Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Nucl Med ; 59(6): 980-985, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29326360

RESUMEN

Inflammatory bowel diseases (IBDs) in humans are characterized in part by aberrant CD4-positive (CD4+) T-cell responses. Currently, identification of foci of inflammation within the gut requires invasive procedures such as colonoscopy and biopsy. Molecular imaging with antibody fragment probes could be used to noninvasively monitor cell subsets causing intestinal inflammation. Here, GK1.5 cys-diabody (cDb), an antimouse CD4 antibody fragment derived from the GK1.5 hybridoma, was used as a PET probe for CD4+ T cells in the dextran sulfate sodium (DSS) mouse model of IBD. Methods: The DSS mouse model of IBD was validated by assessing changes in CD4+ T cells in the spleen and mesenteric lymph nodes (MLNs) using flow cytometry. Furthermore, CD4+ T cell infiltration in the colons of colitic mice was evaluated using immunohistochemistry. 89Zr-labeled GK1.5 cDb was used to image distribution of CD4+ T cells in the abdominal region and lymphoid organs of mice with DSS-induced colitis. Region-of-interest analysis was performed on specific regions of the gut to quantify probe uptake. Colons, ceca, and MLNs were removed and imaged ex vivo by PET. Imaging results were confirmed by ex vivo biodistribution analysis. Results: An increased number of CD4+ T cells in the colons of colitic mice was confirmed by anti-CD4 immunohistochemistry. Increased uptake of 89Zr-maleimide-deferoxamine (malDFO)-GK1.5 cDb in the distal colon of colitic mice was visible in vivo in PET scans, and region-of-interest analysis of the distal colon confirmed increased activity in DSS mice. MLNs from colitic mice were enlarged and visible in PET images. Ex vivo scans and biodistribution confirmed higher uptake in DSS-treated colons (DSS, 1.8 ± 0.40; control, 0.45 ± 0.12 percentage injected dose [%ID] per organ, respectively), ceca (DSS, 1.1 ± 0.38; control, 0.35 ± 0.09 %ID per organ), and MLNs (DSS, 1.1 ± 0.58; control, 0.37 ± 0.25 %ID per organ). Conclusion:89Zr-malDFO-GK1.5 cDb detected CD4+ T cells in the colons, ceca, and MLNs of colitic mice and may prove useful for further investigations of CD4+ T cells in preclinical models of IBD, with potential to guide development of antibody-based imaging in human IBD.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colitis/diagnóstico por imagen , Colitis/inmunología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Animales , Colitis/patología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL
2.
PLoS Pathog ; 12(5): e1005572, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27149619

RESUMEN

Human influenza viruses replicate almost exclusively in the respiratory tract, yet infected individuals may also develop gastrointestinal symptoms, such as vomiting and diarrhea. However, the molecular mechanisms remain incompletely defined. Using an influenza mouse model, we found that influenza pulmonary infection can significantly alter the intestinal microbiota profile through a mechanism dependent on type I interferons (IFN-Is). Notably, influenza-induced IFN-Is produced in the lungs promote the depletion of obligate anaerobic bacteria and the enrichment of Proteobacteria in the gut, leading to a "dysbiotic" microenvironment. Additionally, we provide evidence that IFN-Is induced in the lungs during influenza pulmonary infection inhibit the antimicrobial and inflammatory responses in the gut during Salmonella-induced colitis, further enhancing Salmonella intestinal colonization and systemic dissemination. Thus, our studies demonstrate a systemic role for IFN-Is in regulating the host immune response in the gut during Salmonella-induced colitis and in altering the intestinal microbial balance after influenza infection.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Interferón Tipo I/inmunología , Infecciones por Orthomyxoviridae/inmunología , Salmonelosis Animal/inmunología , Animales , Coinfección/inmunología , Coinfección/microbiología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/complicaciones , Reacción en Cadena en Tiempo Real de la Polimerasa , Salmonelosis Animal/microbiología
3.
Am J Physiol Cell Physiol ; 310(7): C542-57, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26739494

RESUMEN

Given the fundamental role of ß-catenin signaling in intestinal epithelial cell proliferation and the growth-promoting function of protein kinase D1 (PKD1) in these cells, we hypothesized that PKDs mediate cross talk with ß-catenin signaling. The results presented here provide several lines of evidence supporting this hypothesis. We found that stimulation of intestinal epithelial IEC-18 cells with the G protein-coupled receptor (GPCR) agonist angiotensin II (ANG II), a potent inducer of PKD activation, promoted endogenous ß-catenin nuclear localization in a time-dependent manner. A significant increase was evident within 1 h of ANG II stimulation (P< 0.01), peaked at 4 h (P< 0.001), and declined afterwards. GPCR stimulation also induced a marked increase in ß-catenin-regulated genes and phosphorylation at Ser(552) in intestinal epithelial cells. Exposure to preferential inhibitors of the PKD family (CRT006610 or kb NB 142-70) or knockdown of the isoforms of the PKD family prevented the increase in ß-catenin nuclear localization and phosphorylation at Ser(552) in response to ANG II. GPCR stimulation also induced the formation of a complex between PKD1 and ß-catenin, as shown by coimmunoprecipitation that depended on PKD1 catalytic activation, as it was abrogated by cell treatment with PKD family inhibitors. Using transgenic mice that express elevated PKD1 protein in the intestinal epithelium, we detected a marked increase in the localization of ß-catenin in the nucleus of crypt epithelial cells in the ileum of PKD1 transgenic mice, compared with nontransgenic littermates. Collectively, our results identify a novel cross talk between PKD and ß-catenin in intestinal epithelial cells, both in vitro and in vivo.


Asunto(s)
Núcleo Celular/metabolismo , Mucosa Intestinal/metabolismo , Proteína Quinasa C/metabolismo , Receptor Cross-Talk/fisiología , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Transgénicos , Fosforilación , ARN Interferente Pequeño , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/metabolismo , Transfección
4.
PLoS One ; 7(11): e47487, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23189125

RESUMEN

BACKGROUND: Regulation of immune responses is critical for controlling inflammation and disruption of this process can lead to tissue damage. We reported that CXCL13 was induced in fallopian tube tissue following C. trachomatis infection. Here, we examined the influence of the CXCL13-CXCR5 axis in chlamydial genital infection. METHODOLOGY AND PRINCIPAL FINDINGS: Disruption of the CXCL13-CXCR5 axis by injecting anti-CXCL13 Ab to BALB/c mice or using Cxcr5-/- mice increased chronic inflammation in the upper genital tract (UGT; uterine horns and oviducts) after Chlamydia muridarum genital infection (GT). Further studies in Cxcr5-/- mice showed an elevation in bacterial burden in the GT and increased numbers of neutrophils, activated DCs and activated NKT cells early after infection. After resolution, we noted increased fibrosis and the accumulation of a variety of T cells subsets (CD4-IFNγ, CD4-IL-17, CD4-IL-10 & CD8-TNFα) in the oviducts. NKT cell depletion in vitro reduced IL-17α and various cytokines and chemokines, suggesting that activated NKT cells modulate neutrophils and DCs through cytokine/chemokine secretion. Further, chlamydial glycolipids directly activated two distinct types of NKT cell hybridomas in a cell-free CD1d presentation assay and genital infection of Cd1d-/- mice showed reduced oviduct inflammation compared to WT mice. CXCR5 involvement in pathology was also noted using single-nucleotide polymorphism analysis in C. trachomatis infected women attending a sub-fertility clinic. Women who developed tubal pathology after a C. trachomatis infection had a decrease in the frequency of CXCR5 SNP +10950 T>C (rs3922). CONCLUSIONS/SIGNIFICANCE: These experiments indicate that disruption of the CXCL13-CXCR5 axis permits increased activation of NKT cells by type I and type II glycolipids of Chlamydia muridarum and results in UGT pathology potentially through increased numbers of neutrophils and T cell subsets associated with UGT pathology. In addition, CXCR5 appears to contribute to inter-individual differences in human tubal pathology following C. trachomatis infection.


Asunto(s)
Quimiocina CXCL13/fisiología , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/patología , Chlamydia muridarum/inmunología , Células T Asesinas Naturales/inmunología , Receptores CXCR5/fisiología , Infecciones del Sistema Genital/inmunología , Infecciones del Sistema Genital/patología , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Quimiocina CXCL13/metabolismo , Infecciones por Chlamydia/genética , Estudios de Cohortes , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Linfocitos/inmunología , Ratones , Células T Asesinas Naturales/metabolismo , Polimorfismo de Nucleótido Simple , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Infecciones del Sistema Genital/genética , Enfermedades de Transmisión Sexual/genética , Enfermedades de Transmisión Sexual/inmunología , Enfermedades de Transmisión Sexual/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Población Blanca
5.
Blood ; 120(18): 3677-87, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22833548

RESUMEN

Gene therapy (GT) for adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) can provide significant long-term benefit when patients are given nonmyeloablative conditioning and ADA enzyme-replacement therapy (ERT) is withheld before autologous transplantation of γ-retroviral vector-transduced BM CD34+ cells. To determine the contributions of conditioning and discontinuation of ERT to the therapeutic effects, we analyzed these factors in Ada gene knockout mice (Ada(-/-)). Mice were transplanted with ADA-deficient marrow transduced with an ADA-expressing γ-retroviral vector without preconditioning or after 200 cGy or 900 cGy total-body irradiation and evaluated after 4 months. In all tissues analyzed, vector copy numbers (VCNs) were 100- to 1000-fold greater in mice receiving 900 cGy compared with 200 cGy (P < .05). In mice receiving 200 cGy, VCN was similar whether ERT was stopped or given for 1 or 4 months after GT. In unconditioned mice, there was decreased survival with and without ERT, and VCN was very low to undetectable. When recipients were conditioned with 200 cGy and received transduced lineage-depleted marrow, only recipients receiving ERT (1 or 4 months) had detectable vector sequences in thymocytes. In conclusion, cytoreduction is important for the engraftment of gene-transduced HSC, and short-term ERT after GT did not diminish the capacity of gene-corrected cells to engraft and persist.


Asunto(s)
Adenosina Desaminasa/uso terapéutico , Agammaglobulinemia/terapia , Trasplante de Médula Ósea/métodos , Terapia Genética/métodos , Inmunodeficiencia Combinada Grave/terapia , Acondicionamiento Pretrasplante/métodos , Adenosina Desaminasa/deficiencia , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Ratones Noqueados , Retroviridae , Transducción Genética
6.
Mol Ther ; 20(10): 1844-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22760543

RESUMEN

Arginase deficiency is characterized by hyperargininemia and infrequent episodes of hyperammonemia. Human patients suffer from neurological impairment with spasticity, loss of ambulation, seizures, and severe mental and growth retardation. In a murine model, onset of the phenotypic abnormality is heralded by weight loss beginning around day 15 with death occurring typically by postnatal day 17 with hyperargininemia and markedly elevated ammonia. The goal of this study was to address the development of a gene therapy approach for arginase deficiency beginning in the neonatal period. Lifespan extension, body weight, circulating amino acids and ammonia levels were examined as outcome parameters after gene therapy with an adeno-associated viral vector expressing arginase was administered to mice on the second day of life (DOL). One-hundred percent of untreated arginase-deficient mice died by DOL 24, whereas 89% of the adeno-associated virus (AAV)-treated arginase deficient mice have survived for >8 months. While animals at 8 months demonstrate elevated glutamine levels, ammonia is less than three times that of controls and arginine levels are normal. These studies are the first to demonstrate that AAV-based therapy for arginase deficiency is effective and supports the development of gene therapy for this and the other urea cycle disorders.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Hiperargininemia/terapia , Aminoácidos/sangre , Amoníaco/sangre , Animales , Arginasa/genética , Arginasa/metabolismo , Western Blotting , Femenino , Fertilidad/genética , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Genotipo , Glutamina/sangre , Hiperargininemia/genética , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados
7.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G356-66, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22595992

RESUMEN

We have examined the role of protein kinase D1 (PKD1) signaling in intestinal epithelial cell migration. Wounding monolayer cultures of intestinal epithelial cell line IEC-18 or IEC-6 induced rapid PKD1 activation in the cells immediately adjacent to the wound edge, as judged by immunofluorescence microscopy with an antibody that detects the phosphorylated state of PKD1 at Ser(916), an autophosphorylation site. An increase in PKD1 phosphorylation at Ser(916) was evident as early as 45 s after wounding, reached a maximum after 3 min, and persisted for ≥15 min. PKD1 autophosphorylation at Ser(916) was prevented by the PKD family inhibitors kb NB 142-70 and CRT0066101. A kb NB 142-70-sensitive increase in PKD autophosphorylation was also elicited by wounding IEC-6 cells. Using in vitro kinase assays after PKD1 immunoprecipitation, we corroborated that wounding IEC-18 cells induced rapid PKD1 catalytic activation. Further results indicate that PKD1 signaling is required to promote migration of intestinal epithelial cells into the denuded area of the wound. Specifically, treatment with kb NB 142-70 or small interfering RNAs targeting PKD1 markedly reduced wound-induced migration in IEC-18 cells. To test whether PKD1 promotes migration of intestinal epithelial cells in vivo, we used transgenic mice that express elevated PKD1 protein in the small intestinal epithelium. Enterocyte migration was markedly increased in the PKD1 transgenic mice. These results demonstrate that PKD1 activation is one of the early events initiated by wounding a monolayer of intestinal epithelial cells and indicate that PKD1 signaling promotes the migration of these cells in vitro and in vivo.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/fisiología , Proteína Quinasa C/metabolismo , Cicatrización de Heridas/fisiología , Animales , Línea Celular , Células Cultivadas , Activación Enzimática , Ratones , Ratones Transgénicos , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Pirimidinas/farmacología , Transducción de Señal
8.
Exp Hematol ; 40(1): 3-13.e3, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22001673

RESUMEN

Purine analogs such as 6-thioguanine (6TG) cause myelotoxicity upon conversion into nucleotides by hypoxanthine-guanine phosphoribosyltransferase (HPRT). Here we have developed a novel and highly efficient strategy employing 6TG as a single agent for both conditioning and in vivo chemoselection of HPRT-deficient hematopoietic stem cells. The dose-response and time course of 6TG myelotoxicity were first compared in HPRT wild-type mice and HPRT-deficient transgenic mice. Dosage and schedule parameters were optimized to employ 6TG for myelosuppressive conditioning, immediately followed by in vivo chemoselection of HPRT-deficient transgenic donor bone marrow (BM) transplanted into syngeneic HPRT wild-type recipients. At appropriate doses, 6TG induced selective myelotoxicity without any adverse effects on extrahematopoietic tissues in HPRT wild-type mice, while hematopoietic stem cells deficient in HPRT activity were highly resistant to its cytotoxic effects. Combined 6TG conditioning and post-transplantation chemoselection consistently achieved ∼95% engraftment of HPRT-deficient donor BM, with low overall toxicity. Long-term reconstitution of immunophenotypically normal BM was achieved in both primary and secondary recipients. Our results provide proof-of-concept that single-agent 6TG can be used for both myelosuppressive conditioning without requiring irradiation and for in vivo chemoselection of HPRT-deficient donor cells. Our results show that by applying the myelosuppressive effects of 6TG both before (as conditioning) and after transplantation (as chemoselection), highly efficient engraftment of HPRT-deficient hematopoietic stem cells can be achieved.


Asunto(s)
Trasplante de Médula Ósea , Médula Ósea/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Hipoxantina Fosforribosiltransferasa/deficiencia , Tioguanina/farmacología , Acondicionamiento Pretrasplante , Animales , Médula Ósea/enzimología , Relación Dosis-Respuesta a Droga , Células Madre Hematopoyéticas/metabolismo , Hipoxantina Fosforribosiltransferasa/metabolismo , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Tioguanina/administración & dosificación , Tioguanina/efectos adversos , Factores de Tiempo
9.
J Biol Chem ; 287(2): 1158-67, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22094462

RESUMEN

Here, we examined the role of the extracellular Ca(2+)-sensing receptor (CaSR) in the control of colonic epithelial cell proliferation in vivo and changes in ß-catenin triggered by CaSR stimulation in human colonic epithelial cells in vitro. The in vivo studies, using a novel Casr intestinal-specific knock-out mouse, indicate that the genetic ablation of the Casr leads to hyperproliferation of colonic epithelial cells, expansion of the proliferative zone, changes in crypt structure, and enhanced ß-catenin nuclear localization. The in vitro results indicate that stimulation of the CaSR, by Ca(2+) or by the calcimimetic R-568, produced a striking and time-dependent decrease in the phosphorylation of ß-catenin at Ser-552 and Ser-675, two amino acid residues that promote ß-catenin transcriptional activity. The reduced phosphorylation of ß-catenin coincided with a decline in its nuclear localization and a marked redistribution to the plasma membrane. Furthermore, CaSR stimulation promoted a down-regulation of ß-catenin-mediated transcriptional activation. These studies demonstrate that signaling pathways emanating from the CaSR control colonic epithelial cell proliferation in vivo and suggest that the mechanism involves regulation of ß-catenin phosphorylation.


Asunto(s)
Núcleo Celular/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Receptores Sensibles al Calcio/metabolismo , Transcripción Genética/fisiología , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Compuestos de Anilina/farmacología , Animales , Calcio/metabolismo , Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Línea Celular , Núcleo Celular/genética , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Ratones , Ratones Noqueados , Fenetilaminas , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Propilaminas , Receptores Sensibles al Calcio/genética , Transcripción Genética/efectos de los fármacos , beta Catenina/genética
10.
Ann Biomed Eng ; 39(6): 1736-44, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21380571

RESUMEN

Real-time detection of pre-atherosclerotic regions remains an unmet clinical challenge. We previously demonstrated the application of micro-electro-mechanical systems (MEMS) to detect changes in convective heat transfer in terms of sensor output voltages in the zone of flow reversal in an in vitro stenotic model. We hereby demonstrated changes in sensor output voltages in the pre-atherosclerotic regions in the New Zealand White rabbits fed on hypercholesterolemic diet (HD). After 8 weeks, we observed that mean output voltages (V(ave)) were similar in the distal aortic arch, thoracic, and abdominal aortas in the normal standard diet (ND) group, consistent with an absence of atherosclerosis. In HD group, V(ave) increased in the distal aortic arch (HD: V(ave) = 1.05 ± 0.04 V; ND: V(ave) = 0.12 ± 0.01 V, n = 3, p < 0.05) and in the thoracic aortas (HD: V(ave) = 0.72 ± 0.06 V; ND: V(ave) = 0.13 ± 0.024 V, n = 3, p < 0.05), consistent with the histological presence of pre-atherosclerosis. Despite HD diet, V (ave) magnitudes were similar to ND group in the abdominal aortas (HD: V(ave) = 0.14 ± 0.003 V; ND: V(ave) = 0.14 ± 0.004 V, n = 3), corroborating histological absence of pre-atherosclerosis. Hence, MEMS thermal sensors provide a new approach to detect changes in convective heat transfer in the pre-atherosclerotic regions.


Asunto(s)
Aorta/metabolismo , Aorta/fisiopatología , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Dieta Aterogénica , Grasas de la Dieta/efectos adversos , Calor , Conductividad Térmica , Animales , Grasas de la Dieta/administración & dosificación , Masculino , Conejos , Termogénesis
11.
J Biol Chem ; 286(1): 511-20, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21051537

RESUMEN

We examined whether protein kinase D1 (PKD1), the founding member of a new protein kinase family, plays a critical role in intestinal epithelial cell proliferation. Our results demonstrate that PKD1 activation is sustained, whereas that of PKD2 is transient in intestinal epithelial IEC-18 stimulated with the G(q)-coupled receptor agonists angiotensin II or vasopressin. PKD1 gene silencing utilizing small interfering RNAs dramatically reduced DNA synthesis and cell proliferation in IEC-18 cells stimulated with G(q)-coupled receptor agonists. To clarify the role of PKD1 in intestinal epithelial cell proliferation in vivo, we generated transgenic mice that express elevated PKD1 protein in the intestinal epithelium. Transgenic PKD1 exhibited constitutive catalytic activity and phosphorylation at the activation loop residues Ser(744) and Ser(748) and on the autophosphorylation site, Ser(916). To examine whether PKD1 expression stimulates intestinal cell proliferation, we determined the rate of crypt cell DNA synthesis by detection of 5-bromo-2-deoxyuridine incorporated into the nuclei of crypt cells of the ileum. Our results demonstrate a significant increase (p < 0.005) in DNA-synthesizing cells in the crypts of two independent lines of PKD1 transgenic mice as compared with non-transgenic littermates. Morphometric analysis showed a significant increase in the length and in the total number of cells per crypt in the transgenic PKD1 mice as compared with the non-transgenic littermates (p < 0.01). Thus, transgenic PKD1 signaling increases the number of cells per crypt by stimulating the rate of crypt cell proliferation. Collectively, our results indicate that PKD1 plays a role in promoting cell proliferation in intestinal epithelial cells both in vitro and in vivo.


Asunto(s)
ADN/biosíntesis , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Quinasas/metabolismo , Animales , Biocatálisis , Línea Celular , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/enzimología , Cinética , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Proteína Quinasa C/aislamiento & purificación , Proteína Quinasa D2 , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Proteínas Quinasas/aislamiento & purificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores Acoplados a Proteínas G/agonistas
12.
J Mol Cell Cardiol ; 47(4): 552-60, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19683723

RESUMEN

Na,K-ATPase is composed of two essential alpha- and beta-subunits, both of which have multiple isoforms. Evidence indicates that the Na,K-ATPase enzymatic activity as well as its alpha(1), alpha(3) and beta(1) isoforms are reduced in the failing human heart. The catalytic alpha-subunit is the receptor for cardiac glycosides such as digitalis, used for the treatment of congestive heart failure. The role of the Na,K-ATPase beta(1)-subunit (Na,K-beta(1)) in cardiac function is not known. We used Cre/loxP technology to inactivate the Na,K-beta(1) gene exclusively in the ventricular cardiomyocytes. Animals with homozygous Na,K-beta(1) gene excision were born at the expected Mendelian ratio, grew into adulthood, and appeared to be healthy until 10 months of age. At 13-14 months, these mice had 13% higher heart/body weight ratios, and reduced contractility as revealed by echocardiography compared to their wild-type (WT) littermates. Pressure overload by transverse aortic constriction (TAC) in younger mice, resulted in compensated hypertrophy in WT mice, but decompensation in the Na,K-beta(1) KO mice. The young KO survivors of TAC exhibited decreased contractile function and mimicked the effects of the Na,K-beta(1) KO in older mice. Further, we show that intact hearts of Na,K-beta(1) KO anesthetized mice as well as isolated cardiomyocytes were insensitive to ouabain-induced positive inotropy. This insensitivity was associated with a reduction in NCX1, one of the proteins involved in regulating cardiac contractility. In conclusion, our results demonstrate that Na,K-beta(1) plays an essential role in regulating cardiac contractility and that its loss is associated with significant pathophysiology of the heart.


Asunto(s)
Eliminación de Gen , Contracción Miocárdica/efectos de los fármacos , Miocardio/enzimología , Ouabaína/farmacología , Subunidades de Proteína/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Cardiomegalia/enzimología , Cardiomegalia/fisiopatología , Separación Celular , Pruebas de Función Cardíaca , Immunoblotting , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especificidad de Órganos/efectos de los fármacos , Presión , Intercambiador de Sodio-Calcio/metabolismo
13.
Prostate ; 68(14): 1561-9, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18663728

RESUMEN

BACKGROUND: Arginase II (AII) is involved in the polyamine synthetic pathway, and elevated levels of expression have been found in a high proportion of prostate cancer samples and patients. However, the biological function of arginase II in prostate cancer still remains to be elucidated. In this study, we utilized the TRAMP mouse prostate cancer model to better understand the contribution of AII on tumor development. METHODS: AII expression was determined in prostates from TRAMP mice at 23 weeks of age by real-time RT-PCR and Western blot analysis. Additionally, AII expression was disrupted in the TRAMP model by crossbreeding arginase II knockout (AII KO) mice with TRAMP mice in order to generate the TRAMP/AII KO line. In each group, genito-urinary (GU) tract weights were determined and a pathological evaluation of the tumors was completed. RESULTS: AII expression was only detectable in those mice without the presence of macroscopic tumors; it was also absent in the TRAMP-C2 cell line, which is characteristic of an advanced prostate tumor. Assessment of the GU weights revealed larger average GU weights in the TRAMP/AII KO mice compared to TRAMP mice. Additionally, a greater percentage of more advanced pathology was found in the TRAMP/AII KO group compared to the TRAMP cohort. CONCLUSIONS: Based on these results, AII deficiency in the TRAMP model seems to accelerate prostate tumor progression, leading to an overall more advanced cancer stage in these mice. These findings support the possibility that prostatic arginase II could be a potentially useful marker of disease progression.


Asunto(s)
Adenocarcinoma/enzimología , Arginasa/metabolismo , Neoplasias de la Próstata/enzimología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Arginasa/biosíntesis , Arginasa/genética , Western Blotting , Línea Celular Tumoral , Hiperargininemia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neoplasias Hormono-Dependientes/enzimología , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Neoplásico/química , ARN Neoplásico/genética , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Prostaglandins Other Lipid Mediat ; 84(3-4): 98-107, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17991612

RESUMEN

Cyclooxygenases (COX) regulate a variety of inflammatory diseases, including inflammatory bowel disease (IBD). While the pathological effects of COX-1 inhibition by NSAIDs on intestinal ulceration are well established, the role of COX-2 on intestinal inflammation remains under investigation. In this paper, we report a protective role for COX-2 against diet-mediated intestinal inflammation in mice. COX-2(-/-) mice fed an atherogenic diet or diet containing cholate, but not chow or fat alone, had a high mortality whereas COX-1(-/-) mice and wild-type mice were unaffected by the dietary changes. Histological analysis identified the cause of death in COX-2(-/-) mice due to severe intestinal inflammation that was surprisingly limited to the ileo-ceco-colic junction. COX-2 expression is induced in the cecum of wild-type mice fed an atherogenic diet. Our findings show that COX-2 plays an anti-inflammatory role at the ileo-ceco-colic junction in mice, and the pathology of diet-mediated intestinal inflammation in COX-2(-/-) mice offers an excellent model system to elucidate the molecular mechanisms of intestinal inflammation.


Asunto(s)
Enfermedad de Crohn/enzimología , Enfermedad de Crohn/etiología , Ciclooxigenasa 2/deficiencia , Dieta Aterogénica , Animales , Colatos/efectos adversos , Enfermedad de Crohn/mortalidad , Enfermedad de Crohn/patología , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Grasas de la Dieta/efectos adversos , Femenino , Inflamación/inducido químicamente , Intestinos/efectos de los fármacos , Intestinos/patología , Masculino , Ratones
15.
Mol Ther ; 15(7): 1390-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17505480

RESUMEN

Lentiviral vectors (LVs) are potential tools for genetic vaccination. To improve the safety of LV vaccines, we evaluated the selectivity, bio-distribution, persistence of expression, and immune potency of vesicular stomatitis virus G (VSV-G)-pseudotyped vectors transcriptionally targeted to antigen presenting cells (APCs) through a major histocompatibility complex class II (MHCII) promoter. Control vectors contained the ubiquitous cytomegalovirus (CMV) promoter. Bio-distribution studies after intravenous injections of LVs expressing green fluorescent protein (GFP) or luciferase were conducted by a combination of flow cytometry, immunofluorescence, real-time quantitative polymerase chain reaction (RT-Q-PCR) and whole-body bioluminescence analyses. GFP-expressing vectors showed selective expression in MHCII(+) cells of spleen and LV-CMV-GFP administration produced noticeable spleen inflammation, whereas LV-MHCII-GFP did not. Long-term optical imaging analyses of C57BL/6 mice injected with LV-CMV-LUC showed diminishing luciferase expression in the liver and spleen over time. Vaccination/boost with LV-CMV expressing the melanoma antigen tyrosinase-related protein 2 (TRP2) yielded dose-dependent antigen-specific CD8(+) T-cell reactivity and high protection against B16 melanoma challenge. Unexpectedly, administration of LVs containing the MHCII promoter resulted in persistence of luciferase expression and viral integration in MHCII(+) splenocytes and virtually no CD8(+) T-cell responses against TRP2. These studies reveal that APC transduction by LVs could lead to immune reactivity (LV-CMV) or persistence of transgene expression (LV-MHCII), providing a relevant paradigm for vaccination and gene replacement approaches.


Asunto(s)
Citomegalovirus/genética , Expresión Génica/genética , Vectores Genéticos/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Lentivirus/genética , Regiones Promotoras Genéticas/genética , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Vectores Genéticos/administración & dosificación , Antígenos de Histocompatibilidad Clase II/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Sensibilidad y Especificidad , Bazo/inmunología , Bazo/metabolismo , Tasa de Supervivencia , Transgenes/genética , Vacunación , Internalización del Virus
16.
Mol Ther ; 15(5): 971-80, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17375074

RESUMEN

Approaches facilitating generation of dendritic cell (DC) vaccines for clinical trials and enhancing their viability, bio-distribution, and capacity to stimulate antigen-specific immune responses are critical for immunotherapy. We programmed mouse bone marrow (BM) cells with lentiviral vectors (LV-GI4) so that they produced granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) in an autonomous manner. DC/LV-GI4 cells underwent autonomous trans-differentiation to yield typical phenotypic characteristics of DCs. DC/LV-GI4 cells that self-differentiated either ex vivo or in vivo showed persistent and robust viability and stimulated high influx of DCs into draining lymph nodes (LNs). The immunostimulatory efficacy of DC/LV-GI4 cells was evaluated using MART1 and TRP2 as co-expressed melanoma antigens. Mice vaccinated with DC/LV-GI4 cells that self-differentiated in vitro or in vivo produced potent antigen-specific responses against melanoma, which correlated with protective and long-term therapeutic anti-tumor effects. Thus, DC precursors can be genetically engineered after a single ex vivo manipulation, resulting in DC vaccines with improved activity.


Asunto(s)
Células de la Médula Ósea/inmunología , Vacunas contra el Cáncer/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Células Dendríticas/citología , Células Dendríticas/metabolismo , Citometría de Flujo , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunohistoquímica , Inmunoterapia Adoptiva/métodos , Interleucina-4/metabolismo , Lentivirus/genética , Luciferasas/genética , Luciferasas/metabolismo , Antígeno MART-1 , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Reacción en Cadena de la Polimerasa , Análisis de Supervivencia , Transfección
17.
Cancer Res ; 66(13): 6492-6, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16818619

RESUMEN

PTEN is one of the most frequently mutated tumor suppressor genes in human cancers. Germ line mutations of PTEN have been detected in three rare autosomal-dominant disorders. However, identical mutations in the PTEN gene may lead to different symptoms that have traditionally been described as different disorders, such as Cowden disease, Lhermitte-Duclos disease, and Bannayan-Zonana syndromes. This lack of genotype-phenotype correlation prompted us to directly test the possible effects of genetic background or modifier genes on PTEN-controlled tumorigenesis using genetically engineered mouse models. In this study, we generated two animal models in which either exon 5 (Pten(Delta5)) or promoter to exon 3 (Pten(-)) of the murine Pten gene were deleted and compared phenotypes associated with individual mutations on two genetic backgrounds. We found that the onset and spectrum of tumor formation depend significantly on the genetic background but less on the type of mutation generated. Our results suggest that PTEN plays a critical role in cancer development, and genetic background may influence the onset, the spectrum, and the progression of tumorigenesis caused by Pten mutation.


Asunto(s)
Neoplasias Experimentales/genética , Fosfohidrolasa PTEN/deficiencia , Alelos , Animales , Femenino , Genes Supresores de Tumor , Mutación de Línea Germinal , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fosfohidrolasa PTEN/genética
18.
Am J Physiol Gastrointest Liver Physiol ; 291(5): G792-802, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16728727

RESUMEN

In view of the importance of molecular sensing in the function of the gastrointestinal (GI) tract, we assessed whether signal transduction proteins that mediate taste signaling are expressed in cells of the human gut. Here, we demonstrated that the alpha-subunit of the taste-specific G protein gustducin (Galpha(gust)) is expressed prominently in cells of the human colon that also contain chromogranin A, an established marker of endocrine cells. Double-labeling immunofluorescence and staining of serial sections demonstrated that Galpha(gust) localized to enteroendocrine L cells that express peptide YY and glucagon-like peptide-1 in the human colonic mucosa. We also found expression of transcripts encoding human type 2 receptor (hT2R) family members, hT1R3, and Galpha(gust) in the human colon and in the human intestinal endocrine cell lines (HuTu-80 and NCI-H716 cells). Stimulation of HuTu-80 or NCI-H716 cells with the bitter-tasting compound phenylthiocarbamide, which binds hT2R38, induced a rapid increase in the intracellular Ca2+ concentration in these cells. The identification of Galpha(gust) and chemosensory receptors that perceive chemical components of ingested substances, including drugs and toxins, in open enteroendocrine L cells has important implications for understanding molecular sensing in the human GI tract and for developing novel therapeutic compounds that modify the function of these receptors in the gut.


Asunto(s)
Colon/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido YY/metabolismo , Transducina/metabolismo , Calcio/metabolismo , Línea Celular , Cromogranina A/metabolismo , Colon/citología , ADN/genética , ADN/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/metabolismo , Humanos , Inmunohistoquímica , Proteínas de Microfilamentos/metabolismo , Fenilcarbamatos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiocarbamatos/farmacología
19.
Mol Genet Metab ; 86(1-2): 233-43, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15979918

RESUMEN

Mucopolysaccharidosis I (MPS I, alpha-l-iduronidase deficiency disease) is a heritable lysosomal storage disorder involving multiple organs, including the heart. Malfunction of the heart is also a major manifestation in the mouse model of MPS I, progressing in severity from 6 to 10 months (of a 1 year life span). In comparisons of MPS I with wild-type mice, the heart was found enlarged, with thickened septal and posterior walls, primarily because of infiltration of the muscle by storage-laden cells. Heart valves were enlarged and misshapen, and contained large numbers of highly vacuolated interstitial cells. The thickened aortic wall contained vacuolated smooth muscle cells and interrupted elastic fibers. Hemodynamic measurements and echocardiography revealed reduced left ventricular function as well as mitral and aortic regurgitation. But despite these abnormalities, free-roaming MPS I mice implanted with radio telemetry devices showed surprisingly normal heart rate and blood pressure, though their electrocardiograms were abnormal. An incidental finding of the telemetry studies was a disturbed circadian rhythm in the MPS I mice. Restoration of enzyme activity in the heart of one mouse, by transplantation of retrovirally modified bone marrow, resulted in normalization of left ventricular function as well as loss of storage vacuoles in myocytes and endothelial cells, though not in valvular interstitial cells. This study demonstrates the usefulness of the mouse model for in-depth studies of the cardiovascular component of MPS I.


Asunto(s)
Modelos Animales de Enfermedad , Corazón/fisiopatología , Mucopolisacaridosis I/fisiopatología , Animales , Trasplante de Médula Ósea , Ecocardiografía , Electrocardiografía , Terapia Genética , Vectores Genéticos , Glicosaminoglicanos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis I/terapia , Retroviridae/genética
20.
J Nucl Med ; 46(1): 114-20, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632041

RESUMEN

UNLABELLED: The aim of this study was to evaluate, whether PET with (18)F-FDG and 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) may be used to monitor noninvasively the antiproliferative effects of tyrosine kinase inhibitors. METHODS: Using a high-resolution small animal scanner, we measured the effect of the ErbB-selective kinase inhibitor PKI-166 on the (18)F-FDG and (18)F-FLT uptake of ErbB1-overexpressing A431 xenograft tumors. RESULTS: Treatment with PKI-166 markedly lowered tumor (18)F-FLT uptake within 48 h of drug exposure; within 1 wk (18)F-FLT uptake decreased by 79%. (18)F-FLT uptake by the xenografts significantly correlated with the tumor proliferation index as determined by proliferating cell nuclear antigen staining (r = 0.71). Changes in (18)F-FLT uptake did not reflect inhibition of ErbB kinase activity itself but, rather, the effects of kinase inhibition on tumor cell proliferation. Tumor (18)F-FDG uptake generally paralleled the changes seen for (18)F-FLT. However, the baseline signal was significantly lower than that for (18)F-FLT. CONCLUSION: These results indicate that (18)F-FLT PET provides noninvasive, quantitative, and repeatable measurements of tumor cell proliferation during treatment with ErbB kinase inhibitors and provide a rationale for the use this technology in clinical trials of kinase inhibitors.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Carcinoma de Células Escamosas/metabolismo , Didesoxinucleósidos/farmacocinética , Fluorodesoxiglucosa F18/farmacocinética , Invasividad Neoplásica/diagnóstico por imagen , Invasividad Neoplásica/prevención & control , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID , Tomografía de Emisión de Positrones/métodos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Radiofármacos/farmacocinética , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...