Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 51(1): 90-103.e3, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31278057

RESUMEN

The key sites within the gastrointestinal (GI) tract where T cells mediate effector responses and the impact of these responses on intestinal stem cells (ISCs) remain unclear. Using experimental bone marrow transplantation to model immune-mediated GI damage and 3D imaging to analyze T cell localization, we found that the ISC compartment is the primary intestinal site targeted by T cells after transplantation. Recruitment to the crypt base region resulted in direct T cell engagement with the stem cell compartment and loss of crypt base columnar ISCs, which expressed both MHC classes I and II. Vasculature expressing the adhesion molecule MAdCAM-1 clustered near the crypt base, preferentially regulating crypt compartment invasion and ISC reduction without affecting T cell migration to villi. These findings indicate that allogeneic T cells rapidly access the stem cell niche after transplantation, and this targeted recruitment to the stem cell compartment results in ISC loss during immune-mediated GI damage.


Asunto(s)
Células Madre Adultas/inmunología , Trasplante de Médula Ósea , Mucosa Intestinal/inmunología , Nicho de Células Madre/inmunología , Linfocitos T/inmunología , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Citotoxicidad Inmunológica , Femenino , Humanos , Imagenología Tridimensional , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Modelos Animales , Mucoproteínas , Trasplante Homólogo
2.
Nature ; 528(7583): 560-564, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26649819

RESUMEN

Epithelial regeneration is critical for barrier maintenance and organ function after intestinal injury. The intestinal stem cell (ISC) niche provides Wnt, Notch and epidermal growth factor (EGF) signals supporting Lgr5(+) crypt base columnar ISCs for normal epithelial maintenance. However, little is known about the regulation of the ISC compartment after tissue damage. Using ex vivo organoid cultures, here we show that innate lymphoid cells (ILCs), potent producers of interleukin-22 (IL-22) after intestinal injury, increase the growth of mouse small intestine organoids in an IL-22-dependent fashion. Recombinant IL-22 directly targeted ISCs, augmenting the growth of both mouse and human intestinal organoids, increasing proliferation and promoting ISC expansion. IL-22 induced STAT3 phosphorylation in Lgr5(+) ISCs, and STAT3 was crucial for both organoid formation and IL-22-mediated regeneration. Treatment with IL-22 in vivo after mouse allogeneic bone marrow transplantation enhanced the recovery of ISCs, increased epithelial regeneration and reduced intestinal pathology and mortality from graft-versus-host disease. ATOH1-deficient organoid culture demonstrated that IL-22 induced epithelial regeneration independently of the Paneth cell niche. Our findings reveal a fundamental mechanism by which the immune system is able to support the intestinal epithelium, activating ISCs to promote regeneration.


Asunto(s)
Células Epiteliales/citología , Interleucinas/inmunología , Mucosa Intestinal/citología , Intestino Delgado/citología , Regeneración , Células Madre/citología , Células Madre/metabolismo , Animales , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunidad Mucosa , Interleucinas/deficiencia , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Intestino Delgado/inmunología , Intestino Delgado/patología , Ratones , Organoides/citología , Organoides/crecimiento & desarrollo , Organoides/inmunología , Células de Paneth/citología , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Nicho de Células Madre , Interleucina-22
3.
Blood ; 132(26): 2763-2774, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30381375

RESUMEN

Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a ubiquitously expressed transcription factor that is well known for its role in regulating the cellular redox pathway. Although there is mounting evidence suggesting a critical role for Nrf2 in hematopoietic stem cells and innate leukocytes, little is known about its involvement in T-cell biology. In this study, we identified a novel role for Nrf2 in regulating alloreactive T-cell function during allogeneic hematopoietic cell transplantation (allo-HCT). We observed increased expression and nuclear translocation of Nrf2 upon T-cell activation in vitro, especially in CD4+ donor T cells after allo-HCT. Allo-HCT recipients of Nrf2 -/- donor T cells had significantly less acute graft-versus-host disease (GVHD)-induced mortality, morbidity, and pathology. This reduction in GVHD was associated with the persistence of Helios+ donor regulatory T cells in the allograft, as well as defective upregulation of the gut-homing receptor LPAM-1 on alloreactive CD8+ T cells. Additionally, Nrf2 -/- donor CD8+ T cells demonstrated intact cytotoxicity against allogeneic target cells. Tumor-bearing allo-HCT recipients of Nrf2 -/- donor T cells had overall improved survival as a result of preserved graft-versus-tumor activity and reduced GVHD activity. Our findings characterized a previously unrecognized role for Nrf2 in T-cell function, as well as revealed a novel therapeutic target to improve the outcomes of allo-HCT.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Activación de Linfocitos , Factor 2 Relacionado con NF-E2/inmunología , Neoplasias Experimentales/inmunología , Enfermedad Aguda , Aloinjertos , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia
4.
Proc Natl Acad Sci U S A ; 114(18): E3709-E3718, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28420791

RESUMEN

According to current dogma, there is little or no ongoing neurogenesis in the fully developed adult enteric nervous system. This lack of neurogenesis leaves unanswered the question of how enteric neuronal populations are maintained in adult guts, given previous reports of ongoing neuronal death. Here, we confirm that despite ongoing neuronal cell loss because of apoptosis in the myenteric ganglia of the adult small intestine, total myenteric neuronal numbers remain constant. This observed neuronal homeostasis is maintained by new neurons formed in vivo from dividing precursor cells that are located within myenteric ganglia and express both Nestin and p75NTR, but not the pan-glial marker Sox10. Mutation of the phosphatase and tensin homolog gene in this pool of adult precursors leads to an increase in enteric neuronal number, resulting in ganglioneuromatosis, modeling the corresponding disorder in humans. Taken together, our results show significant turnover and neurogenesis of adult enteric neurons and provide a paradigm for understanding the enteric nervous system in health and disease.


Asunto(s)
Apoptosis , Sistema Nervioso Entérico/metabolismo , Nestina/metabolismo , Neurogénesis , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factores de Transcripción SOXE/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Nestina/genética , Receptores de Factor de Crecimiento Nervioso/genética , Factores de Transcripción SOXE/genética
5.
Gastroenterology ; 146(1): 245-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24096005

RESUMEN

BACKGROUND & AIMS: As in other tumor types, progression of pancreatic cancer may require a functionally unique population of cancer stem cells. Although such cells have been identified in many invasive cancers, it is not clear whether they emerge during early or late stages of tumorigenesis. Using mouse models and human pancreatic cancer cell lines, we investigated whether preinvasive pancreatic neoplasia contains a subpopulation of cells with distinct morphologies and cancer stem cell-like properties. METHODS: Pancreatic tissue samples were collected from the KC(Pdx1), KPC(Pdx1), and KC(iMist1) mouse models of pancreatic intraepithelial neoplasia (PanIN) and analyzed by confocal and electron microscopy, lineage tracing, and fluorescence-activated cell sorting. Subpopulations of human pancreatic ductal adenocarcinoma (PDAC) cells were similarly analyzed and also used in complementary DNA microarray analyses. RESULTS: The microtubule regulator DCLK1 marked a morphologically distinct and functionally unique population of pancreatic cancer-initiating cells. These cells displayed morphological and molecular features of gastrointestinal tuft cells. Cells that expressed DCLK1 also expressed high levels of ATAT1, HES1, HEY1, IGF1R, and ABL1, and manipulation of these pathways in PDAC cell lines inhibited their clonogenic potential. Pharmacological inhibition of γ-secretase activity reduced the abundance of these cells in murine PanIN in a manner that correlated with inhibition of PanIN progression. CONCLUSIONS: Human PDAC cells and pancreatic neoplasms in mice contain morphologically and functionally distinct subpopulations that have cancer stem cell-like properties. These populations can be identified at the earliest stages of pancreatic tumorigenesis and provide new cellular and molecular targets for pancreatic cancer treatment and/or chemoprevention.


Asunto(s)
Carcinoma in Situ/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/metabolismo , Animales , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Quinasas Similares a Doblecortina , Citometría de Flujo , Humanos , Ratones , Microscopía Electrónica , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Transducción de Señal , Células Madre/patología
6.
J Clin Invest ; 134(7)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38349762

RESUMEN

Corticosteroid treatment (CST) failure is associated with poor outcomes for patients with gastrointestinal (GI) graft-versus-host disease (GVHD). CST is intended to target the immune system, but the glucocorticoid receptor (GR) is widely expressed, including within the intestines, where its effects are poorly understood. Here, we report that corticosteroids (CS) directly targeted intestinal epithelium, potentially worsening immune-mediated GI damage. CS administered to mice in vivo and intestinal organoid cultures ex vivo reduced epithelial proliferation. Following irradiation, immediate CST mitigated GI damage but delayed treatment attenuated regeneration and exacerbated damage. In a murine steroid-refractory (SR) GVHD model, CST impaired epithelial regeneration, worsened crypt loss, and reduced intestinal stem cell (ISC) frequencies. CST also exacerbated immune-mediated damage in organoid cultures with SR, GR-deficient T cells or IFN-γ. These findings correlated with CS-dependent changes in apoptosis-related gene expression and STAT3-related epithelial proliferation. Conversely, IL-22 administration enhanced STAT3 activity and overcame CS-mediated attenuation of regeneration, reducing crypt loss and promoting ISC expansion in steroid-treated mice with GVHD. Therefore, CST has the potential to exacerbate GI damage if it fails to control the damage-inducing immune response, but this risk may be countered by strategies augmenting epithelial regeneration, thus providing a rationale for clinical approaches combining such tissue-targeted therapies with immunosuppression.


Asunto(s)
Enfermedad Injerto contra Huésped , Intestinos , Humanos , Ratones , Animales , Mucosa Intestinal/metabolismo , Corticoesteroides , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/metabolismo , Esteroides/metabolismo , Regeneración/efectos de la radiación
7.
Diabetologia ; 56(11): 2424-34, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23801221

RESUMEN

AIMS/HYPOTHESIS: Islet Schwann (glial) cells and pericytes are the microorgan's accessory cells positioned at the external and internal boundaries facing the exocrine pancreas and endothelium, respectively, adjacent to the endocrine cells. Plasticity of glial cells and pericytes is shown in the glial scar formation after injury to the central nervous system. It remains unclear whether similar reactive cellular responses occur in insulitis. We applied three-dimensional (3D) histology to perform qualitative and quantitative analyses of the islet Schwann cell network and pericytes in normal, streptozotocin-injected (positive control of gliosis) and NOD mouse models. METHODS: Vessel painting paired with immunostaining of mouse pancreatic tissue was used to reveal the islet Schwann cells and pericytes and their association with vasculature. Transparent islet specimens were prepared by optical clearing to facilitate 3D confocal microscopy for panoramic visualisation of the tissue networks. RESULTS: In-depth microscopy showed that the islet Schwann cell network extends from the peri-islet domain into the core. One week after streptozotocin injection, we observed intra-islet perivascular gliosis and an increase in pericyte density. In early/moderate insulitis in the NOD mice, perilesional gliosis occurred at the front of the lymphocytic infiltration with atypical islet Schwann cell morphologies, including excessive branching and perivascular gliosis. Meanwhile, pericytes aggregated on the walls of the feeding arteriole at the peri- and intralesional domains with a marked increase in surface marker density. CONCLUSIONS/INTERPRETATION: The reactive cellular responses demonstrate plasticity and suggest a stop-gap mechanism consisting of the Schwann cells and pericytes in association with the islet lesion and vasculature when injury occurs.


Asunto(s)
Islotes Pancreáticos/efectos de los fármacos , Pericitos/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Páncreas/efectos de los fármacos , Páncreas/patología , Pericitos/patología , Células de Schwann/patología , Estreptozocina/toxicidad
8.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G1-11, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23086917

RESUMEN

Because of the dispersed nature of nerves and blood vessels, standard histology cannot provide a global and associated observation of the enteric nervous system (ENS) and vascular network. We prepared transparent mouse intestine and combined vessel painting and three-dimensional (3-D) neurohistology for joint visualization of the ENS and vasculature. Cardiac perfusion of the fluorescent wheat germ agglutinin (vessel painting) was used to label the ileal blood vessels. The pan-neuronal marker PGP9.5, sympathetic neuronal marker tyrosine hydroxylase (TH), serotonin, and glial markers S100B and GFAP were used as the immunostaining targets of neural tissues. The fluorescently labeled specimens were immersed in the optical clearing solution to improve photon penetration for 3-D confocal microscopy. Notably, we simultaneously revealed the ileal microstructure, vasculature, and innervation with micrometer-level resolution. Four examples are given: 1) the morphology of the TH-labeled sympathetic nerves: sparse in epithelium, perivascular at the submucosa, and intraganglionic at myenteric plexus; 2) distinct patterns of the extrinsic perivascular and intrinsic pericryptic innervation at the submucosal-mucosal interface; 3) different associations of serotonin cells with the mucosal neurovascular elements in the villi and crypts; and 4) the periganglionic capillary network at the myenteric plexus and its contact with glial fibers. Our 3-D imaging approach provides a useful tool to simultaneously reveal the nerves and blood vessels in a space continuum for panoramic illustration and analysis of the neurovascular complex to better understand the intestinal physiology and diseases.


Asunto(s)
Sistema Nervioso Entérico/anatomía & histología , Intestinos/irrigación sanguínea , Intestinos/inervación , Animales , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/inervación , Capilares/anatomía & histología , Femenino , Colorantes Fluorescentes , Proteína Ácida Fibrilar de la Glía/metabolismo , Imagenología Tridimensional , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Plexo Mientérico/anatomía & histología , Fibras Nerviosas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Red Nerviosa/anatomía & histología , Neuroglía/fisiología , Flujo Sanguíneo Regional/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/metabolismo , Serotonina/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina Tiolesterasa/metabolismo
9.
Nat Commun ; 14(1): 5411, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37669929

RESUMEN

Intestinal stem cells (ISCs) maintain the epithelial lining of the intestines, but mechanisms regulating ISCs and their niche after damage remain poorly understood. Utilizing radiation injury to model intestinal pathology, we report here that the Interleukin-33 (IL-33)/ST2 axis, an immunomodulatory pathway monitored clinically as an intestinal injury biomarker, regulates intrinsic epithelial regeneration by inducing production of epidermal growth factor (EGF). Three-dimensional imaging and lineage-specific RiboTag induction within the stem cell compartment indicated that ISCs expressed IL-33 in response to radiation injury. Neighboring Paneth cells responded to IL-33 by augmenting production of EGF, which promoted ISC recovery and epithelial regeneration. These findings reveal an unknown pathway of niche regulation and crypt regeneration whereby the niche responds dynamically upon injury and the stem cells orchestrate regeneration by regulating their niche. This regenerative circuit also highlights the breadth of IL-33 activity beyond immunomodulation and the therapeutic potential of EGF administration for treatment of intestinal injury.


Asunto(s)
Interleucina-33 , Traumatismos por Radiación , Humanos , Factor de Crecimiento Epidérmico , Imagenología Tridimensional , Inmunomodulación
10.
Gastroenterology ; 137(2): 453-65, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19447107

RESUMEN

BACKGROUND & AIMS: The intrinsic opacity of mouse intestinal tissue prevents its evaluation by high-resolution, in-depth optical microscopy. Instead, intestinal tissue is usually sectioned to expose the interior domains of the mucosa and submucosa for microscopic examination. However, microtome sectioning can cause distortions and artifacts that prevent acquisition of an accurate view of the sample. We therefore attempted to develop a microtome-free 3-dimensional (3D) confocal imaging method for characterization of mouse intestine. METHODS: We applied an optical-clearing solution, FocusClear, to permeate and reduce the opacity of mouse colon and ileum. Tissues were labeled with fluorescent probes and examined by confocal microscopy with efficient fluorescence excitation and emission in the FocusClear solution. The voxel-based confocal micrographs were processed with Amira software for 3D visualization and analysis. RESULTS: Treatment of tissues with the optical-clearing solution improved photon penetration, resulting in the acquisition of images with subcellular-level resolution across the mucosa, submucosa, and muscle layers. Collectively, the acquired image stacks were processed by projection algorithms for 3D analysis of the spatial relations in villi, crypts, and connective tissues. These imaging technologies allowed for identification of spatiotemporal changes in crypt morphology of colon tissues from mice with dextran sulfate sodium-induced colitis as well as detection of transgenic fluorescent proteins expressed in the colon and ileum. CONCLUSIONS: This new optical method for penetrative imaging of mouse intestine does not require tissue sectioning and provides a useful tool for 3D presentation and analysis of diseased and transgenic intestine in an integrated fashion.


Asunto(s)
Colitis Ulcerosa/patología , Colon/ultraestructura , Íleon/ultraestructura , Interpretación de Imagen Asistida por Computador , Imagenología Tridimensional/métodos , Microscopía Confocal/instrumentación , Animales , Colon/patología , Modelos Animales de Enfermedad , Femenino , Tecnología de Fibra Óptica , Íleon/patología , Mucosa Intestinal/patología , Mucosa Intestinal/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Microscopía Confocal/métodos , Probabilidad , Distribución Aleatoria , Valores de Referencia , Sensibilidad y Especificidad
11.
Hum Reprod ; 25(3): 623-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20089521

RESUMEN

BACKGROUND: Prostasin is a glycosylphosphatidylinositol-anchored extracellular serine protease with a role in epidermal growth factor receptor (EGFR) signal modulation. EGFR signaling has been shown to be important for regulating cytotrophoblast (CT) cell proliferation in human placenta. We investigated the impact of prostasin expression regulation on this cellular function as well as the molecular mechanisms involved in human cytotrophoblastic cells. METHODS: An immortalized normal human CT cell line (B6Tert-1) was used as an in vitro cell model. Prostasin expression in B6Tert-1 cells was knocked down by transfection of a short interfering RNA. Lentivirus-mediated expression of recombinant human prostasin under tetracycline regulation was performed to obtain stable B6Tert-1 cell sublines that over-expressed prostasin. Changes in cell proliferation and EGFR signaling were evaluated by immunocytochemistry for Ki67 and western blot analysis, respectively, in B6Tert-1 cells with knocked-down or increased prostasin expression. RESULTS: Prostasin knock-down in B6Tert-1 cells resulted in inhibition of cell proliferation, in association with down-regulated EGFR protein expression (both P < 0.05 versus control) as well as reduced phosphorylation of c-raf, mitogen-activated protein kinase (MAPK) kinases (MEK1/2) and extracellular signal-regulated kinases (Erk1/2) (all P < 0.05 versus control). Over-expression of prostasin led to up-regulation of the EGFR protein, but had no effect on cell proliferation or phosphorylation of MAPK signaling molecules in the B6Tert-1 cells. CONCLUSIONS: Prostasin may regulate trophoblast cell proliferation via modulating the EGFR-MAPK signaling pathway.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptores ErbB/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Serina Endopeptidasas/fisiología , Trofoblastos/fisiología , Línea Celular , Femenino , Silenciador del Gen , Humanos , Embarazo
12.
Microvasc Res ; 80(3): 512-21, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20600164

RESUMEN

Intestinal microvasculature plays a central role in nutrient absorption and immune response against infections. Microscopic visualization of intestinal microvasculature under normal and pathological conditions such as inflammatory bowel disease is essential for understanding the pathophysiology of the disease. Despite the intensive need to characterize the intestinal microstructure and vasculature in an integrated fashion, 3-dimensional (3D) visualization of the gastrointestinal tissue is often limited by the spatial resolution of the imaging tools. In this research, we aimed to apply optical clearing to minimize the random light scattering in the mouse ileum, thereby facilitating photon penetration for high-resolution, 3D optical microscopy of the tissue network without microtome sectioning. We applied cardiac perfusion of lipophilic dialkylcarbocyanine dye DiD (1,1'-dioctadecyl-3,3,3',3'-tetramethylindodicarbocyanine perchlorate) to label the mouse blood vessels, including the intestinal microvasculature. The labeled and paraformaldehyde-fixed ileum was immersed in the aqueous optical-clearing solution to improve photon penetration. Optical clearing revealed the interior domain of the mouse ileal mucosa and submucosa, where random light scattering was suppressed and the size of the microstructure in the fixed specimen remained the same. Using fluorescent labeling, the intestinal microstructure and vasculature were simultaneously imaged by 3D confocal microscopy to allow for an integrated visualization of the tissue network with high definition. This new optical approach provides a useful tool for 3D presentation and analysis of the microvasculature for better understanding the intestinal physiology.


Asunto(s)
Íleon/irrigación sanguínea , Íleon/ultraestructura , Interpretación de Imagen Asistida por Computador , Imagenología Tridimensional , Microscopía Confocal , Microvasos/ultraestructura , Animales , Femenino , Colorantes Fluorescentes , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fosfoglicerato Quinasa/genética
13.
Prostate ; 69(16): 1790-801, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19670249

RESUMEN

BACKGROUND: Prostasin is down-regulated during inflammation and in invasive cancers, and plays a role in regulation of inflammatory gene expression and invasion. METHODS: We used the human benign prostatic hyperplasia cell line BPH-1 to investigate gene expression changes associated with siRNA-mediated loss of prostasin expression. Quantitative PCR and/or western blotting were used to evaluate the expression changes of iNOS, ICAM-1, cyclin D1, IL-6, and IL-8. Gene expression changes were also evaluated in the presence of a PAR-2 antagonist. The PC-3 human prostate cancer cell line was used for evaluation of gene expression in response to prostasin re-expression. RESULTS: Prostasin silencing in BPH-1 was associated with up-regulation of iNOS, ICAM-1, IL-6, and IL-8, and down-regulation of cyclin D1; as well as reduced proliferation and invasion. The iNOS up-regulation and cyclin D1 down-regulation associated with prostasin silencing were inhibited by a PAR-2 antagonist. Re-expression of prostasin, a serine active-site mutant, and a GPI-anchor-free mutant, in the PC-3 cells resulted in PAR-2 and cyclin D1 transcription up-regulation. Transcription up-regulation of IL-6 and IL-8 was associated with re-expression of the serine active-site mutant prostasin in the PC-3 cells. Transcription up-regulation of IL-8, but to a lesser extent, was also observed in PC-3 cells expressing the wild-type prostasin. Expression of a serine protease active prostasin, GPI-anchored or anchor-free, prevented the IL-6 induction in response to PAR-2. The GPI-anchor-free prostasin also prevented the IL-8 induction. CONCLUSIONS: Prostasin plays a negative regulatory role on PAR-2-mediated signaling in prostate epithelial cells.


Asunto(s)
Ciclina D1/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Receptor PAR-2/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Western Blotting , Línea Celular , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Reacción en Cadena de la Polimerasa , Próstata/patología , Hiperplasia Prostática/patología , ARN Interferente Pequeño/farmacología , Serina Endopeptidasas/genética , Regulación hacia Arriba/efectos de los fármacos
14.
Histochem Cell Biol ; 132(6): 639-46, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19847458

RESUMEN

Controlled invasion of the uterine wall by the trophoblast cells is pivotal for the successful pregnancy, and various kinds of protease are involved in this process. Serine protease prostasin has been shown to participate in the proteolytic activation of epithelial sodium channel as well as cleavage of epidermal growth factor receptor extracellular domain in human epithelial cells. Its physiological significance in human placentation has been suggested but not validated. In the present study, we found that prostasin was expressed at a relatively high level in human placenta trophoblasts in early pregnant weeks. In the in vitro cultured human choriocarcinomal JEG-3 cells, treatment with functional antibody against prostasin led to promotion in cell invasion capability, as well as increase in the production of MMP-2, MMP-26, TIMP-1, and TIMP-4. Our data indicated that this serine protease may function as an invasion suppressor in human trophoblast, participating in the invasion-restrictive regulation of trophoblasts to avoid their over-penetration into the uterine wall.


Asunto(s)
Coriocarcinoma/patología , Invasividad Neoplásica , Serina Endopeptidasas/fisiología , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasas de la Matriz Secretadas/biosíntesis , Embarazo , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidores Tisulares de Metaloproteinasas/biosíntesis , Trofoblastos/citología , Útero/citología , Inhibidor Tisular de Metaloproteinasa-4
15.
Biochim Biophys Acta ; 1773(7): 1133-40, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17532063

RESUMEN

Expression of prostasin in the PC-3 human prostate carcinoma cells inhibited in vitro invasion, but the molecular mechanisms are unknown. Wild-type human prostasin or a serine active-site mutant prostasin was expressed in the PC-3 cells. Molecular changes were measured at the mRNA and the protein levels. Cell signaling changes were evaluated by measuring phosphorylation of the extracellular signal-regulated kinases (Erk1/2) following epidermal growth factor (EGF) treatment of the cells. Protein expression of the EGF receptor (EGFR) was differentially down-regulated by the wild-type and the active-site mutant prostasin. The mRNA expression of EGFR and the transcription repressor SLUG was reduced in cells expressing wild-type prostasin but not the active-site mutant. Phosphorylation of Erk1/2 in response to EGF was greatly reduced by the wild-type prostasin but not by the active-site mutant. The mRNA expression of the urokinase-type plasminogen activator (uPA), the uPA receptor (uPAR), cyclooxygenase-2 (COX-2), and the inducible nitric oxide synthase (iNOS) was decreased by the wild-type and the active-site mutant prostasin. The mRNA or protein expression of granulocyte-macrophage colony-stimulating factor (GM-CSF), matriptase, and E-cadherin was greatly increased by the active-site mutant prostasin. In conclusion, prostasin expression elicits both protease-dependent and independent molecular changes in the PC-3 cells.


Asunto(s)
Línea Celular Tumoral , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/metabolismo , Sitios de Unión , Cadherinas/genética , Cadherinas/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
Cancer Res ; 77(8): 1868-1879, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28386018

RESUMEN

Nerves are a notable feature of the tumor microenvironment in some epithelial tumors, but their role in the malignant progression of pancreatic ductal adenocarcinoma (PDAC) is uncertain. Here, we identify dense innervation in the microenvironment of precancerous pancreatic lesions, known as pancreatic intraepithelial neoplasms (PanIN), and describe a unique subpopulation of neuroendocrine PanIN cells that express the neuropeptide substance P (SP) receptor neurokinin 1-R (NK1-R). Using organoid culture, we demonstrated that sensory neurons promoted the proliferation of PanIN organoids via SP-NK1-R signaling and STAT3 activation. Nerve-responsive neuroendocrine cells exerted trophic influences and potentiated global PanIN organoid growth. Sensory denervation of a genetically engineered mouse model of PDAC led to loss of STAT3 activation, a decrease in the neoplastic neuroendocrine cell population, and impaired PanIN progression to tumor. Overall, our data provide evidence that nerves of the PanIN microenvironment promote oncogenesis, likely via direct signaling to neoplastic neuroendocrine cells capable of trophic influences. These findings identify neuroepithelial cross-talk as a potential novel target in PDAC treatment. Cancer Res; 77(8); 1868-79. ©2017 AACR.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Células Neuroendocrinas/patología , Páncreas/inervación , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/patología , Células Receptoras Sensoriales/patología , Células 3T3 , Animales , Carcinogénesis , Modelos Animales de Enfermedad , Ganglios Espinales/patología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Células Neuroendocrinas/metabolismo , Páncreas/patología , Factor de Transcripción STAT3/metabolismo , Células Receptoras Sensoriales/metabolismo , Sustancia P/biosíntesis
18.
J Appl Physiol (1985) ; 117(7): 765-76, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25103977

RESUMEN

Obstructive sleep apnea causes chronic intermittent hypoxia (IH) and is associated with impaired glucose metabolism, but mechanisms are unknown. Carotid bodies orchestrate physiological responses to hypoxemia by activating the sympathetic nervous system. Therefore, we hypothesized that carotid body denervation would abolish glucose intolerance and insulin resistance induced by chronic IH. Male C57BL/6J mice underwent carotid sinus nerve dissection (CSND) or sham surgery and then were exposed to IH or intermittent air (IA) for 4 or 6 wk. Hypoxia was administered by decreasing a fraction of inspired oxygen from 20.9% to 6.5% once per minute, during the 12-h light phase (9 a.m.-9 p.m.). As expected, denervated mice exhibited blunted hypoxic ventilatory responses. In sham-operated mice, IH increased fasting blood glucose, baseline hepatic glucose output (HGO), and expression of a rate-liming hepatic enzyme of gluconeogenesis phosphoenolpyruvate carboxykinase (PEPCK), whereas the whole body glucose flux during hyperinsulinemic euglycemic clamp was not changed. IH did not affect glucose tolerance after adjustment for fasting hyperglycemia in the intraperitoneal glucose tolerance test. CSND prevented IH-induced fasting hyperglycemia and increases in baseline HGO and liver PEPCK expression. CSND trended to augment the insulin-stimulated glucose flux and enhanced liver Akt phosphorylation at both hypoxic and normoxic conditions. IH increased serum epinephrine levels and liver sympathetic innervation, and both increases were abolished by CSND. We conclude that chronic IH induces fasting hyperglycemia increasing baseline HGO via the CSN sympathetic output from carotid body chemoreceptors, but does not significantly impair whole body insulin sensitivity.


Asunto(s)
Cuerpo Carotídeo/metabolismo , Desnervación , Hiperglucemia/prevención & control , Hipoxia/complicaciones , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Hipoxia/metabolismo , Hipoxia/fisiopatología , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apnea Obstructiva del Sueño/metabolismo , Apnea Obstructiva del Sueño/fisiopatología , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología
19.
PLoS One ; 7(8): e43042, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22912784

RESUMEN

Maternal cigarette smoking has adverse effects on pregnancy outcomes. The granulocyte-macrophage colony-stimulating factor (GM-CSF) is an essential cytokine for a normal pregnancy. We investigated the impact of cigarette smoke extract (CSE) on GM-CSF expression in human cytotrophoblast cells and suggested a cellular mechanism underlying the CSE-induced GM-CSF expression. An immortalized normal human trophoblast cell line (B6Tert-1) was treated with CSE. The viability and proliferation of the CSE-treated B6Tert-1 cells were evaluated, and the expression of GM-CSF in these cells was quantified at the mRNA and the protein levels by means of reverse-transcription and quantitative polymerase chain reaction (RT-qPCR); and enzyme-linked immunosorbent assay (ELISA), respectively. Human trophoblast cells treated with CSE had an increased expression of GM-CSF at both the mRNA and the protein levels. The CSE-induced GM-CSF expression was synergistically enhanced by the addition of the proteasome inhibitor MG-132, but inhibited by AG-1478, an inhibitor of the epidermal growth factor receptor (EGFR) kinase. Furthermore, CSE treatment increased the phosphorylation of the extracellular-signal regulated kinases (ERK1/2) in the trophoblast cells. The expression of other growth factors such as heparin-binding epidermal growth factor-like growth factor (HB-EGF) and vascular endothelial growth factor (VEGF) was also evaluated. Our data suggested that cigarette smoking and proteasome inhibition synergistically up-regulate GM-CSF cytokine expression by activating the EGFR signaling pathway.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Nicotiana/química , Inhibidores de Proteasoma/farmacología , ARN Mensajero/metabolismo , Humo/efectos adversos , Trofoblastos/metabolismo , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/antagonistas & inhibidores , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leupeptinas/farmacología , Microscopía Fluorescente , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Embarazo , Quinazolinas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirfostinos/farmacología
20.
ACS Nano ; 4(10): 6278-84, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20886812

RESUMEN

In this study, we aimed to use gold nanorods (Au-NRs) as luminescent substrates for labeling of the mouse intestinal blood vessels for tissue imaging. The labeled intestine was subjected to 3-D confocal microscopy to reveal the intricate morphology of the intestinal capillaries. Using the Au-NR's unique near-infrared excitation and visible fluorescence emission, we observed low noise background compared to the tissue's high autofluorescence from blue laser excitation. We took advantage of this sharp contrast in optical properties to achieve 3-D visualization of the intestinal microstructure and vasculature with capillary-level resolution. This new optical application demonstrates the Au-NR's distinctive properties in vascular labeling and fluorescence microscopy for 3-D illustration of intestinal vasculature.


Asunto(s)
Oro/química , Intestinos/patología , Nanopartículas del Metal/química , Nanotecnología/métodos , Animales , Vasos Sanguíneos/patología , Imagenología Tridimensional , Intestinos/irrigación sanguínea , Rayos Láser , Luminiscencia , Ratones , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Nanotubos/química , Óptica y Fotónica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA