Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Stem Cell Res Ther ; 19(5): 662-668, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37055904

RESUMO

Diabetes mellitus (DM) is a multifaceted pathological condition, which at present is being considered an epidemic disease keeping the rampant rate of its increase in almost all population groups of the world in consideration. Out of the two types of DM described, T1D is characterized as an autoimmune condition that leads to the destruction of pancreatic ß-cells by macrophages and T-cells, thereby, adversely affecting the production of insulin. On the other hand, T2D, often caused by insulin resistance, is commonly related to unhealthy habits, and therefore, it can be prevented in most cases. In both of the conditions, high levels of proinflammatory cytokines like IL-6, TNF-α, and INF-Æ´, lead to chronic inflammation, and elevated oxidative stress resulting in apoptosis and destruction of tissues. Although several treatments are available to treat the symptoms, the underlying causes are not well addressed. One of the most promising approaches to tackle the ill effects and the primary causes of DM is mesenchymal stem cell (MSC) therapy. The use of MSC therapy, because of the immunomodulatory and regenerative properties recorded in this type of cells in a number of experiments carried out in animal models and clinical trials of the disease, has reported positive outcomes. This review covers the principal mechanisms of action induced during MSC therapy in reference to the described pathophysiological pathways of both T1D and T2D. In addition, how this therapeutic intervention can counteract the ill effects of this condition leading to the promotion of tissue regeneration has been covered.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Diabetes Mellitus Tipo 1/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/terapia
2.
Stem Cells Int ; 2022: 3530520, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958032

RESUMO

Innate lymphoid cells have the capability to communicate with other immune cell types to coordinate the immune system functioning during homeostasis and inflammation. However, these cells behave differently at the functional level, unlike T cells, these cells do not need antigen receptors for activation because they are activated by the interaction of their receptor ligation. In hematopoietic stem cell transplantation (HSCT), T cells and NK cells have been extensively studied but very few studies are available on ILCs. In this review, an attempt has been made to provide current information related to NK and ILCs cell-based stem cell therapies and role of the stem cells in the regulation of ILCs as well. Also, the latest information on the differentiation of NK cells and ILCs from CD34+ hematopoietic stem cells is covered in the article.

3.
Stem Cell Rev Rep ; 17(1): 144-152, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32740890

RESUMO

An unfortunate emergence of a new virus SARS-CoV-2, causing a disease known as COVID-19, has spread all around the globe and has caused a pandemic. It primarily affects the respiratory tract and lungs in some cases causing severe organ damage and pneumonia due to overwhelming immune responses. Clinical reports show that the most commons symptoms are fever, dry cough, and shortness of breath, along with several other symptoms. It is thought that an immense cytokine dysregulation in COVID-19 patients is caused following the virus infection. Notably, if patients present with pre-existing specific comorbidities like diabetes or high blood pressure, rates of COVID-19 induced complications and deaths are escalated. Mesenchymal stem cell (MSC) therapy has been shown to alleviate pneumonia and acute respiratory syndrome (ARDS) symptoms, through their immunomodulatory activities in COVID-19 patients. Although more research studies and clinical trial results are needed to elucidate the exact mechanism by which MSCs provide relief to COVID-19 infected patients. Results from clinical trials are encouraging as patients treated with MSCs, regain lung functions and have restored levels of cytokines and trophic factors underscoring the fact that stem cell therapy can be, at least, a complementary therapy to alleviate sufferings in COVID-19 patients. This review discusses the possible therapeutic uses of MSCs for treating COVID-19. Graphical Abstract.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19/terapia , Terapia Baseada em Transplante de Células e Tecidos , Transplante de Células-Tronco Mesenquimais , COVID-19/epidemiologia , COVID-19/virologia , Humanos , Pandemias , SARS-CoV-2/patogenicidade
4.
World J Stem Cells ; 12(10): 1050-1066, 2020 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-33178391

RESUMO

Inflammatory bowel disease (IBD), consisting primarily of ulcerative colitis and Crohn's disease, is a group of debilitating auto-immune disorders, which also increases the risk of colitis-associated cancer. However, due to the chronic nature of the disease and inconsistent treatment outcomes of current anti-IBD drugs (e.g., approximately 30% non-responders to anti-TNFα agents), and related serious side effects, about half of all IBD patients (in millions) turn to alternative treatment options. In this regard, mucosal healing is gaining acceptance as a measure of disease activity in IBD patients as recent studies have correlated the success of mucosal healing with improved prognosis. However, despite the increasing clinical realization of the significance of the concept of mucosal healing, its regulation and means of therapeutic targeting remain largely unclear. Here, stem-cell therapy, which uses hematopoietic stem cells or mesenchymal stem cells, remains a promising option. Stem cells are the pluripotent cells with ability to differentiate into the epithelial and/or immune-modulatory cells. The over-reaching concept is that the stem cells can migrate to the damaged areas of the intestine to provide curative help in the mucosal healing process. Moreover, by differentiating into the mature intestinal epithelial cells, the stem cells also help in restoring the barrier integrity of the intestinal lining and hence prevent the immunomodulatory induction, the root cause of the IBD. In this article, we elaborate upon the current status of the clinical management of IBD and potential role of the stem cell therapy in improving IBD therapy and patient's quality of life.

5.
Curr Pharm Des ; 26(37): 4815-4821, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32744962

RESUMO

BACKGROUND: Normal skin pigmentation pattern is an extremely important component of the appearance of a person, as it can be a significant factor in the social context of any person. A condition known as vitiligo is caused by the death of melanocytes leading to pigmentation loss in the skin. This affects all races across the globe and sometimes leads to social avoidance as in some communities, it is stigmatized. Although there are different pathobiological processes suspected because of the different underlying causes of vitiligo, autoimmunity and oxidative stress are suspected to be the most probable ones. OBJECTIVE: In this review, we present an overview of the underlying mechanisms causing and developing the disease. Also, some of the most successful treatments along with the clinical applications of Mesenchymal Stem Cells (MSCs) as a comprehensive approach for treating this condition will be covered. RESULTS: Autoreactive CD8+ T-cells are the primary suspect considered to be responsible for the destruction of melanocytes. Therefore, topical use of autoimmune inhibitors including those derived from MSCs, thanks to their immune-modulatory properties, have been reported to be successful in the promotion of repigmentation. MSCs can suppress the proliferation of CD8+T via the NKG2D pathway while inducing T-cell apoptosis. The use of pharmacological agents for reducing cellular oxidative stress with the help of topical application of antioxidants and growth factors also have been in use. Intravenous administration of MSCs has been shown to regulate the level of reactive oxidative species (ROS) in a mice model. Growth factors derived from platelet-rich-plasma (PRP) or from MSCs caused rapid tissue regeneration. CONCLUSIONS: Finally, MSC therapy also has been shown to stimulate the mobilization of healthy melanocytes, leading to successful repigmentation of skin lesions in vitiligo patients.


Assuntos
Células-Tronco Mesenquimais , Vitiligo , Animais , Humanos , Melanócitos , Camundongos , Pele , Transplante de Células-Tronco , Vitiligo/tratamento farmacológico
6.
J Thorac Oncol ; 15(9): 1522-1534, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32599072

RESUMO

OBJECTIVES: SCLC represents 15% of all lung cancer diagnoses in the United States and has a particularly poor prognosis. We hypothesized that kinases regulating SCLC survival pathways represent therapeutically targetable vulnerabilities whose inhibition may improve SCLC outcome. METHODS: A short-hairpin RNA (shRNA) library targeting all human kinases was introduced in seven chemonaive patient-derived xenografts (PDX) and the cells were cultured in vitro and in vivo. On harvest, lost or depleted shRNAs were considered as regulating-cell survival pathways and deemed essential kinases. RESULTS: Unsupervised hierarchical cluster analysis of recovered shRNAs separated the PDXs into two clusters, suggesting kinase-based heterogeneity among the SCLC PDXs. A total of 23 kinases were identified as essential in two or more PDXs, with mechanistic Target of Rapamycin (mTOR) a candidate essential kinase in four. mTOR phosphorylation status correlated with PDX sensitivity to mTOR kinase inhibition, and mTOR inhibition sensitized the PDX to cisplatin and etoposide. In the PDX in which mTOR was defined as essential, mTOR inhibition caused a 43% decrease in tumor volume at 21 days (p < 0.01). Combining mTOR inhibition with cisplatin and etoposide decreased PDX tumor volume 96% compared with cisplatin and etoposide alone at 70 days (p < 0.002). Chemoresistance did not develop with the combination of mTOR inhibition and cisplatin and etoposide in mTOR-essential PDX over 105 days. The prevalence of phospho-mTOR-Ser-2448 in a tissue microarray of chemonaive SCLC was 27%, thus, identifying an important SCLC subtype that might benefit from the addition of mTOR inhibition to standard chemotherapy. CONCLUSIONS: These studies reveal that kinases can define SCLC subgroups, can identify therapeutic vulnerabilities, and can potentially be used to optimize therapeutic approaches. Significance We used functional genomics to identify kinases regulating SCLC survival. mTOR was identified as essential in a subset of PDXs. mTOR inhibition decreased PDX growth, sensitized PDX to cisplatin and etoposide, and prevented chemoresistance.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Cisplatino/farmacologia , Etoposídeo/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Sirolimo , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/genética , Serina-Treonina Quinases TOR , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biochem Biophys Res Commun ; 513(4): 794-799, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31000198

RESUMO

A clear understanding of the mechanisms that regulate the alveolar epithelium's barrier is critical to develop new therapeutic strategies to mitigate lung injury. The HER2/HER3 receptor tyrosine kinase complex plays a central role in maintaining the alveolar-capillary barrier. This receptor complex is activated by its ligand, neuregulin-1 (NRG-1). Interleukin-6 (IL-6) is also known to induce HER2 signaling through HER2 transphosphorylation by the IL-6 receptor (IL-6R) complex (1). Due to this interaction, we hypothesized that NRG-1 and IL-6 cooperatively interacted to activate the HER2/HER3 complex. Studies were performed in cultured pulmonary epithelial cells measuring the HER2/IL-6/IL-6R/GP130 interaction and receptor activation by western blotting and confocal microscopy, IL-6 production by ELISA, and IL-6 inhibition using specific antibodies, small molecule inhibitors and shRNA. We found that IL-6 was required for NRG-1 induced activation of HER2 in pulmonary epithelial cells. IL-6 inhibition led to a decrease in NRG-1 induced HER2 activation. The IL-6R and GP130, a subunit of the IL-6R complex, were physically associated with HER2 and were required for NRG-1 induced HER2 activation. Inhibition of GP130, the ß-subunit of the IL-6 receptor decreased NRG-1 induced HER2 activation lower than control by 38% Finally, HER2 activation increased IL-6 secretion more than two-fold over resting cells (526 ± 131 vs 231 ± 39.7 pg/ml), and inhibition of HER2 gene expression decreased basal IL-6 secretion over 80% (89 + 4.6 vs 1.3 + 0.8 pg/ml). These findings identify a requirement for IL-6 and the IL-6R complex to allow NRG-1 mediated HER2 activation, and a HER2 driven IL-6 production feedback loop.


Assuntos
Epitélio/metabolismo , Interleucina-6/metabolismo , Pulmão/metabolismo , Neuregulina-1/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Humanos , Fosforilação , Ligação Proteica , Receptores de Interleucina-6/metabolismo
8.
Am J Respir Cell Mol Biol ; 61(4): 481-491, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30917006

RESUMO

The discovery of mutant tyrosine kinases as oncogenic drivers of lung adenocarcinomas has changed the basic understanding of lung cancer development and therapy. Yet, expressed kinases (kinome) in lung cancer progenitor cells, as well as whether kinase expression and the overall kinome changes or is reprogrammed upon transformation, is incompletely understood. We hypothesized that the kinome differs between lung cancer progenitor cells, alveolar type II cells (ATII), and basal cells (BC) and that their respective kinomes undergo distinct lineage-specific reprogramming to adenocarcinomas and squamous cell carcinomas upon transformation. We performed RNA sequencing on freshly isolated human ATII, BC, and lung cancer cell lines to define the kinome in nontransformed cells and transformed cells. Our studies identified a unique kinome for ATII and BC and changes in their kinome upon transformation to their respective carcinomas.


Assuntos
Células-Tronco Adultas/enzimologia , Células Epiteliais Alveolares/enzimologia , Transformação Celular Neoplásica , Neoplasias Pulmonares/enzimologia , Pulmão/enzimologia , Proteínas de Neoplasias/análise , Proteínas Tirosina Quinases/análise , Adenocarcinoma/enzimologia , Adenocarcinoma/genética , Animais , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/genética , Linhagem da Célula , Células Cultivadas , Indução Enzimática , Humanos , Pulmão/citologia , Neoplasias Pulmonares/genética , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/enzimologia , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , RNA Mensageiro/análise , RNA Neoplásico/análise , Transcriptoma
9.
PLoS One ; 13(4): e0196256, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29672593

RESUMO

Alveolar epithelial cells are critical to the pathogenesis of pulmonary inflammation and fibrosis, which are associated with overexpression of type 2 cytokine IL-13. IL-13 is known to induce the production of profibrotic (e.g., periostin) and pro-inflammatory (e.g., eotaxin-3) mediators in human airway epithelial cells, but it remains unclear if human primary alveolar epithelial cells increase periostin and eotaxin expression following IL-13 stimulation. The goals of this study are to determine if alveolar epithelial cells increase periostin and eotaxin expression upon IL-13 stimulation, and if alveolar and airway epithelial cells from the same subjects have similar responses to IL-13. Paired alveolar and airway epithelial cells were isolated from donors without any lung disease, and cultured under submerged or air-liquid interface conditions with or without IL-13. Up-regulation of periostin protein and mRNA was observed in IL-13-stimulated alveolar epithelial cells, which was comparable to that in IL-13-stimulated paired airway epithelial cells. IL-13 also increased eotaxin-3 expression in alveolar epithelial cells, but the level of eotaxin mRNA was lower in alveolar epithelial cells than in airway epithelial cells. Our findings demonstrate that human alveolar epithelial cells are able to produce periostin and eotaxin in responses to IL-13 stimulation. This study suggests the need to further determine the contribution of alveolar epithelial cell-derived mediators to pulmonary fibrosis.


Assuntos
Células Epiteliais Alveolares , Moléculas de Adesão Celular/genética , Quimiocina CCL11/genética , Células Epiteliais , Interleucina-13/farmacologia , Mucosa Respiratória , Adolescente , Adulto , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Quimiocina CCL11/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Mol Neurobiol ; 54(1): 238-254, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26738854

RESUMO

Diabetes is a systemic disease mainly characterized by chronic hyperglycemia and with extensive and long-lasting spiteful complications in central nervous systems (CNS). Astrocytes play an important role in the defense mechanism of CNS, with great ability of withstanding accumulation of toxic substances. Apart from functional disorders, hyperglycemia leads to slow progressive structural abnormalities in the CNS through oxidative stress pathways. However, the molecular mechanism by which neurons die under oxidative stress induced by high glucose (HG) remains largely unclear. Here, we report that HG-induced inflammation and neurodegeneration in brain tissues, brain astrocytes (C6), and pheochromocytoma (PC-12) cells are cultured in HG conditions. Our results show that the increases in phosphorylation of Akt and ERK1/2MAPK are associated with increased accumulations of reactive oxygen species (ROS) in neuronal cells, which simultaneously enhanced phosphorylations of tuberous sclerosis complex-2 (TSC-2) and mammalian target of rapamycin (mTOR) in the diabetic brain and in HG-exposed neuronal cells. Pharmacologic inhibition of Akt or ERK1/2 or siRNA-mediated gene silencing of TSC-2 suppressed the strong downregulation of TSC-2-mTOR activation. Findings of this study also demonstrate that HG resulted in phosphorylation of NF-κB, coinciding with the increased production of inflammatory mediators and activation of neurodegenerative markers. Pretreatment of cells with antioxidants, phosphoinositide3-kinase (PI3-K)/Akt, and ERK1/2 inhibitors significantly reduced HG-induced TSC-2 phosphorylation and restored NF-κB protein expression leading to decreased production of inflammatory mediators and neurodegenerative markers. These results illustrate that ROS functions as a key signaling component in the regulatory pathway induced by elevated glucose in neuronal cell activation leading to inflammation and neurodegeneration.


Assuntos
Hiperglicemia/metabolismo , Mediadores da Inflamação/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Antioxidantes/farmacologia , Células Cultivadas , Hiperglicemia/complicações , Inflamação/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Masculino , Doenças Neurodegenerativas/etiologia , Neurônios/efeitos dos fármacos , Nitrosação/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/antagonistas & inibidores
11.
Am J Respir Cell Mol Biol ; 54(6): 853-64, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26600084

RESUMO

The airway epithelium constitutes a protective barrier against inhaled insults, such as viruses, bacteria, and toxic fumes, including cigarette smoke (CS). Maintenance of bronchial epithelial integrity is central for airway health, and defective epithelial barrier function contributes to the pathogenesis of CS-mediated diseases, such as chronic obstructive pulmonary disease. Although CS has been shown to increase epithelial permeability, current understanding of the mechanisms involved in CS-induced epithelial barrier disruption remains incomplete. We have previously identified that the receptor tyrosine kinase human epidermal receptor (HER) 2 growth factor is activated by the ligand neuregulin-1 and increases epithelial permeability in models of inflammatory acute lung injury. We hypothesized that CS activates HER2 and that CS-mediated changes in barrier function would be HER2 dependent in airway epithelial cells. We determined that HER2 was activated in whole lung, as well as isolated epithelial cells, from smokers, and that acute CS exposure resulted in HER2 activation in cultured bronchial epithelial cells. Mechanistic studies determined that CS-mediated HER2 activation is independent of neuregulin-1 but required upstream activation of the epidermal growth factor receptor. HER2 was required for CS-induced epithelial permeability as knockdown of HER2 blocked increases in permeability after CS. CS caused an increase in IL-6 production by epithelial cells that was dependent on HER2-mediated extracellular signal-regulated kinases (Erk) activation. Finally, blockade of IL-6 attenuated CS-induced epithelial permeability. Our data indicate that CS activates pulmonary epithelial HER2 and that HER2 is a central mediator of CS-induced epithelial barrier dysfunction.


Assuntos
Permeabilidade da Membrana Celular , Células Epiteliais/metabolismo , Receptor ErbB-2/metabolismo , Fumar/efeitos adversos , Animais , Linhagem Celular , Impedância Elétrica , Receptores ErbB/metabolismo , Humanos , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Neuregulina-1/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais
12.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L199-207, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25326580

RESUMO

The receptor tyrosine kinase human epidermal growth factor receptor-2 (HER2) is known to regulate pulmonary epithelial barrier function; however, the mechanisms behind this effect remain unidentified. We hypothesized that HER2 signaling alters the epithelial barrier through an interaction with the adherens junction (AJ) protein ß-catenin, leading to dissolution of the AJ. In quiescent pulmonary epithelial cells, HER2 and ß-catenin colocalized along the lateral intercellular junction. HER2 activation by the ligand neuregulin-1 was associated with tyrosine phosphorylation of ß-catenin, dissociation of ß-catenin from E-cadherin, and decreased E-cadherin-mediated cell adhesion. All effects were blocked with the HER2 inhibitor lapatinib. ß-Catenin knockdown using shRNA significantly attenuated neuregulin-1-induced decreases in pulmonary epithelial resistance in vitro. Our data indicate that HER2 interacts with ß-catenin, leading to dissolution of the AJ, decreased cell-cell adhesion, and disruption of the pulmonary epithelial barrier.


Assuntos
Junções Aderentes/metabolismo , Adesão Celular/fisiologia , Receptor ErbB-2/metabolismo , Mucosa Respiratória/metabolismo , beta Catenina/metabolismo , Linhagem Celular , Impedância Elétrica , Ativação Enzimática , Humanos , Lapatinib , Pulmão/fisiologia , Neuregulina-1/metabolismo , Permeabilidade , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptor ErbB-2/antagonistas & inibidores , Transdução de Sinais , Junções Íntimas/metabolismo , beta Catenina/genética
13.
Bioinformatics ; 30(17): 2393-8, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24812339

RESUMO

MOTIVATION: Non-small-cell lung cancer (NSCLC) is the leading cause of cancer death in the United States. Targeted tyrosine kinase inhibitors (TKIs) directed against the epidermal growth factor receptor (EGFR) have been widely and successfully used in treating NSCLC patients with activating EGFR mutations. Unfortunately, the duration of response is short-lived, and all patients eventually relapse by acquiring resistance mechanisms. RESULT: We performed an integrative systems biology approach to determine essential kinases that drive EGFR-TKI resistance in cancer cell lines. We used a series of bioinformatics methods to analyze and integrate the functional genetics screen and RNA-seq data to identify a set of kinases that are critical in survival and proliferation in these TKI-resistant lines. By connecting the essential kinases to compounds using a novel kinase connectivity map (K-Map), we identified and validated bosutinib as an effective compound that could inhibit proliferation and induce apoptosis in TKI-resistant lines. A rational combination of bosutinib and gefitinib showed additive and synergistic effects in cancer cell lines resistant to EGFR TKI alone. CONCLUSIONS: We have demonstrated a bioinformatics-driven discovery roadmap for drug repurposing and development in overcoming resistance in EGFR-mutant NSCLC, which could be generalized to other cancer types in the era of personalized medicine. AVAILABILITY AND IMPLEMENTATION: K-Map can be accessible at: http://tanlab.ucdenver.edu/kMap. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Inibidores de Proteínas Quinases/farmacologia , Compostos de Anilina/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Biologia Computacional , Resistencia a Medicamentos Antineoplásicos/genética , Gefitinibe , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Quinases/genética , Quinazolinas/farmacologia , Quinolinas/farmacologia , Análise de Sequência de RNA
14.
Eur Respir J ; 41(2): 396-401, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22599357

RESUMO

Shedding of neuregulin (NRG)-1 from the pulmonary epithelium leads to activation of the epithelial human epidermal growth factor receptor (HER)2 receptor, increased pulmonary epithelial permeability and acute lung injury (ALI). We sought to determine whether NRG-1 was detectable and elevated in bronchoalveolar lavage (BAL) and plasma from patients with ALI compared with controls and to determine whether a correlation exists between NRG-1 and inflammation and outcome in ALI. Matched BAL and plasma samples were obtained from 23 ALI patients requiring intubation and mechanical ventilation. Control patients (n=5) included healthy volunteers. NRG-1 and indices of inflammation were measured in BAL and plasma via ELISA. The mean±sd BAL NRG-1 concentration in ALI patients was 187.0±21.35 pg·mL(-1) compared with 85.50±9.2 pg·mL(-1) in controls (p=0.001). Increased BAL NRG-1 was associated with markers of inflammation, and inversely correlated with ventilator-free days (VFDs; r= -0.51, p=0.015). Plasma NRG-1 was elevated in ALI patients compared with controls (611.7±354.2 versus 25.17±19.33 pg·mL(-1), p<0.001) and inversely correlated with VFDs (r= -0.51, p=0.04). These results confirm shedding of NRG-1 in ALI and suggest that the NRG-1-HER2 pathway is active in patients with ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Líquido da Lavagem Broncoalveolar/imunologia , Neuregulina-1/metabolismo , Adulto , Biomarcadores/metabolismo , Estudos de Casos e Controles , Epitélio/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptor ErbB-2/metabolismo , Respiração Artificial
15.
J Thorac Oncol ; 8(12): 1492-501, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24389431

RESUMO

INTRODUCTION: KRAS mutations are poor prognostic markers for patients with non-small-cell lung cancer (NSCLC). RALA and RALB GTPases lie downstream of RAS and are implicated in RAS-mediated tumorigenesis. However, their biological or prognostic role in the context of KRAS mutation in NSCLC is unclear. METHODS: Using expression analysis of human tumors and a panel of cell lines coupled with functional in vivo and in vitro experiments, we evaluated the prognostic and functional importance of RAL in NSCLC and their relationship to KRAS expression and mutation. RESULTS: Immunohistochemical (N = 189) and transcriptomic (N = 337) analyses of NSCLC patients revealed high RALA and RALB expression was associated with poor survival. In a panel of 14 human NSCLC cell lines, RALA and RALB had higher expression in KRAS mutant cell lines whereas RALA but not RALB activity was higher in KRAS mutant cell lines. Depletion of RAL paralogs identified cell lines that are dependent on RAL expression for proliferation and anchorage independent growth. Overall, growth of NSCLC cell lines that carry a glycine to cystine KRAS mutation were more sensitive to RAL depletion than those with wild-type KRAS. The use of gene expression and outcome data from 337 human tumors in RAL-KRAS interaction analysis revealed that KRAS and RAL paralog expression jointly impact patient prognosis. CONCLUSION: RAL GTPase expression carries important additional prognostic information to KRAS status in NSCLC patients. Simultaneously targeting RAL may provide a novel therapeutic approach in NSCLC patients harboring glycine to cystine KRAS mutations.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo , Animais , Apoptose , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Feminino , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Camundongos , Camundongos Nus , Estadiamento de Neoplasias , Prognóstico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteínas ral de Ligação ao GTP/genética , Proteínas ras/genética
16.
J Biol Chem ; 286(12): 10660-70, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21247898

RESUMO

The mechanisms behind the loss of epithelial barrier function leading to alveolar flooding in acute lung injury (ALI) are incompletely understood. We hypothesized that the tyrosine kinase receptor human epidermal growth factor receptor-2 (HER2) would be activated in an inflammatory setting and participate in ALI. Interleukin-1ß (IL-1ß) exposure resulted in HER2 activation in human epithelial cells and markedly increased conductance across a monolayer of airway epithelial cells. Upon HER2 blockade, conductance changes were significantly decreased. Mechanistic studies revealed that HER2 trans-activation by IL-1ß required a disintegrin and metalloprotease 17 (ADAM17)-dependent shedding of the ligand neuregulin-1 (NRG-1). In murine models of ALI, NRG-1-HER2 signaling was activated, and ADAM17 blockade resulted in decreased NRG-1 shedding, HER2 activation, and lung injury in vivo. Finally, NRG-1 was detectable and elevated in pulmonary edema fluid from patients with ALI. These results suggest that the ADAM17-NRG-1-HER2 axis modulates the alveolar epithelial barrier and contributes to the pathophysiology of ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Neuregulina-1/metabolismo , Receptor ErbB-2/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Barreira Alveolocapilar/metabolismo , Barreira Alveolocapilar/patologia , Linhagem Celular , Modelos Animais de Doenças , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Neuregulina-1/genética , Permeabilidade , Receptor ErbB-2/genética , Mucosa Respiratória/patologia , Ativação Transcricional/genética
17.
J Biol Chem ; 283(16): 10461-9, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18250161

RESUMO

In wound healing, myofibroblast transdifferentiation (MFT) is a metaplastic change in phenotype producing profibrotic effector cells that secrete and remodel the extracellular matrix. Unlike pathways that induce MFT, the molecular mechanisms that negatively regulate MFT are poorly understood. Here, we report that AMP-activated protein kinase (AMPK) blocks MFT in response to transforming growth factor-beta (TGFbeta). Pharmacological activation of AMPK inhibited TGFbeta-induced secretion of extracellular matrix proteins collagen types I and IV and fibronectin. AMPK activation also prevented induction of the myofibroblast phenotype markers alpha-smooth muscle actin and the ED-A fibronectin splice variant. AMPK activators did not prevent MFT in cells transduced with an adenovirus expressing dominant negative, kinase-dead AMPKalpha2. Moreover, AMPK activators did not inhibit MFT induction in AMPK(alpha1,2)(-/-) fibroblasts, demonstrating a requirement for AMPK(alpha) expression. Adenoviral transduction of constitutively active AMPK(alpha2) was sufficient to prevent TGFbeta-induced collagen I, alpha-smooth muscle actin, and ED-A fibronectin. AMPK did not reduce TGFbeta-stimulated Smad3 COOH-terminal phosphorylation and nuclear translocation, which are necessary for MFT. However, AMPK activation inhibited TGFbeta-induced transcription driven by Smad3-binding cis-elements. Consistent with a role for AMPK in transcriptional regulation, nuclear translocation of AMPKalpha2 correlated with the appearance of active AMPKalpha in the nucleus. Collectively, these results demonstrate that AMPK inhibits TGFbeta-induced transcription downstream of Smad3 COOH-terminal phosphorylation and nuclear translocation. Furthermore, activation of AMPK is sufficient to negatively regulate MFT in vitro.


Assuntos
Fibroblastos/metabolismo , Complexos Multienzimáticos/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteína Smad3/fisiologia , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases Ativadas por AMP , Transporte Ativo do Núcleo Celular , Adenoviridae/metabolismo , Núcleo Celular/metabolismo , Transdiferenciação Celular , Colágeno/metabolismo , Ativação Enzimática , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Modelos Biológicos , Fenótipo
18.
Arterioscler Thromb Vasc Biol ; 28(3): 541-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18096831

RESUMO

OBJECTIVE: High circulating free fatty acids, commonly associated with obesity and insulin resistance, impair structure and function of the microvasculature. However, the mechanisms by which fatty acids cause microvascular remodeling are unclear. Using the mesangial cell model of microvascular pericytes, we demonstrate that the monounsaturated free fatty acid oleate induces a myofibroblast phenotype, an important cell fate transition in fibrotic remodeling of the extracellular matrix. MATERIALS AND RESULTS: Oleate induced a time- and dose-dependent increase in secretion of collagen I and fibronectin. Oleate also induced the myofibroblast phenotype markers alpha smooth muscle actin and ED-A fibronectin, and the magnitude of marker protein expression was similar to that for transforming growth factor (TGF)-beta. Oleate raised TGF-beta secretion 2.2-fold, and processing of latent to bioactive TGF-beta was also elevated. Oleate rapidly stimulated extracellular signal-regulated kinase1/2, and a pharmacological MEK inhibitor blocked TGF-beta secretion and conversion to the myofibroblast phenotype. A neutralizing TGF-beta antibody and a TGF-beta receptor kinase inhibitor blocked oleate-induced collagen I, alpha smooth muscle actin, and ED-A fibronectin, suggesting that oleate-stimulated TGF-beta was necessary for inducing myofibroblasts. CONCLUSIONS: Collectively, these results demonstrate that oleate can induce a myofibroblast phenotype in mesangial cells, which suggests a mechanism whereby elevated free fatty acids might promote microvascular remodeling in vivo.


Assuntos
Fibronectinas/metabolismo , Células Mesangiais/efeitos dos fármacos , Ácido Oleico/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Ensaio de Imunoadsorção Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Fibronectinas/efeitos dos fármacos , Imunofluorescência , Humanos , Células Mesangiais/fisiologia , Fenótipo , Sensibilidade e Especificidade , Transdução de Sinais , Fator de Crescimento Transformador beta/efeitos dos fármacos
19.
Biochem Biophys Res Commun ; 361(4): 953-9, 2007 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-17692287

RESUMO

The primary objective of this work is to determine the repairing potential of murine embryonic stem cells (ES) in murine model of Crohn's disease (CD). Colitis, induced in IL10-/- KO mice using piroxicam, was associated with the increased levels of IL-12. Enhanced yellow fluorescent protein (EYFP) marked murine ES cells (R1/129) and control non-fluorescent ES cells were subjected to in vitro differentiation into intestinal epithelial cells. IL 10-/- KO mice were injected with pre-differentiated ES-YFP cells and sacrificed after 2 and 3 months. Histopathological analysis of intestines demonstrated a progressive improvement in colitis (from grade-4 to grade-1 and -0) and decreased levels of IL-12 cytokine following transplantation. Fluorescent and confocal microscopy demonstrated presence of ES-EYFP cells in the colon, small intestine, liver, and thymus tissues but none in the spleen and bone marrow. The EYFP signal was not detected in sham (non-transplanted mice with induced colitis) and control IL10-/- KO mice. Engraftment, detected at 3 months post-transplant, correlated with markedly improved grading in colon histology (grade-1 or -0) and weight gain, as well as with decreased rectal prolapses. In vitro pre-differentiated ES cells migrated and homed exclusively into the colon, small intestine, and the liver, engrafted for long term, reduced inflammation and tissue damage, and restored immune balance. These findings suggest that pre-differentiated ES cells may become alternative source of stem cell therapy for CD with dual functions i.e. regenerating damaged epithelium and restoring immune imbalance occurring in this disease.


Assuntos
Doença de Crohn/terapia , Células-Tronco Embrionárias/transplante , Interleucina-10/genética , Animais , Colite/induzido quimicamente , Colite/imunologia , Colite/terapia , Colo/patologia , Doença de Crohn/induzido quimicamente , Doença de Crohn/imunologia , Interleucina-10/metabolismo , Camundongos , Camundongos Knockout , Piroxicam
20.
Stem Cells ; 25(6): 1456-61, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17379761

RESUMO

The role of thrombopoietin (TPO) in adult hematopoiesis is well-established. A recent report suggests that TPO and vascular endothelial growth factor (VEGF) play a role in promoting formation of early erythropoietic progenitors in a nonhuman primate embryonic stem cell (ES) model. No such report exists for human ES cells as yet. Because TPO may become an important factor promoting human ES cell-derived hematopoiesis, we sought to investigate whether TPO in combination with VEGF can enhance human ES-derived hematopoiesis in an EB-derived culture system. The emphasis of this work was to demonstrate the molecular mechanisms involved in this process, specifically the role of c-mpl and its ligand TPO. Human ES cells were cultured to the EB state, and EB-derived secondary cultures supporting hematopoietic differentiation were established: condition 1, control (stem cell factor [SCF] and Flt3 ligand [Flt3L]); condition 2, SCF, Flt3L, and TPO; and condition 3, SCF, Flt3L, TPO, and VEGF. Cells were harvested daily, starting at day 2 and continuing until day 8, for reverse transcription-polymerase chain reaction and Western blot. There was no evidence of expression of c-mpl and VEGF receptor on the gene or protein level until day 8, when the formation of well-established hematopoietic colonies began. This correlated with the formation of CD34+/CD31- negative progenitors, mostly found in blast-forming units-erythroid-like colonies. We concluded that TPO and VEGF play an important synergistic role in the formation of early ES-derived hematopoietic progenitors that occurs through the c-mpl and VEGF receptors. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Antígenos CD34/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Trombopoetina/farmacologia , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Humanos , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Trombopoetina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...