Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(1): 80-93, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25259922

RESUMEN

The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Calcitriol/análogos & derivados , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptores de Calcitriol/metabolismo , Adenocarcinoma/patología , Animales , Calcitriol/farmacología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neoplasias Pancreáticas/patología , Pancreatitis/tratamiento farmacológico , Pancreatitis/prevención & control , Transducción de Señal , Células del Estroma/patología
2.
Proteomics ; 24(11): e2300067, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570832

RESUMEN

Small extracellular vesicles (sEVs) are cell-derived vesicles evolving as important elements involved in all stages of cancers. sEVs bear unique protein signatures that may serve as biomarkers. Pancreatic cancer (PC) records a very poor survival rate owing to its late diagnosis and several cancer cell-derived proteins have been reported as candidate biomarkers. However, given the pivotal role played by stellate cells (PSCs, which produce the collagenous stroma in PC), it is essential to also assess PSC-sEV cargo in biomarker discovery. Thus, this study aimed to isolate and characterise sEVs from mouse PC cells and PSCs cultured alone or as co-cultures and performed proteomic profiling and pathway analysis. Proteomics confirmed the enrichment of specific markers in the sEVs compared to their cells of origin as well as the proteins that are known to express in each of the culture types. Most importantly, for the first time it was revealed that PSC-sEVs are enriched in proteins (including G6PI, PGAM1, ENO1, ENO3, and LDHA) that mediate pathways related to development of diabetes, such as glucose metabolism and gluconeogenesis revealing a potential role of PSCs in pancreatic cancer-related diabetes (PCRD). PCRD is now considered a harbinger of PC and further research will enable to identify the role of these components in PCRD and may develop as novel candidate biomarkers of PC.


Asunto(s)
Vesículas Extracelulares , Neoplasias Pancreáticas , Células Estrelladas Pancreáticas , Proteómica , Animales , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Ratones , Vesículas Extracelulares/metabolismo , Proteómica/métodos , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proteoma/análisis , Proteoma/metabolismo
3.
Pancreatology ; 24(4): 522-527, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38704341

RESUMEN

BACKGROUND: The global incidence of acute pancreatitis (AP) is increasing, but little information exists about trends in Australia. This study aimed to describe incidence trends, along with clinical and socio-demographic associations, in the state of Tasmania over a recent 12-year period. METHODS: The study cohort was obtained by linking clinical and administrative datasets encompassing the whole Tasmanian population between 2007 and 2018, inclusive. Pancreatitis case definition was based on relevant ICD-10 hospitalization codes, or elevated serum lipase or amylase in pathology data. Age-standardised incidence rates were estimated, overall and stratified by sex, aetiology, and Index of Relative Socio-economic Disadvantage (IRSD). RESULTS: In the study period, 4905 public hospital AP episodes were identified in 3503 people. The age-standardised person-based incidence rate across the entire period was 54 per 100,000 per year. Incidence was inversely related to IRSD score; 71 per 100,000 per year in the most disadvantaged quartile compared to 32 in the least disadvantaged. Biliary AP incidence was higher than that of alcohol-related AP, although the greatest incidence was in "unspecified" cases. There was an increase in incidence for the whole cohort (average annual percent change 3.23 %), largely driven by the two most disadvantaged IRSD quartiles; the least disadvantaged quartile saw a slight overall decrease. CONCLUSION: This is the first Australian study providing robust evidence that AP incidence is increasing and is at the upper limit of population-based studies worldwide. This increased incidence is greatest in socio-economically disadvantaged areas, meriting further research to develop targeted, holistic management strategies.


Asunto(s)
Pancreatitis , Humanos , Tasmania/epidemiología , Pancreatitis/epidemiología , Masculino , Femenino , Incidencia , Persona de Mediana Edad , Anciano , Adulto , Estudios de Cohortes , Anciano de 80 o más Años , Enfermedad Aguda , Factores Socioeconómicos , Adulto Joven , Adolescente
4.
Pancreatology ; 21(1): 103-114, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33309014

RESUMEN

BACKGROUND: The detection and quantification of circulating tumour cells (CTCs) in pancreatic cancer (PC) has the potential to provide prognostic information. The aim of this review was to provide an overview of the literature surrounding CTCs in PC. METHODS: A systematic literature review on CTCs in PC between 2005-2020 was performed. Data based on peripheral vein samples were used to determine the positivity rate of CTCs, their prognostic significance and their relative numbers compared to portal vein (PV) samples. RESULTS: The overall CTC detection rate in forty-four articles was 65% (95%CI: 55-75%). Detection rate for CellSearch was 26% (95%CI: 14-38%), which was lower than for both filtration and microfluidic techniques. In nine studies with >50 patients, overall survival was worse with CTC positivity (HR 1.82; 95%CI: 1.61-2.05). Five of seven studies which described PV CTC collection provided patient-level data. PV CTC yield was 7.7-fold (95%CI 1.35-43.9) that of peripheral blood. CONCLUSIONS: CTCs were detected in the peripheral circulation of most patients with PC and may be related to prognosis and disease stage. PV blood contains more CTCs than peripheral blood sampling. This review points to the maturation of techniques of CTC enrichment, and its evidence base for eventual clinical deployment.


Asunto(s)
Células Neoplásicas Circulantes/patología , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/patología , Humanos
5.
Br J Cancer ; 122(10): 1486-1495, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32203220

RESUMEN

BACKGROUND: Stromal-tumour interactions facilitate pancreatic cancer (PC) progression. The hepatocyte growth factor (HGF)/c-MET pathway is upregulated in PC and mediates the interaction between cancer cells and stromal pancreatic stellate cells (PSCs). This study assessed the effect of HGF/c-MET inhibition plus gemcitabine (G) on the progression of advanced PC. METHODS: Orthotopic PC was produced by implantation of luciferase-tagged human cancer cells + human PSCs into mouse pancreas. Tumours were allowed to develop without treatment for 4 weeks. Mice were then treated for 6 weeks with one of the following: IgG, G, HGF inhibitor (Hi), c-MET inhibitor (Ci), Hi + Ci, Hi + G, Ci + G, or Hi + Ci + G. RESULTS: Bioluminescence imaging showed similar tumour sizes in all mice at the initiation of treatments. Triple therapy (Hi + Ci + G): (1) completely eliminated metastasis; (2) significantly reduced tumour size as assessed by bioluminescence and at necropsy; (3) significantly reduced proliferating cancer cell density and stem cell marker DCLK1 expression in tumours. In vitro 3D culture studies supported our in vivo findings. CONCLUSION: Even at an advanced disease stage, a two-pronged approach, targeting (a) HGF/c-MET with relevant inhibitors and (b) cancer cells with chemotherapy, completely eliminated metastasis and significantly decreased tumour growth, suggesting that this is a promising treatment approach for PC.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Quinasas Similares a Doblecortina , Factor de Crecimiento de Hepatocito/genética , Humanos , Inmunoglobulina G/farmacología , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Células Madre Neoplásicas , Páncreas/efectos de los fármacos , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
6.
Pancreatology ; 20(3): 409-418, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31928917

RESUMEN

Tumour-stromal interactions have now been acknowledged to play a major role in pancreatic cancer (PC) progression. The abundant collagenous stroma is produced by a specific cell type in the pancreas-the pancreatic stellate cell (PSC). Pancreatic stellate cells (PSCs) are a unique resident cell type of pancreas and with a critical role in both healthy and diseased pancreas. Accumulating evidence indicates that PSCs interact closely with cancer cells as well as with other cell types of the stroma such as immune cells, endothelial cells and neuronal cells, to set up a growth permissive microenvironment for pancreatic tumours, which facilitates local tumour growth as well as distant metastasis. Consequently, recent work in the field has focused on the development of novel therapeutic approaches targeting the stroma to inhibit PC progression. Such a multi-pronged approach targeting both tumour and stromal elements of PC has been successfully applied in pre-clinical settings. The challenge now is to translate the pre-clinical findings into the clinical setting to achieve better outcomes for pancreatic cancer patients.


Asunto(s)
Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Progresión de la Enfermedad , Humanos , Microambiente Tumoral
7.
Int J Mol Sci ; 21(23)2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271944

RESUMEN

Pancreatic cancer (pancreatic ductal adenocarcinoma (PDAC/PC)) has been an aggressive disease that is associated with early metastases. It is characterized by dense and collagenous desmoplasia/stroma, predominantly produced by pancreatic stellate cells (PSCs). PSCs interact with cancer cells as well as other stromal cells, facilitating disease progression. A candidate growth factor pathway that may mediate this interaction is the hepatocyte growth factor (HGF)/c-MET pathway. HGF is produced by PSCs and its receptor c-MET is expressed on pancreatic cancer cells and endothelial cells. The current review discusses the role of the MET/HGF axis in tumour progression and dissemination of pancreatic cancer. Therapeutic approaches that were developed targeting either the ligand (HGF) or the receptor (c-MET) have not been shown to translate well into clinical settings. We discuss a two-pronged approach of targeting both the components of this pathway to interrupt the stromal-tumour interactions, which may represent a potential therapeutic strategy to improve outcomes in PC.


Asunto(s)
Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Humanos , Estadificación de Neoplasias , Neovascularización Patológica , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Transducción de Señal , Microambiente Tumoral
8.
Gastroenterology ; 153(6): 1674-1686, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847752

RESUMEN

BACKGROUND & AIMS: Smoking, an independent risk factor for pancreatitis, accelerates the development of alcoholic pancreatitis. Alcohol feeding of mice induces up-regulation of spliced X-box binding protein 1 (XBP1s), which regulates the endoplasmic reticulum (ER) unfolded protein response and promotes cell survival upon ER stress. We examined whether smoking affects the adaptive mechanisms induced by alcohol and accelerates disorders of the ER in pancreatic acinar cells. METHODS: We studied the combined effects of ethanol (EtOH) and cigarette smoke extract (CSE) on ER stress and cell death responses in mouse and human primary acini and the acinar cell line AR42J. Cells were incubated with EtOH (50 mmol/L), CSE (20-40 µg/mL), or both (CSE+EtOH), and analyzed by immunoblotting, quantitative reverse-transcription polymerase chain reaction, and cell death assays. Some cells were incubated with MKC-3946, an inhibitor of endoplasmic reticulum to nucleus signaling 1 (ERN1, also called IRE1) that blocks XBP1s formation. Male Sprague-Dawley rats were fed isocaloric amounts of an EtOH-containing (Lieber-DeCarli) or control diet for 11 weeks and exposed to cigarette smoke or room air in an exposure chamber for 2 hours each day. During the last 3 weeks, a subset of rats received intravenous injections of lipopolysaccharide (LPS, 3 mg/kg per week) to induce pancreatitis or saline (control). Pancreatic tissues were collected and analyzed by histology and immunostaining techniques. RESULTS: In AR42J and primary acini, CSE+EtOH induced cell death (necrosis and apoptosis), but neither agent alone had this effect. Cell death was associated with a significant decrease in expression of XBP1s. CSE+EtOH, but neither agent alone, slightly decreased adenosine triphosphate levels in AR42J cells, but induced oxidative stress and sustained activation (phosphorylation) of eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3, also called PERK) and increased protein levels of DNA damage inducible transcript 3 (DDIT3, also called CHOP). CHOP regulates transcription to promote apoptosis. Incubation of AR42J or primary mouse or human acinar cells with MKC-3946 reduced expression of XBP1s, increased levels of CHOP, and induced cell death. In rats fed an EtOH diet, exposure to cigarette smoke increased ER stress in acinar cells and sensitized the pancreas to LPS-induced pathology. CONCLUSIONS: Cigarette smoke promotes cell death and features of pancreatitis in EtOH-sensitized acinar cells by suppressing the adaptive unfolded protein response signaling pathway. It also activates ER stress pathways that promote acinar cell death.


Asunto(s)
Células Acinares/efectos de los fármacos , Consumo de Bebidas Alcohólicas/efectos adversos , Fumar Cigarrillos/efectos adversos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Etanol/toxicidad , Páncreas Exocrino/efectos de los fármacos , Pancreatitis Alcohólica/etiología , Humo/efectos adversos , Células Acinares/metabolismo , Células Acinares/patología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Humanos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Estrés Oxidativo/efectos de los fármacos , Páncreas Exocrino/metabolismo , Páncreas Exocrino/patología , Pancreatitis Alcohólica/metabolismo , Pancreatitis Alcohólica/patología , Ratas Sprague-Dawley , Factores de Riesgo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Respuesta de Proteína Desplegada/efectos de los fármacos
9.
Carcinogenesis ; 38(6): 588-591, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28379317

RESUMEN

Pancreatic stellate cells (PSCs) are known to play an important role in facilitating pancreatic cancer progression-both in terms of local tumour growth as well as the establishment of metastases. We have previously demonstrated that PSCs from the primary cancer seed to distant metastatic sites. We therefore hypothesise that PSCs circulate along with pancreatic cancer cells (circulating tumour cells-CTCs) to help create a growth permissive microenvironment at distant metastatic sites. This review aims to explore the concept of circulating PSCs in pancreatic cancer and suggests future directions for research in this area.


Asunto(s)
Células Neoplásicas Circulantes/patología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Microambiente Tumoral , Animales , Comunicación Celular , Humanos , Metástasis de la Neoplasia , Células del Estroma
10.
Curr Opin Gastroenterol ; 33(5): 366-373, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28590306

RESUMEN

PURPOSE OF REVIEW: Pancreatic stellate cells (PSCs) play an integral role in the pathogenesis of pancreatitis and pancreatic cancer. With the developing knowledge of this important cell type, we are at the cusp of developing effective therapies for the above diseases based upon targeting the PSC and modulating its function. RECENT FINDINGS: The major themes of the recent PSC literature include: PSC interactions with the extracellular matrix and other stromal components; intracellular calcium physiology as drivers of mechanical interactions and necrosis; the relationship between proinflammatory, protumoural, angiogenic, and metabolic pathways in pancreatic necrosis, fibrosis, and carcinogenesis; and targeting of the stroma for antitumoural and antifibrotic effects. SUMMARY: Traditionally, there have been few treatment options for pancreatitis and pancreatic cancer. The elucidation of the wide-ranging functions of PSCs provide an opportunity for treatments based on stromal reprogramming.


Asunto(s)
Fibrosis/terapia , Terapia Molecular Dirigida , Neoplasias Pancreáticas/terapia , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Pancreatitis/terapia , Biomarcadores de Tumor , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/fisiología , Fibrosis/patología , Humanos , Terapia Molecular Dirigida/tendencias , Neoplasias Pancreáticas/patología , Pancreatitis/patología
11.
Med J Aust ; 207(4): 161-165, 2017 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-28814218

RESUMEN

INTRODUCTION: In 2015, the Australasian Pancreatic Club (APC) published the Australasian guidelines for the management of pancreatic exocrine insufficiency (http://pancreas.org.au/2016/01/pancreatic-exocrine-insufficiency-guidelines). Pancreatic exocrine insufficiency (PEI) occurs when normal digestion cannot be sustained due to insufficient pancreatic digestive enzyme activity. This may be related to a breakdown, at any point, in the pancreatic digestive chain: pancreatic stimulation; synthesis, release or transportation of pancreatic enzymes; or synchronisation of secretions to mix with ingested food. Main recommendations: The guidelines provide advice on diagnosis and management of PEI, noting the following: A high prevalence of PEI is seen in certain diseases and conditions, such as cystic fibrosis, acute and chronic pancreatitis, pancreatic cancer and pancreatic surgery. The main symptoms of PEI are steatorrhoea or diarrhoea, abdominal pain, bloating and weight loss. These symptoms are non-specific and often go undetected and untreated. PEI diagnosis is predominantly based on clinical findings and the presence of underlying disease. The likelihood of PEI in suspected patients has been categorised into three groups: definite, possible and unlikely. If left untreated, PEI may lead to complications related to fat malabsorption and malnutrition, and have an impact on quality of life. Pancreatic enzyme replacement therapy (PERT) remains the mainstay of PEI treatment with the recommended adult initial enzyme dose being 25 000-40 000 units of lipase per meal, titrating up to a maximum of 75 000-80 000 units of lipase per meal. Adjunct acid-suppressing therapy may be useful when patients still experience symptoms of PEI on high dose PERT. Nutritional management by an experienced dietitian is essential. Changes in management as a result of these guidelines: These are the first guidelines to classify PEI as being definite, possible or unlikely, and provide a diagnostic algorithm to facilitate the early diagnosis of PEI and appropriate use of PERT.


Asunto(s)
Manejo de la Enfermedad , Terapia de Reemplazo Enzimático , Insuficiencia Pancreática Exocrina/diagnóstico , Insuficiencia Pancreática Exocrina/terapia , Australia , Humanos , Apoyo Nutricional , Guías de Práctica Clínica como Asunto , Calidad de Vida , Sociedades Médicas
12.
Br J Cancer ; 114(3): 269-80, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26766740

RESUMEN

BACKGROUND: Pancreatic stellate cells (PSCs, which produce the stroma of pancreatic cancer (PC)) interact with cancer cells to facilitate PC growth. A candidate growth factor pathway that may mediate this interaction is the HGF-c-MET pathway. METHODS: Effects of HGF inhibition (using a neutralising antibody AMG102) alone or in combination with gemcitabine were assessed (i) in vivo using an orthotopic model of PC, and (ii) in vitro using cultured PC cells (AsPC-1) and human PSCs. RESULTS: We have shown that human PSCs (hPSCs) secrete HGF but do not express the receptor c-MET, which is present predominantly on cancer cells. HGF inhibition was as effective as standard chemotherapy in inhibiting local tumour growth but was significantly more effective than gemcitabine in reducing tumour angiogenesis and metastasis. HGF inhibition has resulted in reduced metastasis; however, interestingly this antimetastatic effect was lost when combined with gemcitabine. This suggests that gemcitabine treatment selects out a subpopulation of cancer cells with increased epithelial-mesenchymal transition (EMT) and stem-cell characteristics, as supported by our findings of increased expression of EMT and stem-cell markers in tumour sections from our animal model. In vitro studies showed that hPSC secretions induced proliferation and migration, but inhibited apoptosis, of cancer cells. These effects were countered by pretreatment of hPSC secretions with a HGF-neutralising antibody but not by gemcitabine, indicating a key role for HGF in PSC-PC interactions. CONCLUSIONS: Our studies suggest that targeted therapy to inhibit stromal-tumour interactions mediated by the HGF-c-MET pathway may represent a novel therapeutic approach in PC that will require careful modelling for optimal integration with existing treatment modalities.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/metabolismo , Desoxicitidina/análogos & derivados , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células Estrelladas Pancreáticas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
14.
Curr Opin Gastroenterol ; 31(5): 416-23, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26125317

RESUMEN

PURPOSE OF REVIEW: Ever since the first descriptions of methods to isolate pancreatic stellate cells (PSCs) from rodent and human pancreas 17 years ago, rapid advances have been made in our understanding of the biology of these cells and their functions in health and disease. This review updates recent literature in the field, which indicates an increasingly complex role for the cells in normal pancreas, pancreatitis and pancreatic cancer. RECENT FINDINGS: Work reported over the past 12 months includes improved methods of PSC immortalization, a role for PSCs in islet fibrosis, novel factors causing PSC activation as well as those inducing quiescence, and translational research aimed at inhibiting the facilitatory effects of PSCs on disease progression in chronic pancreatitis as well as pancreatic cancer. SUMMARY: Improved understanding of the role of PSCs in pancreatic pathophysiology has prompted a focus on translational studies aimed at developing novel approaches to modulate PSC function in a bid to improve clinical outcomes of two major fibrotic diseases of the pancreas: chronic pancreatitis and pancreatic cancer.


Asunto(s)
Fibrosis/fisiopatología , Páncreas/patología , Neoplasias Pancreáticas/fisiopatología , Células Estrelladas Pancreáticas/fisiología , Pancreatitis Crónica/fisiopatología , Actinas/biosíntesis , Comunicación Celular , Progresión de la Enfermedad , Regulación de la Expresión Génica/fisiología , Humanos , Páncreas/citología , Transducción de Señal
15.
Alcohol Clin Exp Res ; 39(11): 2123-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26463405

RESUMEN

BACKGROUND: Chronic pancreatitis, a known complication of alcohol abuse, is characterized histopathologically by prominent fibrosis. Pancreatic stellate cells (PSCs) are responsible for producing this fibrous tissue in chronic pancreatitis and are activated by alcohol. Progression of alcoholic chronic pancreatitis (as assessed by calcification and fibrosis) is thought to be facilitated by concurrent smoking, but the mechanisms are unknown. This study aimed to (a) determine whether human PSCs (hPSCs) and rat PSCs express nicotinic acetylcholine receptors (nAChRs), which are known to bind 2 important components of cigarette smoke, namely nicotine and nicotine-derived nitrosamine ketone (NNK), and (b) examine the effects of cigarette smoke components in the presence and absence of alcohol on PSC activation in vitro. METHODS: Western blotting was used to detect the presence of nAChRs in primary cultures of PSCs. Clinically relevant concentrations of cigarette smoke components (either cigarette smoke extract [CSE], NNK, or nicotine) ± ethanol (EtOH) were used to treat primary cultures of PSCs, and stellate cell activation was assessed by cell migration, proliferation, collagen production, and apoptosis. RESULTS: We demonstrate, for the first time, that PSCs express nAChRs (isoforms α3, α7, ß, ε) and that the expression of the α7 isoform in hPSCs is induced by CSE + EtOH. We also provide novel findings that PSCs are activated by CSE and NNK (both alone and in combination with EtOH) as evidenced by an increase in cell migration and/or proliferation. Further, we demonstrate that activation of PSCs by CSE + EtOH and NNK + EtOH may be mediated via nAChRs on the cells. CONCLUSIONS: PSCs are activated by clinically relevant concentrations of cigarette smoke components (CSE and NNK), alone and in combination with EtOH. Thus, in alcoholics who smoke, progression of pancreatic fibrosis may be facilitated by the combined effects of alcohol and cigarette smoke components on hPSC behavior.


Asunto(s)
Etanol/toxicidad , Nicotiana/toxicidad , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/patología , Pancreatitis Alcohólica/patología , Humo/efectos adversos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Pancreatitis Alcohólica/inducido químicamente , Fumar/efectos adversos , Fumar/patología
16.
Carcinogenesis ; 35(8): 1891-900, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24876152

RESUMEN

Activated cancer-associated human pancreatic stellate cells (CAhPSCs, which produce the collagenous stroma of pancreatic cancer [PC]) are known to play a major role in PC progression. Apart from inducing cancer cell proliferation and migration, CAhPSCs have also been implicated in neoangiogenesis in PC. However, the mechanisms mediating the observed angiogenic effects of CAhPSCs are unknown. A candidate pathway that may be involved in this process is the hepatocyte growth factor (HGF)/c-MET pathway and its helper molecule, urokinase-type plasminogen activator (uPA). This study investigated the effects of CAhPSC secretions on endothelial cell function in the presence and absence of HGF, c-MET and uPA inhibitors. HGF levels in CAhPSC secretions were quantified using ELISA. CAhPSC secretions were then incubated with human microvascular endothelial cells (HMEC-1) and angiogenesis assessed by quantifying HMEC-1 tube formation and proliferation. CAhPSC-secreted HGF significantly increased HMEC-1 tube formation and proliferation; notably, these effects were downregulated by inhibition of HGF, its receptor c-MET and uPA. Phosphorylation of p38 mitogen-activated protein kinase was downregulated during inhibition of the HGF/c-MET pathway, whereas phosphatidylinositol-3 kinase and ERK1/2 remained unaffected. Our studies have shown for the first time that CAhPSCs induce proliferation and tube formation of HMEC-1 and that the HGF/c-MET pathway plays a major role in this induction. Given that standard antiangiogenic treatment targeting vascular endothelial growth factor has had limited success in the clinical setting, the findings of the current study provide strong support for a novel, alternative antiangiogenic approach targeting the HGF/c-MET and uPA pathways in PC.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Comunicación Celular/fisiología , Endotelio Vascular/patología , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Ductal Pancreático/metabolismo , Comunicación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Gastroenterology ; 144(6): 1210-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23622130

RESUMEN

Pancreatic ductal adenocarcinoma is a devastating disease, and patient outcomes have not improved in decades. Treatments that target tumor cells have largely failed. This could be because research has focused on cancer cells and the influence of the stroma on tumor progression has been largely ignored. The focus of pancreatic cancer research began to change with the identification of pancreatic stellate cells, which produce the pancreatic tumor stroma. There is compelling in vitro and in vivo evidence for the influence of pancreatic stellate cells on pancreatic cancer development; several recent preclinical studies have reported encouraging results with approaches designed to target pancreatic stellate cells and the stroma. We review the background and recent advances in these areas, along with important areas of future research that could improve therapy.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Páncreas/patología , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología , Microambiente Tumoral , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Células Estrelladas Pancreáticas/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/patología
18.
Am J Physiol Gastrointest Liver Physiol ; 305(6): G408-17, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23868411

RESUMEN

Activated pancreatic stellate cells (PSCs) are responsible for the fibrotic matrix of chronic pancreatitis and pancreatic cancer. In vitro protocols examining PSC biology have usually involved PSCs cultured on plastic, a nonphysiological surface. However, PSCs cultured on physiological matrices, e.g., Matrigel (normal basement membrane) and collagen (fibrotic pancreas), may have distinctly different behaviors compared with cells cultured on plastic. Therefore, we aimed to 1) compare PSC gene expression after culture on plastic, Matrigel, and collagen I; 2) validate the gene array data for transgelin, the most highly dysregulated gene in PSCs grown on activating vs. nonactivating matrices, at mRNA and protein levels; 3) examine the role of transgelin in PSC function; and 4) assess transgelin expression in human chronic pancreatitis sections. Culture of PSCs on different matrices significantly affected their gene expression pattern. 146, 619, and 432 genes, respectively, were differentially expressed (P < 0.001) in PSCs cultured on collagen I vs. Matrigel, Matrigel vs. plastic, and collagen I vs. plastic. The highest fold change (12.5-fold upregulation) in gene expression in cells on collagen I vs. Matrigel was observed for transgelin (an actin stress fiber-associated protein). Transgelin was significantly increased in activated PSCs vs. quiescent PSCs. Silencing transgelin expression decreased PSC proliferation and also reduced platelet-derived growth factor-induced PSC migration. Notably, transgelin was highly expressed in chronic pancreatitis in stromal areas and periacinar spaces but was absent in acinar cells. These findings suggest that transgelin is a potentially useful target protein to modulate PSC function so as to ameliorate pancreatic fibrosis.


Asunto(s)
Matriz Extracelular/química , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Transcripción Genética , Animales , Proliferación Celular , Células Cultivadas , Colágeno/farmacología , Combinación de Medicamentos , Matriz Extracelular/metabolismo , Humanos , Laminina/farmacología , Masculino , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/fisiología , Plásticos/farmacología , Proteoglicanos/farmacología , Ratas , Ratas Sprague-Dawley
19.
Proc Natl Acad Sci U S A ; 107(40): 17397-402, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20852067

RESUMEN

The pancreatic secretagogue cholecystokinin (CCK) is widely thought to stimulate enzyme secretion by acinar cells indirectly via activation of the vagus nerve. We postulate an alternative pathway for CCK-induced pancreatic secretion. We hypothesize that neurally related pancreatic stellate cells (PSCs; located in close proximity to the basolateral aspect of acinar cells) play a regulatory role in pancreatic secretion by serving as an intermediate target for CCK and secreting the neurotransmitter acetylcholine (ACh), which, in turn, stimulates acinar enzyme secretion. To determine whether PSCs (i) exhibit CCK-dependent ACh secretion and (ii) influence acinar enzyme secretion, primary cultures of human and rat PSCs were used. Immunoblotting and/or immunofluorescence was used to detect choline acetyltransferase (ACh synthesizing enzyme), vesicular ACh transporter (VAChT), synaptophysin, and CCK receptors 1 and 2. Synaptic-like vesicles in PSCs were identified by EM. ACh secretion by PSCs exposed to 20 pM CCK was measured by LC-MS/MS. Amylase secretion by acini [pretreated with and without the muscarinic receptor antagonist atropine (10 µM) and cocultured with PSCs] was measured by colorimetry. PSCs express ACh synthesizing enzyme, VAChT, synaptophysin, and CCK receptors; exhibit CCK-dependent ACh secretion; and stimulate amylase secretion by acini, which is blocked by atropine. In conclusion, PSCs express the essential elements for ACh synthesis and secretion. CCK stimulates ACh secretion by PSCs, which, in turn, induces amylase secretion by acini. Therefore, PSCs may represent a previously unrecognized intrapancreatic pathway regulating CCK-induced pancreatic exocrine secretion.


Asunto(s)
Acetilcolina/metabolismo , Páncreas Exocrino , Amilasas/metabolismo , Animales , Células Cultivadas , Colecistoquinina/metabolismo , Colina O-Acetiltransferasa/metabolismo , Técnicas de Cocultivo , Vesículas Citoplasmáticas/metabolismo , Humanos , Páncreas Exocrino/citología , Páncreas Exocrino/metabolismo , Ratas , Receptores de Colecistoquinina/metabolismo , Sinaptofisina/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
20.
Pancreatology ; 12(4): 344-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22898636

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease. The prognosis is poor; less than 5% of those diagnosed are still alive five years after diagnosis, and complete remission is still rare. Tobacco smoking is a major risk factor of pancreatic cancer. However, the mechanism(s) through which it causes the disease remains unknown. Accumulating evidence indicates that carcinogenic compounds in cigarette smoke stimulate pancreatic cancer progression through induction of inflammation and fibrosis which act in concert with genetic factors leading to the inhibition of cell death and stimulation of proliferation resulting in the promotion of the PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Neoplasias Pancreáticas/etiología , Fumar/efectos adversos , Animales , Carcinógenos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Inflamación , Masculino , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Riesgo , Humo/análisis , Nicotiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA