, the mechanism underlying the anti-atherosclerotic effect by ß-elemene is not clear yet. In this study, we aimed to investigate the effects of ß-elemene on the monocyte-endothelial cells interactions in the initiation of atherosclerosis in vitro. Our results showed that ß-elemene protects human umbilical vein endothelial cells (HUVECs) from hydrogen peroxide-induced endothelial cells injury in vitro. Besides, this molecule inhibits monocyte adhesion and transendothelial migration across inflamed endothelium through the suppression of the nuclear factor-kappa B-dependent expression of cell adhesion molecules. Further, ß-elemene decreases generation of reactive oxygen species (ROS) and prevents the activation of mitogen-activated protein kinase (MAPK) signaling pathway in HUVECs. In conclusion, this study would provide a new pharmacological evidence of the significance of ß-elemene as a future drug for prevention and treatment of atherosclerosis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Sesquiterpenos/farmacologia , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/metabolismo , Monócitos/fisiologia , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética
12.
Oxid Med Cell Longev ; 2014: 313602, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25404990

RESUMO

Oxidative stress interferes with hepatic lipid metabolism at various levels ranging from benign lipid storage to so-called second hit of inflammation activation. Isoquercitrin (IQ) is widely present flavonoid but its effects on hepatic lipid metabolism remain unknown. We used free fatty acids (FFA) induced lipid overload and oxidative stress model in two types of liver cells and measured cell viability, intracellular lipids, and reactive oxygen species (ROS) within hepatocytes. In addition, Intracellular triglycerides (TG), superoxide dismutase (SOD), and malondialdehyde (MDA) were examined. A novel in vitro model was used to evaluate correlation between lipid lowering and antioxidative activities. Furthermore, 34 major cytokines and corresponding ROS levels were analyzed in FFA/LPS induced coculture model between hepatocytes and Kupffer cells. At molecular level AMPK pathway was elucidated. We showed that IQ attenuated FFA induced lipid overload and ROS within hepatocytes. Further, IQ reversed FFA induced increase in intracellular TG SOD and MDA. It was shown that antioxidative activity of IQ correlates with its lipid lowering potentials. IQ reversed major proinflammatory cytokines and oxidative stress in FFA/LPS induced coculture model. Finally, AMPK pathway was found responsible for metabolic benefits at molecular level. IQ strikingly manifests antioxidative and related lipid lowering activities in hepatocytes.


Assuntos
Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Quercetina/análogos & derivados , Triglicerídeos/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Células de Kupffer/citologia , Lipopolissacarídeos/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Ácido Oleico/administração & dosagem , Palmitatos/administração & dosagem , Quercetina/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo
13.
Eur J Pharmacol ; 724: 219-30, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24380828

RESUMO

Peganum harmala L is an important medicinal plant that has been used from ancient time due to its alkaloids rich of ß-carbolines. Harmane is a naturally occurring ß-carboline extracted from Peganum harmala L, that exhibits a wide range of biological, psychopharmacological, and toxicological actions. The synthesis of novel derivatives with high anti-cancer activity and less side effects is necessary. In the present study, B-9-3-a semi-synthetic compound that is formed of two harmane molecules bound by a butyl group-showed a strong anti-cancer activity against a human lung cancer cell line, a human breast cancer cell line, and a human colorectal carcinoma cell line. B-9-3 anti-proliferative effect followed a similar pattern in the three cell lines. This pattern includes a dose-dependent induction of apoptosis, or necroptosis as confirmed by Hoechst staining, flow cytometry and western blot analyses, and the inhibition of cancer cells migration that was shown to be dependent on the drug׳s concentration as well. Moreover, B-9-3 inhibited tube formation in human umbilical vascular endothelial cell line (HUVEC), which indicates an anti-angiogenesis activity in vitro. In summary, B-9-3, a semi-synthetic derivative of ß-carboline, has an anti-proliferative effect against tumor cells via induction of apoptosis and inhibition of cell migration.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Carbolinas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Peganum
14.
Mol Med Rep ; 5(3): 852-8, 2012 03.
Artigo em Inglês | MEDLINE | ID: mdl-22200881

RESUMO

Reactive oxygen species (ROS) are involved in myocardial injury. ROS are known to inactivate lipid phosphatase and tension homolog on chromosome 10 (PTEN), an enzyme that increases apoptosis in neonatal cardiomyocytes. BpV(pic) and bpV(phen), two bisperoxovanadium molecules and PTEN inhibitors, may be involved in limiting myocardial infarction. To compare the protective effects of bpV(pic) and bpV(phen) on ROS-induced cardiomyocyte injury and their possible mechanisms, we selected two popular models of hypoxia/reoxygenation (H/R) and H2O2-induced injury in H9c2 cardiomyoblasts to investigate their effects against injury. We found that pre-treatment with bpV(pic) and bpV(phen) increased the viability and protected the morphology of H9c2 cells under the conditions of H/R and H2O2 by inhibiting LDH release, apoptosis and caspases 3/8/9 activities. However, their respective inhibitory abilities in the two models were different, suggesting that the quantity of ROS from the two models might be different. However, the conflict between ROS and PTEN may affect the action of bpV(pic) and bpV(phen). Taken together, the results demonstrate that bpV(pic) and bpV(phen) have inhibitory effects on oxidative stress-induced cardiomyocyte injury that may be partially modulated by the action of ROS on PTEN.


Assuntos
Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Peróxido de Hidrogênio/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Fenantrolinas/farmacologia , Compostos de Vanádio/farmacologia , Animais , Caspases/metabolismo , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , L-Lactato Desidrogenase/metabolismo , Compostos Organometálicos/química , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/metabolismo , Fenantrolinas/química , Ratos , Compostos de Vanádio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA