Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Carcinogenesis ; 41(5): 561-570, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31369062

RESUMO

The lack of tools for early detection of pancreatic ductal adenocarcinoma (PDAC) is directly correlated with the abysmal survival rates in patients. In addition to several potential detection tools under active investigation, we tested the gut microbiome and its metabolic complement as one of the earliest detection tools that could be useful in patients at high risk for PDAC. We used a combination of 16s rRNA pyrosequencing and whole-genome sequencing of gut fecal microbiota in a genetically engineered PDAC murine model (KRASG12DTP53R172HPdxCre or KPC). Metabolic reconstruction of microbiome was done using the HUMAnN2 pipeline. Serum polyamine levels were measured from murine and patient samples using chromogenic assay. Our results showed a Proteobacterial and Firmicutes dominance in gut microbiota in early stages of PDAC development. Upon in silico reconstruction of active metabolic pathways within the altered microbial flora, polyamine and nucleotide biosynthetic pathways were significantly elevated. These metabolic products are known to be actively assimilated by the host and eventually utilized by rapidly dividing cells for proliferation validating their importance in the context of tumorigenesis. In KPC mice, as well as PDAC patients, we show significantly elevated serum polyamine concentrations. Therefore, at the early stages of tumorigenesis, there is a strong correlation between microbial changes and release of metabolites that foster host tumorigenesis, thereby fulfilling the 'vicious cycle hypothesis' of the role of microbiome in health and disease states. Our results provide a potential, precise, noninvasive tool for early detection of PDAC, which may result in improved outcomes.


Assuntos
Carcinoma Ductal Pancreático/diagnóstico , Disbiose/complicações , Detecção Precoce de Câncer , Microbioma Gastrointestinal , Neoplasias Pancreáticas/diagnóstico , Poliaminas/metabolismo , Animais , Carcinogênese , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/microbiologia , Carcinoma Ductal Pancreático/patologia , Disbiose/microbiologia , Fezes/microbiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/microbiologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína Supressora de Tumor p53/genética
2.
Gastroenterology ; 156(7): 2073-2084, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30716326

RESUMO

Pancreatic ductal adenocarcinoma is one of the deadliest cancers, and its incidence on the rise. The major challenges in overcoming the poor prognosis with this disease include late detection and the aggressive biology of the disease. Intratumoral heterogeneity; presence of a robust, reactive, and desmoplastic stroma; and the crosstalk between the different tumor components require complete understanding of the pancreatic tumor biology to better understand the therapeutic challenges posed by this disease. In this review, we discuss the processes involved during tumorigenesis encompassing the inherent plasticity of the transformed cells, development of tumor stroma crosstalk, and enrichment of cancer stem cell population during tumorigenesis.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Plasticidade Celular , Transformação Celular Neoplásica/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Biomarcadores Tumorais/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundário , Carcinoma Ductal Pancreático/terapia , Comunicação Celular , Movimento Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Humanos , Mediadores da Inflamação/metabolismo , Mutação , Invasividade Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Fenótipo , Transdução de Sinais , Microambiente Tumoral
3.
Microb Cell Fact ; 19(1): 75, 2020 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32204699

RESUMO

Resistance to therapy is one of the major factors that contribute to dismal survival statistics in pancreatic cancer. While there are many tumor intrinsic and tumor microenvironment driven factors that contribute to therapy resistance, whether pre-existing metabolic diseases like type 2 diabetes (T2D) contribute to this has remained understudied. It is well accepted that hyperglycemia associated with type 2 diabetes changes the gut microbiome. Further, hyperglycemia also enriches for a "stem-like" population within the tumor. In the current study, we observed that in a T2D mouse model, the microbiome changed significantly as the hyperglycemia developed in these animals. Our results further showed that, tumors implanted in the T2D mice responded poorly to gemcitabine/paclitaxel (Gem/Pac) standard of care compared to those in the control group. A metabolomic reconstruction of the WGS of the gut microbiota further revealed that an enrichment of bacterial population involved in drug metabolism in the T2D group. Additionally, we also observed an increase in the CD133+ tumor cells population in the T2D model. These observations indicated that in an animal model for T2D, microbial dysbiosis is associated with increased resistance to chemotherapeutic compounds.


Assuntos
Diabetes Mellitus Tipo 2/microbiologia , Resistencia a Medicamentos Antineoplásicos , Disbiose/microbiologia , Hiperglicemia/microbiologia , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Microbioma Gastrointestinal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/microbiologia , Gencitabina , Neoplasias Pancreáticas
4.
Gastroenterology ; 155(1): 33-37.e6, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29630898

RESUMO

We studied the effects of gut microbiome depletion by oral antibiotics on tumor growth in subcutaneous and liver metastases models of pancreatic cancer, colon cancer, and melanoma. Gut microbiome depletion significantly reduced tumor burden in all the models tested. However, depletion of gut microbiome did not reduce tumor growth in Rag1-knockout mice, which lack mature T and B cells. Flow cytometry analyses demonstrated that gut microbiome depletion led to significant increase in interferon gamma-producing T cells with corresponding decrease in interleukin 17A and interleukin 10-producing T cells. Our results suggest that gut microbiome modulation could emerge as a novel immunotherapeutic strategy.


Assuntos
Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Metástase Neoplásica/imunologia , Neoplasias/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Antibacterianos/farmacologia , Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/efeitos dos fármacos , Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-17/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Melanoma/imunologia , Melanoma/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias de Tecidos Moles/imunologia , Neoplasias de Tecidos Moles/secundário , Linfócitos T/imunologia , Microambiente Tumoral/imunologia
5.
Gastroenterology ; 155(3): 880-891.e8, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29909021

RESUMO

BACKGROUND & AIMS: Immunotherapies are ineffective against pancreatic cancer. We investigated whether the activity of nuclear factor (NF)κB in pancreatic stromal cells contributes to an environment that suppresses antitumor immune response. METHODS: Pancreata of C57BL/6 or Rag1-/- mice were given pancreatic injections of a combination of KrasG12D/+; Trp53 R172H/+; Pdx-1cre (KPC) pancreatic cancer cells and pancreatic stellate cells (PSCs) extracted from C57BL/6 (control) or mice with disruption of the gene encoding the NFκB p50 subunit (Nfkb1 or p50-/- mice). Tumor growth was measured as an endpoint. Other mice were given injections of Lewis lung carcinoma (LLC) lung cancer cells or B16-F10 melanoma cells with control or p50-/- fibroblasts. Cytotoxic T cells were depleted from C57BL/6 mice by administration of antibodies against CD8 (anti-CD8), and growth of tumors from KPC cells, with or without control or p50-/- PSCs, was measured. Some mice were given an inhibitor of CXCL12 (AMD3100) and tumor growth was measured. T-cell migration toward cancer cells was measured using the Boyden chamber assay. RESULTS: C57BL/6 mice coinjected with KPC cells (or LLC or B16-F10 cells) and p50-/- PSCs developed smaller tumors than mice given injections of the cancer cells along with control PSCs. Tumors that formed when KPC cells were injected along with p50-/- PSCs had increased infiltration by activated cytotoxic T cells along with decreased levels of CXCL12, compared with tumors grown from KPC cells injected along with control PSCs. KPC cells, when coinjected with control or p50-/- PSCs, developed the same-size tumors when CD8+ T cells were depleted from C57BL/6 mice or in Rag1-/- mice. The CXCL12 inhibitor slowed tumor growth and increased tumor infiltration by cytotoxic T cells. In vitro expression of p50 by PSCs reduced T-cell migration toward and killing of cancer cells. When cultured with cancer cells, control PSCs expressed 10-fold higher levels of CXCL12 than p50-/- PSCs. The CXCL12 inhibitor increased migration of T cells toward KPC cells in culture. CONCLUSIONS: In studies of mice and cell lines, we found that NFκB activity in PSCs promotes tumor growth by increasing expression of CXCL12, which prevents cytotoxic T cells from infiltrating the tumor and killing cancer cells. Strategies to block CXCL12 in pancreatic tumor cells might increase antitumor immunity.


Assuntos
Quimiocina CXCL12/fisiologia , Linfócitos do Interstício Tumoral/fisiologia , NF-kappa B/fisiologia , Neoplasias Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Linfócitos T Citotóxicos/fisiologia , Animais , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Imunidade Celular , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/imunologia , Células Estreladas do Pâncreas/imunologia , Regulação para Cima
6.
J Transl Med ; 17(1): 301, 2019 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-31484534

RESUMO

Following publication of the original article [1], the authors found an error in Figure 3. The middle panel of Figure 3a was inadvertently duplicated.

7.
J Transl Med ; 17(1): 163, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31109340

RESUMO

BACKGROUND: There is an urgent need for novel and effective treatment options for acute myeloid leukemia (AML). Triptolide, a diterpenoid tri-epoxide compound isolated from the herb Tripterygium wilfordii and its water-soluble pro-drug-Minnelide have shown promising anti-cancer activity. A recent clinical trial for patients with solid tumors confirmed the safety and efficacy at biologically equivalent doses of 0.2 mg/kg/day and lower. METHODS: Cell viability of multiple AML cell lines as well as patient apheresis samples were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) based assay. Apoptosis was evaluated by estimating the amount of cleaved caspase. AML cell line (THP1-Luc) was implanted in immunocompromised mice and treated with indicated doses of Minnelide. Leukemic burden before and after treatment was evaluated by imaging in an In Vivo Imaging System (IVIS). RESULTS: In the current study, we show that Minnelide, at doses below maximum tolerated dose (MTD) demonstrates leukemic clearance of both primary AML blasts and luciferase expressing THP-1 cells in mice. In vitro, multiple primary AML apheresis samples and AML cell lines (THP-1, KG1, Kasumi-1, HL-60) were sensitive to triptolide mediated cell death and apoptosis in low doses. Treatment with triptolide led to a significant decrease in the colony forming ability of AML cell lines as well as in the expression of stem cell markers. Additionally, it resulted in the cell cycle arrest in the G1/S phase with significant downregulation of c-Myc, a major transcriptional regulator mediating cancer cell growth and stemness. CONCLUSION: Our results suggest that Minnelide, with confirmed safety and activity in the clinic, exerts a potent anti-leukemic effect in multiple models of AML at doses easily achievable in patients.


Assuntos
Diterpenos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Organofosfatos/uso terapêutico , Fenantrenos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Diterpenos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Compostos de Epóxi , Humanos , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Organofosfatos/farmacologia , Fenantrenos/farmacologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
8.
IUBMB Life ; 71(2): 152-165, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30466159

RESUMO

It is now well appreciated that the human microbiome plays a significant role in a number of processes in the body, significantly affecting its metabolic, inflammatory, and immune homeostasis. Recent research has revealed that almost every mucosal surface in the human body is associated with a resident commensal microbiome of its own. While the gut microbiome and its role in regulation of host metabolism along with its alteration in a disease state has been well studied, there is a lacuna in understanding the resident microbiota of other mucosal surfaces. Among these, the scientific information on the role of lung microbiota in pulmonary diseases is currently severely limited. Historically, lungs have been considered to be sterile and lung diseases have only been studied in the context of bacterial pathogenesis. Recently however, studies have revealed a resilient microbiome in the upper and lower respiratory tracts and there is increased evidence on its central role in respiratory diseases. Knowledge of lung microbiome and its metabolic fallout (local and systemic) is still in its nascent stages and attracting immense interest in recent times. In this review, we will provide a perspective on lung-associated metabolic disorders defined for lung diseases (e.g., chronic obstructive pulmonary disease, asthma, and respiratory depression due to infection) and correlate it with lung microbial perturbation. Such perturbations may be due to altered biochemical or metabolic stress as well. Finally, we will draw evidence from microbiome and classical microbiology literature to demonstrate how specific lung morbidities associate with specific metabolic characteristics of the disease, and with the role of microbiome in this context. © 2018 IUBMB Life, 71(1):152-165, 2019.


Assuntos
Asma/metabolismo , Fibrose Cística/metabolismo , Neoplasias Pulmonares/metabolismo , Pneumonia Bacteriana/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Actinobacteria/imunologia , Actinobacteria/metabolismo , Actinobacteria/patogenicidade , Asma/imunologia , Asma/microbiologia , Asma/patologia , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Firmicutes/imunologia , Firmicutes/metabolismo , Firmicutes/patogenicidade , Homeostase/imunologia , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/microbiologia , Neoplasias Pulmonares/patologia , Microbiota/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Proteobactérias/imunologia , Proteobactérias/metabolismo , Proteobactérias/patogenicidade , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Doença Pulmonar Obstrutiva Crônica/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia
9.
Gut ; 67(4): 600-602, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28642332

RESUMO

BACKGROUND: Opioids such as morphine are widely used for the management of pain associated with acute pancreatitis. Interestingly, opioids are also known to affect the immune system and modulate inflammatory pathways in non-pancreatic diseases. However, the impact of morphine on the progression of acute pancreatitis has never been evaluated. In the current study, we evaluated the impact of morphine on the progression and severity of acute pancreatitis. METHODS: Effect of morphine treatment on acute pancreatitis in caerulein, L-arginine and ethanol-palmitoleic acid models was evaluated after induction of the disease. Inflammatory response, gut permeability and bacterial translocation were compared. Experiments were repeated in mu (µ) opioid receptor knockout mice (MORKO) and in wild-type mice in the presence of opioid receptor antagonist naltrexone to evaluate the role of µ-opioid receptors in morphine's effect on acute pancreatitis. Effect of morphine treatment on pathways activated during pancreatic regeneration like sonic Hedgehog and activation of embryonic transcription factors like pdx-1 and ptf-1 were measured by immunofluorescence and quantitative PCR. RESULTS: Histological data show that treatment with morphine after induction of acute pancreatitis exacerbates the disease with increased pancreatic neutrophilic infiltration and necrosis in all three models of acute pancreatitis. Morphine also exacerbated acute pancreatitis-induced gut permeabilisation and bacteraemia. These effects were antagonised in the MORKO mice or in the presence of naltrexone suggesting that morphine's effect on severity of acute pancreatitis are mediated through the µ-opioid receptors. Morphine treatment delayed macrophage infiltration, sonic Hedgehog pathway activation and expression of pdx-1 and ptf-1. CONCLUSION: Morphine treatment worsens the severity of acute pancreatitis and delays resolution and regeneration. Considering our results, the safety of morphine for analgesia during acute pancreatitis should be re-evaluated in future human studies.


Assuntos
Analgésicos Opioides/efeitos adversos , Morfina/efeitos adversos , Pâncreas/patologia , Pancreatite/diagnóstico , Doença Aguda , Analgésicos Opioides/administração & dosagem , Animais , Arginina , Ceruletídeo , Modelos Animais de Doenças , Progressão da Doença , Ácidos Graxos Monoinsaturados , Camundongos , Camundongos Knockout , Morfina/administração & dosagem , Pancreatite/induzido quimicamente , Pancreatite/tratamento farmacológico , Índice de Gravidade de Doença , Fatores de Tempo
10.
J Bioenerg Biomembr ; 50(3): 205-211, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29204729

RESUMO

Whether embryonic, hematopoietic or cancer stem cells, this metabolic reprogramming is dependent on the nutrient-status and bioenergetic pathways that is influenced by the micro-environmental niches like hypoxia. Thus, the microenvironment plays a vital role in determining the stem cell fate by inducing metabolic reprogramming. Under the influence of the microenvironment, like hypoxia, the stem cells have increased glucose and glutamine uptake which result in activation of hexosamine biosynthesis pathway (HBP) and increased O-GlcNAc Transferase (OGT). The current review is focused on understanding how HBP, a nutrient-sensing pathway (that leads to increased OGT activity) is instrumental in regulating self-renewal not only in embryonic and hematopoietic stem cells (ESC/HSC) but also in cancer stem cells.


Assuntos
Vias Biossintéticas , Autorrenovação Celular , N-Acetilglucosaminiltransferases/metabolismo , Nutrientes/metabolismo , Animais , Metabolismo Energético , Hexosaminas/biossíntese , Humanos
11.
Prostate ; 77(6): 584-596, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28144973

RESUMO

BACKGROUND: With almost 30,000 deaths per year, prostate cancer is the second-leading cause of cancer-related death in men. Androgen Deprivation Therapy (ADT) has been the corner stone of prostate cancer treatment for decades. However, despite an initial response of prostate cancer to ADT, this eventually fails and the tumors recur, resulting in Castration Resistant Prostate Cancer (CRPC). Triptolide, a diterpene triepoxide, has been tested for its anti-tumor properties in a number of cancers for over a decade. Owing to its poor solubility in aqueous medium, its clinical application had been limited. To circumvent this problem, we have synthesized a water-soluble pro-drug of triptolide, Minnelide, that is currently being evaluated in a Phase 1 clinical trial against gastrointestinal tumors. In the current study, we assessed the therapeutic potential of Minnelide and its active compound triptolide against androgen dependent prostate cancer both in vitro as well as in vivo. METHODS: Cell viability was measured by a MTT based assay after treating prostate cancer cells with multiple doses of triptolide. Apoptotic cell death was measured using a caspase 3/7 activity. Androgen Receptor (AR) promoter-binding activity was evaluated by using luciferase reporter assay. For evaluating the effect in vivo, 22Rv1 cells were implanted subcutaneously in animals, following which, treatment was started with 0.21 mg/kg Minnelide. RESULTS: Our study showed that treatment with triptolide induced apoptotic cell death in CRPC cells. Triptolide treatment inhibited AR transcriptional activity and decreased the expression of AR and its splice variants both at the mRNA and the protein level. Our studies show that triptolide inhibits nuclear translocation of Sp1, resulting in its decreased transcriptional activity leading to downregulation of AR and its splice variants in prostate cancer cells. In vivo, Minnelide (0.21 mg/kg) regressed subcutaneous tumors derived from CRPC 22RV1 at our study endpoint. Our animal studies further confirmed that Minnelide was more efficacious than the standard of care therapies, Docetaxel and Enzalutamide. CONCLUSION: Our study indicates that Minnelide is very effective as a therapeutic option against CRPC at a dose that is currently tolerated by patients in the ongoing clinical trials. Prostate 77: 584-596, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Regulação Neoplásica da Expressão Gênica , Organofosfatos/farmacologia , Fenantrenos/farmacologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Isoformas de Proteínas/biossíntese , Receptores Androgênicos/biossíntese , Animais , Linhagem Celular Tumoral , Diterpenos , Relação Dose-Resposta a Droga , Compostos de Epóxi , Humanos , Masculino , Camundongos , Camundongos Nus , Organofosfatos/uso terapêutico , Fenantrenos/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Distribuição Aleatória , Receptores Androgênicos/genética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Gastroenterology ; 151(4): 747-758.e5, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27519471

RESUMO

BACKGROUND & AIMS: Experimental studies in acute pancreatitis (AP) suggest a strong association of acinar cell injury with cathepsin B-dependent intracellular activation of trypsin. However, the molecular events subsequent to trypsin activation and their role, if any, in cell death is not clear. In this study, we have explored intra-acinar events downstream of trypsin activation that lead to acinar cell death. METHODS: Acinar cells prepared from the pancreas of rats or mice (wild-type, trypsinogen 7, or cathepsin B-deleted) were stimulated with supramaximal cerulein, and the cytosolic activity of cathepsin B and trypsin was evaluated. Permeabilized acini were used to understand the differential role of cytosolic trypsin vs cytosolic cathepsin B in activation of apoptosis. Cell death was evaluated by measuring specific markers for apoptosis and necrosis. RESULTS: Both in vitro and in vivo studies have suggested that during AP cathepsin B leaks into the cytosol from co-localized organelles, through a mechanism dependent on active trypsin. Cytosolic cathepsin B but not trypsin activates the intrinsic pathway of apoptosis through cleavage of bid and activation of bax. Finally, excessive release of cathepsin B into the cytosol can lead to cell death through necrosis. CONCLUSIONS: This report defines the role of trypsin in AP and shows that cytosolic cathepsin B but not trypsin activates cell death pathways. This report also suggests that trypsin is a requisite for AP only because it causes release of cathepsin B into the cytosol.


Assuntos
Células Acinares/enzimologia , Catepsina B/fisiologia , Morte Celular/fisiologia , Citosol/enzimologia , Pancreatite/enzimologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pâncreas/citologia , Pancreatite/patologia , Ratos , Ratos Wistar , Tripsina/fisiologia
13.
Curr Opin Gastroenterol ; 33(5): 404-410, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28682795

RESUMO

PURPOSE OF REVIEW: Pancreatic cancer, despite years of study and some progress, presents with a grim prognosis in almost all cases. In the current review, we have discussed recent studies that have attempted to decipher the genetic makeup of pancreatic ductal adenocarcinoma and preneoplastic pancreatic cystic neoplasms. RECENT FINDINGS: With the advent of high throughput sequencing, the genetic code of pancreatic cancer is beginning to unravel and this new-found information heralds an era of precision cancer care where treatment will be guided by the genetic code of the neoplasm. Results from these studies have pointed towards the complexity and heterogeneity of the pancreatic cancer genome, provided avenues to "tailor therapy" based as well as shed light on progression of preneoplastic pancreatic neoplasms into full blown invasive pancreatic ductal adenocarcinoma. SUMMARY: While this progress has made us closer to the model of precision medicine, significant obstacles need to be overcome to use this new-found information to change the way we manage patients with pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/genética , Genômica , Cisto Pancreático/patologia , Neoplasias Pancreáticas/genética , Medicina de Precisão , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/fisiopatologia , Carcinoma Ductal Pancreático/terapia , Progressão da Doença , Predisposição Genética para Doença , Genômica/tendências , Humanos , Neoplasias Pancreáticas/fisiopatologia , Neoplasias Pancreáticas/terapia , Medicina de Precisão/tendências , Prognóstico
14.
Pancreatology ; 17(1): 7-12, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28034553

RESUMO

Every year, nearly 300,000 people are diagnosed with pancreatic cancer worldwide, and an equivalent number succumb to this disease. One of the major challenges of pancreatic cancer that contributes to its poor survival rates is the development of resistance to the standard chemotherapy. Heterogeneity of the tumor, the dense fibroblastic stroma, and the aggressive biology of the tumor all contribute to the chemoresistant phenotype. In addition, the acellular components of the tumor microenvironment like hypoxia, stress pathways in the stromal cells, and the cytokines that are secreted by the immune cells, have a definitive role in orchestrating the chemoresistant property of the tumor. In this review, we systematically focus on the role played by the different microenvironmental components in determining chemoresistance of pancreatic tumors.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/fisiopatologia , Microambiente Tumoral/fisiologia
15.
J Surg Oncol ; 116(1): 114-122, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28543919

RESUMO

The heat shock response in pancreatitis that is activated via HSP70 protects acinar cells through multiple simultaneous mechanisms. It inhibits trypsinogen activation and modulates NF-κB signaling to limit acinar cell injury. On the other hand, HSP70 is overexpressed in pancreatic cancer and is hijacked by the cellular machinery to inhibit apoptosis. Inhibition of HSP70 in pancreatic cancer by a novel compound, Minnelide, has shown considerable clinical promise.


Assuntos
Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/metabolismo , Organofosfatos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Fenantrenos/farmacologia , Animais , Ensaios Clínicos Fase I como Assunto , Diterpenos , Compostos de Epóxi , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/metabolismo , Pancreatite/metabolismo
16.
J Biol Chem ; 290(21): 13321-43, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25825498

RESUMO

Mycobacterium tuberculosis employs various strategies to modulate host immune responses to facilitate its persistence in macrophages. The M. tuberculosis cell wall contains numerous glycoproteins with unknown roles in pathogenesis. Here, by using Concanavalin A and LC-MS analysis, we identified a novel mannosylated glycoprotein phosphoribosyltransferase, encoded by Rv3242c from M. tuberculosis cell walls. Homology modeling, bioinformatic analyses, and an assay of phosphoribosyltransferase activity in Mycobacterium smegmatis expressing recombinant Rv3242c (MsmRv3242c) confirmed the mass spectrometry data. Using Mycobacterium marinum-zebrafish and the surrogate MsmRv3242c infection models, we proved that phosphoribosyltransferase is involved in mycobacterial virulence. Histological and infection assays showed that the M. marinum mimG mutant, an Rv3242c orthologue in a pathogenic M. marinum strain, was strongly attenuated in adult zebrafish and also survived less in macrophages. In contrast, infection with wild type and the complemented ΔmimG:Rv3242c M. marinum strains showed prominent pathological features, such as severe emaciation, skin lesions, hemorrhaging, and more zebrafish death. Similarly, recombinant MsmRv3242c bacteria showed increased invasion in non-phagocytic epithelial cells and longer intracellular survival in macrophages as compared with wild type and vector control M. smegmatis strains. Further mechanistic studies revealed that the Rv3242c- and mimG-mediated enhancement of intramacrophagic survival was due to inhibition of autophagy, reactive oxygen species, and reduced activities of superoxide dismutase and catalase enzymes. Infection with MsmRv3242c also activated the MAPK pathway, NF-κB, and inflammatory cytokines. In summary, we show that a novel mycobacterial mannosylated phosphoribosyltransferase acts as a virulence and immunomodulatory factor, suggesting that it may constitute a novel target for antimycobacterial drugs.


Assuntos
Autofagia , Macrófagos/imunologia , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Nicotinamida Fosforribosiltransferase/metabolismo , Estresse Oxidativo , Tuberculose/imunologia , Peixe-Zebra/imunologia , Animais , Apoptose , Western Blotting , Adesão Celular , Movimento Celular , Proliferação de Células , Parede Celular/metabolismo , Células Cultivadas , Feminino , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Mycobacterium marinum/crescimento & desenvolvimento , Mycobacterium tuberculosis/crescimento & desenvolvimento , NF-kappa B , Nicotinamida Fosforribosiltransferase/genética , Fagocitose , Conformação Proteica , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Tuberculose/metabolismo , Tuberculose/microbiologia , Virulência/imunologia , Peixe-Zebra/metabolismo , Peixe-Zebra/microbiologia
17.
J Cell Biochem ; 117(2): 279-88, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26206252

RESUMO

Pancreatic cancer is estimated to be the 12th most common cancer in the United States in 2014 and yet this malignancy is the fourth leading cause of cancer-related death in the United States. Late detection and resistance to therapy are the major causes for its dismal prognosis. Apoptosis is an actively orchestrated cell death mechanism that serves to maintain tissue homoeostasis. Cancer develops from normal cells by accruing significant changes through one or more mechanisms, leading to DNA damage and mutations, which in a normal cell would induce this programmed cell death pathway. As a result, evasion of apoptosis is one of the hallmarks of cancer cells. PDAC is notoriously resistant to apoptosis, thereby explaining its aggressive nature and resistance to conventional treatment modalities. The current review is focus on understanding different intrinsic and extrinsic pathways in pancreatic cancer that may affect apoptosis in this disease.


Assuntos
Apoptose , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/patologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proteínas Reguladoras de Apoptose/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Humanos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/tratamento farmacológico
18.
Lab Invest ; 96(12): 1268-1278, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27775688

RESUMO

NF-κB has an essential role in the initiation and progression of pancreatic cancer and specifically mediates the induction of epithelial-mesenchymal transition and invasiveness. In this study, we demonstrate the importance of activated NF-κB signaling in EMT induction, lymphovascular metastasis, and neural invasion. Modulation of NF-κB activity was accomplished through the specific NF-κB inhibitor (BAY 11-7085), triptolide, and Minnelide treatment, as well as overexpression of IKBα repressor and IKK activator plasmids. In the classical lymphovascular metastatic cascade, inhibition of NF-κB decreased the expression of several EMT transcription factors (SNAI1, SNAI2, and ZEB1) and mesenchymal markers (VIM and CDH2) and decreased in vitro invasion, which was rescued by IKK activation. This was further demonstrated in vivo via BAY 11-7085 treatment in a orthotopic model of pancreatic cancer. In vivo NF-κB inhibition decreased tumor volume; decreased tumor EMT gene expression, while restoring cell-cell junctions; and decreasing overall metastasis. Furthermore, we demonstrate the importance of active NF-κB signaling in neural invasion. Triptolide treatment inhibits Nerve Growth Factor (NGF) mediated, neural-tumor co-culture in vitro invasion, and dorsal root ganglia (DRG) neural outgrowth through a disruption in tumor-neural cross talk. In vivo, Minnelide treatment decreased neurotrophin expression, nerve density, and sciatic nerve invasion. Taken together, this study demonstrates the importance of NF-κB signaling in the progression of pancreatic cancer through the modulation of EMT induction, lymphovascular invasion, and neural invasion.


Assuntos
Transição Epitelial-Mesenquimal , NF-kappa B/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Nervos Periféricos/metabolismo , Neoplasias do Sistema Nervoso Periférico/secundário , Transdução de Sinais , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Humanos , Metástase Linfática/patologia , Metástase Linfática/prevenção & controle , Camundongos , Camundongos Nus , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/antagonistas & inibidores , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Nervos Periféricos/citologia , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Neoplasias do Sistema Nervoso Periférico/prevenção & controle , Proteínas Recombinantes/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Transdução de Sinais/efeitos dos fármacos
19.
Am J Physiol Gastrointest Liver Physiol ; 311(5): G974-G980, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27686613

RESUMO

In the current study, we have characterized the global miRNA expression profile in mouse pancreatic acinar cells and during acute pancreatitis using next-generation RNA sequencing. We identified 324 known and six novel miRNAs that are expressed in mouse pancreatic acinar cells. In the basal state, miR-148a-3p, miR-375-3p, miR-217-5p, and miR-200a-3p were among the most abundantly expressed, whereas miR-24-5p and miR-421-3p were the least abundant. Treatment of acinar cells with caerulein (100 nM) and taurolithocholic acid 3-sulfate [TLC-S (250 µM)] induced numerous changes in miRNA expression profile. In particular, we found significant overexpression of miR-21-3p in acini treated with caerulein and TLC-S. We further looked at the expression of miR-21-3p in caerulein, l-arginine, and caerulein + LPS-induced acute pancreatitis mouse models and found 12-, 21-, and 50-fold increased expression in the pancreas, respectively. In summary, this is the first comprehensive analysis of global miRNA expression profile of mouse pancreatic acinar cells in normal and disease conditions. Our analysis shows that miR-21-3p expression level correlates with the severity of the disease.


Assuntos
Células Acinares/metabolismo , MicroRNAs/metabolismo , Pancreatite/metabolismo , Células Acinares/efeitos dos fármacos , Animais , Ceruletídeo/farmacologia , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , MicroRNAs/genética , Pancreatite/genética , Ácido Taurolitocólico/análogos & derivados , Ácido Taurolitocólico/farmacologia
20.
Curr Opin Gastroenterol ; 32(5): 401-407, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27454027

RESUMO

PURPOSE OF REVIEW: Pancreatic cancer is the most devastating of all cancers with an extremely poor prognosis. In US alone, over 50 000 new cases of pancreatic cancer are reported annually, and about the same number succumb to it, making pancreatic cancer the third most common cause of cancer deaths. Most patients with pancreatic cancer present with advanced disease, which cannot be resected surgically, and for these patients chemotherapy is the only option. Even patients who undergo resection require adjuvant therapy to decrease the risk of recurrence. Since the 1950s, a variety of different agents, like antimetabolites, nucleoside analogs, and DNA intercalating compounds, have been used against pancreatic cancer, alone or in combination, with little improvement in the survival statistics. The current article reviews the evolution of chemotherapy for pancreatic cancer, and discusses some novel therapeutic options that are emerging in recent times, with special emphasis on Minnelide, a novel HSP70 inhibitor, which is currently in clinical trials. RECENT FINDINGS: Approaches towards developing therapies for pancreatic cancer have evolved tremendously over the past decade. Research has shown that apart from the inherent drug resistance, drug delivery to pancreatic cancer has also posed a major challenge. The extensive desmoplastic stroma of pancreatic cancer is believed to create inordinately high interstitial fluid pressures leading to vascular collapse and substantial barrier to perfusion of chemotherapeutics, thus creating an additional layer of protection for pancreatic cancer. Recent research thus is focused not only on understanding the biology and developing strategies to target cancer cells, but also is targeted towards the depletion of stroma in order to ensure better delivery of chemotherapeutic compounds to the tumor. SUMMARY: The current article describes the novel therapies that are constantly being evaluated to address and overcome the challenges that make pancreatic cancer a difficult disease to treat.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa