Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(9): e2210836120, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36821580

RESUMEN

Defining the ontogeny of tumor-associated macrophages (TAM) is important to develop therapeutic targets for mesothelioma. We identified two distinct macrophage populations in mouse peritoneal and pleural cavities, the monocyte-derived, small peritoneal/pleural macrophages (SPM), and the tissue-resident large peritoneal/pleural macrophages (LPM). SPM rapidly increased in tumor microenvironment after tumor challenge and contributed to the vast majority of M2-like TAM. The selective depletion of M2-like TAM by conditional deletion of the Dicer1 gene in myeloid cells (D-/-) promoted tumor rejection. Sorted SPM M2-like TAM initiated tumorigenesis in vivo and in vitro, confirming their capacity to support tumor development. The transcriptomic and single-cell RNA sequencing analysis demonstrated that both SPM and LPM contributed to the tumor microenvironment by promoting the IL-2-STAT5 signaling pathway, inflammation, and epithelial-mesenchymal transition. However, while SPM preferentially activated the KRAS and TNF-α/NFkB signaling pathways, LPM activated the IFN-γ response. The importance of LPM in the immune response was confirmed by depleting LPM with intrapleural clodronate liposomes, which abrogated the antitumoral memory immunity. SPM gene signature could be identified in pleural effusion and tumor from patients with untreated mesothelioma. Five genes, TREM2, STAB1, LAIR1, GPNMB, and MARCO, could potentially be specific therapeutic targets. Accordingly, Trem2 gene deletion led to reduced SPM M2-like TAM with compensatory increase in LPM and slower tumor growth. Overall, these experiments demonstrate that SPM M2-like TAM play a key role in mesothelioma development, while LPM more specifically contribute to the immune response. Therefore, selective targeting of monocyte-derived TAM may enhance antitumor immunity through compensatory expansion of tissue-resident TAM.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Animales , Ratones , Mesotelioma Maligno/metabolismo , Mesotelioma Maligno/patología , Macrófagos Asociados a Tumores/patología , Macrófagos/metabolismo , Mesotelioma/metabolismo , Monocitos/patología , Microambiente Tumoral , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(21): e2214936120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192162

RESUMEN

Influenza A virus (IAV) enters host cells mostly through clathrin-dependent receptor-mediated endocytosis. A single bona fide entry receptor protein supporting this entry mechanism remains elusive. Here we performed proximity ligation of biotin to host cell surface proteins in the vicinity of attached trimeric hemagglutinin-HRP and characterized biotinylated targets using mass spectrometry. This approach identified transferrin receptor 1 (TfR1) as a candidate entry protein. Genetic gain-of-function and loss-of-function experiments, as well as in vitro and in vivo chemical inhibition, confirmed the functional involvement of TfR1 in IAV entry. Recycling deficient mutants of TfR1 do not support entry, indicating that TfR1 recycling is essential for this function. The binding of virions to TfR1 via sialic acids confirmed its role as a directly acting entry factor, but unexpectedly even headless TfR1 promoted IAV particle uptake in trans. TIRF microscopy localized the entering virus-like particles in the vicinity of TfR1. Our data identify TfR1 recycling as a revolving door mechanism exploited by IAV to enter host cells.


Asunto(s)
Virus de la Influenza A , Transferrina , Virus de la Influenza A/fisiología , Internalización del Virus , Endocitosis/fisiología , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo
3.
Int J Mol Sci ; 19(7)2018 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949929

RESUMEN

Malignant pleural mesothelioma (MPM) is a thoracic aggressive cancer caused by asbestos exposure, which is difficult to diagnose and treat. Here, we characterized an in vivo orthotopic xenograft model consisting of human mesothelioma cells (designed as H2052/484) derived from a pleural NCI-H2052 tumor injected in partially immunodeficient athymic mice. We assessed tumor formation and tumor-dependent patterns of inflammation. H2052/484 cells conserved their mesothelioma phenotype and most characteristics from the parental NCI-H2052 cells. After intra-thoracic injection of H2052/484 cells, thoracic tumors developed in nearly all mice (86%) within 14 days, faster than from parental NCI-H2052 cells. When the mice were euthanized, the pleural lavage fluid was examined for immune cell profiles. The pleural immune cell population increased with tumor development. Interestingly, the proportion of myeloid-derived suppressor cell and macrophage (especially CD206⁺ M2 macrophages) populations increased in the pleural fluid of mice with large mesothelioma development, as previously observed in immunocompetent mice. This reliable orthotopic model recapitulates human mesothelioma and may be used for the study of new treatment strategies.


Asunto(s)
Neoplasias Pulmonares/patología , Mesotelioma/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Líquidos Corporales/metabolismo , Carcinogénesis/patología , Recuento de Células , Línea Celular Tumoral , Supervivencia Celular , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Macrófagos/metabolismo , Mesotelioma/genética , Mesotelioma/inmunología , Mesotelioma Maligno , Ratones Desnudos
4.
Cancers (Basel) ; 15(23)2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38067281

RESUMEN

Lung cancer is the leading cause of cancer mortality worldwide, with a median survival rate at 5 years of less than 20%. While molecular mapping aids in selecting appropriate therapies, it cannot predict personalized treatment response and long-term efficacy. For addressing these challenges, there is a great need for functional tests. Within this context, we developed patient-derived spheroids (PDS) from tumor and adjacent normal tissue to biomimic the respective tissue for assessing the personalized drug treatment response in vitro. Surgically resected lung specimens were used to generate spheroids using a two-step culture procedure. Flow cytometry and immune staining enabled the characterization of different cell populations resulting from the lung samples. PDS phenotype, cell proliferation and apoptosis were evaluated. Differential gene expression between tumor and adjacent normal tissue was analyzed via RT-qPCR. PDS drug sensitivity was assessed using a cell metabolic assay in response to two chemotherapeutic drug combinations. Cellular and molecular analysis revealed the proportion of epithelial cells, fibroblasts, and immune cells in the patients' tissue samples. Subsequently, PDS models from tumor and normal lung were successfully established using the expanded epithelial cells. As a proof of concept, an analysis of the drug treatment using PDS of lung adenoid cystic carcinoma exhibited a dose-dependent effect in response to cisplatin/etoposide and cisplatin/paclitaxel. Our spheroid model of both tumor and non-tumor lung cells holds great promise for enhancing the treatment efficacy in the cancer patients.

5.
Mol Oncol ; 16(22): 3949-3974, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36221913

RESUMEN

We previously observed increased levels of adenosine-deaminase-acting-on-dsRNA (Adar)-dependent RNA editing during mesothelioma development in mice exposed to asbestos. The aim of this study was to characterize and assess the role of ADAR-dependent RNA editing in mesothelioma. We found that tumors and mesothelioma primary cultures have higher ADAR-mediated RNA editing compared to mesothelial cells. Unsupervised clustering of editing in different genomic regions revealed heterogeneity between tumor samples as well as mesothelioma primary cultures. ADAR2 expression levels are higher in BRCA1-associated protein 1 wild-type tumors, with corresponding changes in RNA editing in transcripts and 3'UTR. ADAR2 knockdown and rescue models indicated a role in cell proliferation, altered cell cycle, increased sensitivity to antifolate treatment, and type-1 interferon signaling upregulation, leading to changes in the microenvironment in vivo. Our data indicate that RNA editing contributes to mesothelioma heterogeneity and highlights an important role of ADAR2 not only in growth regulation in mesothelioma but also in chemotherapy response, in addition to regulating inflammatory response downstream of sensing nucleic acid structures.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Animales , Ratones , Edición de ARN/genética , Microambiente Tumoral/genética , Resistencia a Antineoplásicos/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Mesotelioma/genética
6.
Hum Mol Genet ; 18(3): 428-39, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19000992

RESUMEN

Previous studies have documented that the insulin-producing beta-cells of laboratory rodents are coupled by gap junction channels made solely of the connexin36 (Cx36) protein, and have shown that loss of this protein desynchronizes beta-cells, leading to secretory defects reminiscent of those observed in type 2 diabetes. Since human islets differ in several respects from those of laboratory rodents, we have now screened human pancreas, and islets isolated thereof, for expression of a variety of connexin genes, tested whether the cognate proteins form functional channels for islet cell exchanges, and assessed whether this expression changes with beta-cell function in islets of control and type 2 diabetics. Here, we show that (i) different connexin isoforms are differentially distributed in the exocrine and endocrine parts of the human pancreas; (ii) human islets express at the transcript level different connexin isoforms; (iii) the membrane of beta-cells harbors detectable levels of gap junctions made of Cx36; (iv) this protein is concentrated in lipid raft domains of the beta-cell membrane where it forms gap junctions; (v) Cx36 channels allow for the preferential exchange of cationic molecules between human beta-cells; (vi) the levels of Cx36 mRNA correlated with the expression of the insulin gene in the islets of both control and type 2 diabetics. The data show that Cx36 is a native protein of human pancreatic islets, which mediates the coupling of the insulin-producing beta-cells, and contributes to control beta-cell function by modulating gene expression.


Asunto(s)
Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Expresión Génica , Células Secretoras de Insulina/metabolismo , Insulina/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Conexinas/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Uniones Comunicantes/genética , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Páncreas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína delta-6 de Union Comunicante
7.
Oncotarget ; 12(24): 2375-2387, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34853659

RESUMEN

Advanced lung cancers and mesothelioma remain incurable diseases. Despite some promising new therapy strategies, predicting whether an individual patient will be sensitive to a given therapy is challenging. The purpose of this study is to establish and evaluate the efficiency of a three-dimensional spheroid model of human thoracic cancer in predicting the efficacy of drugs. Human mesothelioma and lung tumor spheroids were established from cell lines and primary cells derived from the patient. The growth kinetics and cell viability of microtumors were assessed using spheroid size and intracellular ATP level. The sensitivity of the mesothelioma spheroids to the cisplatin or cisplatin/pemetrexed combination was determined. We determined that studying the kinetics of the spheroid growth for 15 days after seeding 1000 cells/well in a 96-well plate was optimal. Monitoring the growth kinetic and intracellular ATP of spheroids allowed the identification of early changes in spheroid viability. Finally, we validated this model by measuring a dose-dependent reduction in the cell viability of mesothelioma H2052/484 spheroids treated with both first-line treatments, cisplatin and the cisplatin/pemetrexed combination. In conclusion, we have developed a three-dimensional spheroid model of thoracic tumor cells useful for tailoring the medical treatment to the specific characteristics of each patient.

8.
Xenotransplantation ; 17(5): 362-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20973278

RESUMEN

Embryonic stem cells (ESC) can differentiate into all cell lineages, and ESC-like cells were shown to induce hematopoietic chimerism and tolerance in allogeneic models. The aim of our study was to test the capacity of mouse ESC (mESC) to engraft in rats in a xenotransplantation setting. Forty-six rats were transplanted intravenously with 1 million mESC, without immunosuppression (group 1, n = 23) or with cyclosporine (group 2, n = 23). Three months after mESC transplantation, skin grafts were performed from allogeneic, xenogeneic identical to mESC, or xenogeneic third party donors. At day 27 post-transplant, we detected circulating mouse cells in the blood of 4/23 and 5/23 animals of group 1 and group 2, respectively. Chimerism was confirmed by PCR. We also identified long-term surviving murine cells within livers of chimeric animals. Skin grafts showed no difference in survival between allogeneic and xenogeneic donors. Transplantation of xenogeneic mouse ESC induced short-term chimerism in the blood and persistent tissue chimerism in the liver of recipient rats, but did not induce tolerance to skin grafts. Improved immunosuppressive protocols should be tested to prolong chimerism and allow tolerance.


Asunto(s)
Quimerismo , Células Madre Embrionarias/fisiología , Trasplante de Células Madre , Trasplante Heterólogo , Animales , Ciclosporina/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Supervivencia de Injerto , Sistema Hematopoyético , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Trasplante de Piel , Tolerancia al Trasplante
9.
Cell Transplant ; 18(1): 101-10, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19476213

RESUMEN

The aim of this study was to establish hepatocyte isolation in pigs, and to evaluate function of isolated hepatocytes after encapsulation, cryopreservation, and transplantation (Tx) in a mouse model of fulminant liver failure (FLF). After isolation, porcine hepatocytes were microencapsulated with alginate-poly-L-Lysine-alginate membranes and cryopreserved. In vitro, albumin production of free and encapsulated hepatocytes were measured by enzyme linked-immunoadsorbent assay. In vivo, encapsulated hepatocytes were transplanted into different groups of mice with FLF and the following experimental groups were performed: group 1, Tx of empty capsules; group 2, Tx of free primary porcine hepatocytes; group 3, Tx of fresh encapsulated porcine hepatocytes; group 4, Tx of cryopreserved encapsulated porcine hepatocytes. In vitro, fresh or cryopreserved encapsulated porcine hepatocytes showed a continuous decreasing metabolic function over 1 week (albumin and urea synthesis, drug catabolism). In vivo, groups 1 and 2 showed similar survival (18% and 25%, respectively, p > 0.05). In groups 3 and 4, Tx of fresh or cryopreserved encapsulated porcine hepatocytes significantly increased survival rate to 75% and 68%, respectively (p < 0.05). Primary porcine hepatocytes maintained metabolic functions after encapsulation and cryopreservation. In mice with FLF, Tx of encapsulated xenogeneic hepatocytes significantly improved survival. These results indicate that porcine hepatocytes can successfully be isolated, encapsulated, stored using cryopreservation, and transplanted into xenogeneic recipients with liver failure and sustain liver metabolic functions.


Asunto(s)
Criopreservación/métodos , Hepatocitos/trasplante , Fallo Hepático Agudo/terapia , Trasplante Heterólogo/métodos , Animales , Células Cultivadas , Composición de Medicamentos , Supervivencia de Injerto , Hepatocitos/metabolismo , Fallo Hepático Agudo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Porcinos
10.
J Vis Exp ; (154)2019 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-31904012

RESUMEN

Malignant pleural mesothelioma (MPM) is a rare and aggressive tumor arising in the mesothelium that covers the lungs, the heart, and the thoracic cavity. MPM development is mainly associated with asbestos. Treatments provide only modest survival since the median survival average is 9-18 months from the time of diagnosis. Therefore, more effective treatments must be identified. Most data describing new therapeutic targets were obtained from in vitro experiments and need to be validated in reliable in vivo preclinical models. This article describes one such reliable MPM orthotopic model obtained after injection of a human MPM cell line H2052/484 into the pleural cavity of immunodeficient athymic mice. Transplantation in the orthotopic site allows studying the progression of tumor in the natural in vivo environment. Positron emission tomography/computed tomography (PET/CT) molecular imaging using the clinical [18F]-2-fluoro-2-deoxy-D-glucose ([18F]FDG) radiotracer is the diagnosis method of choice for examining patients with MPM. Accordingly, [18F]FDG-PET/CT was used to longitudinally monitor the disease progression of the H2052/484 orthotopic model. This technique has a high 3R potential (Reduce the number of animals, Refine to lessen pain and discomfort, and Replace animal experimentation with alternatives) since the tumor development can be monitored non-invasively and the number of animals required could be significantly reduced. This model displays a high development rate, a rapid tumor growth, is cost-efficient and allows for rapid clinical translation. By using this orthotopic xenograft MPM model, researchers can assess biological responses of a reliable MPM model following therapeutic interventions.


Asunto(s)
Neoplasias Pulmonares/diagnóstico por imagen , Mesotelioma/diagnóstico por imagen , Neoplasias Pleurales/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Proliferación Celular , Fluorodesoxiglucosa F18/química , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mesotelioma/metabolismo , Mesotelioma/patología , Mesotelioma Maligno , Ratones Desnudos , Tamaño de los Órganos , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología
11.
Sci Rep ; 9(1): 9380, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31253838

RESUMEN

Toll-like receptor 4 (TLR4) is involved in CD4+ T lymphocyte-mediated pathologies. Here, we demonstrate that CD4+ T lymphocytes express functional TLR4 that contributes to their activation, proliferation and cytokine secretion. In addition, we demonstrate that TLR4-induced responses are mediated by macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine. We also demonstrate that MIF regulates suboptimal TCR/CD3-mediated activation of T lymphocytes. On one hand, MIF prevents excessive TCR/CD3-mediated activation of CD4+ T lymphocytes under suboptimal stimulation conditions and, on the other hand, MIF enables activated CD4+ T lymphocytes to sense their microenvironment and adapt their effector response through TLR4. Therefore, MIF appears to be a major regulator of the activation of CD4+ T lymphocytes and the intensity of their effector response. TLR4-mediated activation is thus an important process for T cell-mediated immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Regulación de la Expresión Génica , Oxidorreductasas Intramoleculares/metabolismo , Activación de Linfocitos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Receptor Toll-Like 4/genética , Animales , Biomarcadores , Inmunomodulación , Inmunofenotipificación , Lipopolisacáridos/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Receptor Toll-Like 4/metabolismo
12.
Lung Cancer ; 127: 138-145, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30642542

RESUMEN

Here we summarize the most recent update of mesothelioma research in basic science presented at the 14th iMig2018 international conference. The symposium of basic science track mainly focused on the drivers of mesothelioma initiation and progression, molecular pathogenesis, and perspectives on potential therapeutic approaches. This review covers several promising fields including strategies efficiently inhibiting YAP/TAZ functions or their critical downstream targets, heparanase inhibitors, RAN depletion, and MIF/CD74 inhibitors that may be developed as novel therapeutic approaches. In addition, targeting mesothelioma stem cells by depleting M2-polarized macrophages in tumor microenvironment or blocking Tnfsf18 (GITRL)-GITR signalling might be translated into therapeutic modalities in mesothelioma treatment.


Asunto(s)
Cooperación Internacional , Neoplasias Pulmonares/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Investigación , Animales , Antineoplásicos/uso terapéutico , Congresos como Asunto , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Mesotelioma/patología , Mesotelioma/terapia , Mesotelioma Maligno , Terapia Molecular Dirigida , Neoplasias Pleurales/patología , Neoplasias Pleurales/terapia , Opinión Pública , Microambiente Tumoral
13.
Oncogene ; 37(20): 2645-2659, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29507420

RESUMEN

Chronic exposure to intraperitoneal asbestos triggered a marked response in the mesothelium well before tumor development. Macrophages, mesothelial precursor cells, cytokines, and growth factors accumulated in the peritoneal lavage. Transcriptome profiling revealed YAP/TAZ activation in inflamed mesothelium with further activation in tumors, paralleled by increased levels of cells with nuclear YAP/TAZ. Arg1 was one of the highest upregulated genes in inflamed tissue and tumor. Inflamed tissue showed increased levels of single-nucleotide variations, with an RNA-editing signature, which were even higher in the tumor samples. Subcutaneous injection of asbestos-treated, but tumor-free mice with syngeneic mesothelioma tumor cells resulted in a significantly higher incidence of tumor growth when compared to naïve mice supporting the role of the environment in tumor progression.


Asunto(s)
Asbesto Crocidolita/efectos adversos , Redes Reguladoras de Genes , Neoplasias Pulmonares/genética , Mesotelioma/genética , Edición de ARN , Activación Transcripcional , Proteínas Adaptadoras Transductoras de Señales , Animales , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/metabolismo , Activación de Macrófagos , Mesotelioma/inducido químicamente , Mesotelioma/metabolismo , Mesotelioma Maligno , Ratones , Mutación , Fosfoproteínas , Polimorfismo de Nucleótido Simple , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
14.
Oncotarget ; 7(10): 11512-25, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26883190

RESUMEN

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine implicated in acute and chronic inflammatory diseases. MIF is overexpressed in various tumors. It displays a number of functions that provide a direct link between the process of inflammation and tumor growth. Our group recently identified the MIF-receptor CD74 as an independent prognostic factor for overall survival in patients with malignant pleural mesothelioma.In the present study, we compared the levels of expression of MIF and CD74 in different human mesothelioma cell lines and investigated their physiopathological functions in vitro and in vivo.Human mesothelioma cells expressed more CD74 and secreted less MIF than non tumoral MeT5A cells, suggesting a higher sensitivity to MIF. In mesothelioma cells, high MIF levels were associated with a high multiplication rate of cells. In vitro, reduction of MIF or CD74 levels in both mesothelioma cell lines showed that the MIF/CD74 signaling pathway promoted tumor cell proliferation and protected MPM cells from apoptosis. Finally, mesothelioma cell lines expressing high CD74 levels had a low tumorigenic potential after xenogeneic implantation in athymic nude mice.All these data highlight the complexity of the MIF/CD74 signaling pathway in the development of mesothelioma.


Asunto(s)
Antígenos CD/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias Pulmonares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurales/metabolismo , Sialiltransferasas/metabolismo , Animales , Antígenos CD/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Xenoinjertos , Humanos , Oxidorreductasas Intramoleculares/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Factores Inhibidores de la Migración de Macrófagos/genética , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Ratones Desnudos , Neoplasias Pleurales/genética , Neoplasias Pleurales/patología , Sialiltransferasas/genética , Transducción de Señal , Transfección
15.
Diabetes ; 52(2): 417-24, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12540616

RESUMEN

Cx36 is the predominant connexin isoform expressed by pancreatic beta-cells. However, little is known about the role of this protein in the functioning of insulin-secreting cells. To address this question, we searched for a cell line expressing Cx36 and having glucose-induced insulin secretion comparable to that of primary beta-cells. By evaluating Cx36 expression in MIN6, betaTC3, RIN2A, INS1, and HIT cell lines, which differ in their sensitivity to glucose, we found that wild-type MIN6 cells fit these requirements. Therefore, we stably transfected MIN6 cells with a cDNA coding for a Cx36 antisense sequence to study the role of Cx36 in these cells. Independent clones of MIN6 cells were obtained that had a markedly reduced Cx36 expression. Loss of Cx36 decreased functional gap junctional conductance in these clones. This alteration impaired the synchronization of glucose-induced [Ca(2+)](i) oscillations and insulin secretion in response to glucose, to secretagogues that increase [cAMP](i), and to depolarizing conditions. These data provide the first evidence that Cx36-made channels 1) mediate functional coupling in MIN6 cells, 2) provide for synchronous [Ca(2+)](i) oscillations, and 3) are necessary for proper insulin secretion in response to metabolizable and nonmetabolizable secretagogues.


Asunto(s)
Señalización del Calcio/fisiología , Conexinas/genética , Insulina/metabolismo , Anfotericina B/farmacología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Conductividad Eléctrica , Electrofisiología/métodos , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/fisiología , Regulación de la Expresión Génica/fisiología , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína delta-6 de Union Comunicante
16.
PLoS One ; 6(9): e25442, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21980458

RESUMEN

BACKGROUND: Cytokine administration is a potential therapy for acute liver failure by reducing inflammatory responses and favour hepatocyte regeneration. The aim of this study was to evaluate the role of interleukin-1 receptor antagonist (IL-1ra) during liver regeneration and to study the effect of a recombinant human IL-1ra on liver regeneration. METHODS: We performed 70%-hepatectomy in wild type (WT) mice, IL-1ra knock-out (KO) mice and in WT mice treated by anakinra. We analyzed liver regeneration at regular intervals by measuring the blood levels of cytokines, the hepatocyte proliferation by bromodeoxyuridin (BrdU) incorporation, proliferating cell nuclear antigen (PCNA) and Cyclin D1 expression. The effect of anakinra on hepatocyte proliferation was also tested in vitro using human hepatocytes. RESULTS: At 24h and at 48 h after hepatectomy, IL-1ra KO mice had significantly higher levels of pro-inflammatory cytokines (IL-6, IL-1ß and MCP-1) and a reduced and delayed hepatocyte proliferation measured by BrdU incorporation, PCNA and Cyclin D1 protein levels, when compared to WT mice. IGFBP-1 and C/EBPß expression was significantly decreased in IL-1ra KO compared to WT mice. WT mice treated with anakinra showed significantly decreased levels of IL-6 and significantly higher hepatocyte proliferation at 24h compared to untreated WT mice. In vitro, primary human hepatocytes treated with anakinra showed significantly higher proliferation at 24h compared to hepatocytes without treatment. CONCLUSION: IL1ra modulates the early phase of liver regeneration by decreasing the inflammatory stress and accelerating the entry of hepatocytes in proliferation. IL1ra might be a therapeutic target to improve hepatocyte proliferation.


Asunto(s)
Hepatectomía , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Regeneración Hepática/efectos de los fármacos , Alanina Transaminasa/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Proteína Antagonista del Receptor de Interleucina 1/deficiencia , Proteína Antagonista del Receptor de Interleucina 1/genética , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Regeneración Hepática/genética , Masculino , Ratones , Factores de Tiempo
17.
Cell Transplant ; 20(11-12): 1791-803, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21396154

RESUMEN

The aim of this study was to evaluate the effects of intraperitoneal transplantation of encapsulated human hepatocytes on liver metabolism and regeneration of mice with acute liver failure. Primary human hepatocytes were immortalized using lentiviral vectors coding for antiapoptotic genes and microencapsulated using alginate-polylysine polymers. In vitro, immortalized human hepatocytes showed low, but stable, synthetic and catabolitic functions over time, when compared to primary hepatocytes. In vivo, mice with acute liver failure and transplanted with encapsulated immortalized human hepatocytes had a significantly improved survival and biochemical profile, compared to mice transplanted with empty capsules. Serum levels of cytokines implicated in liver regeneration were lower in mice transplanted with hepatocytes compared to mice receiving empty capsules. This decrease was significant for IL-6 and HGF at 3 h. Measurement of liver regeneration showed no significant difference between mice transplanted with hepatocytes compared to control groups. Intraperitoneal transplantation of encapsulated immortalized hepatocytes significantly improved survival of mice with acute liver failure by providing metabolic support and without modifying liver regeneration. The lower levels of cytokines implicated in liver regeneration suggest that the metabolic support provided by the encapsulated hepatocytes reduced the inflammatory stress on the liver and herein decreased the regenerative trigger on residual hepatocytes. These data emphasize that metabolic function and regeneration of hepatocytes are two distinct aspects that need to be studied and approached separately during acute liver failure.


Asunto(s)
Hepatocitos/citología , Fallo Hepático Agudo/terapia , Regeneración Hepática , Animales , Aspartato Aminotransferasas/sangre , Bilirrubina/sangre , Cápsulas/química , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citocinas/sangre , Factor de Crecimiento de Hepatocito/sangre , Hepatocitos/trasplante , Humanos , Interleucina-6/sangre , Hígado/fisiología , Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/mortalidad , Masculino , Ratones , Ratones Endogámicos C57BL , Albúmina Sérica/metabolismo , Tasa de Supervivencia , Trasplante Heterólogo
18.
Diabetes ; 60(9): 2331-40, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752956

RESUMEN

OBJECTIVE: Streptozotocin (STZ) is the most widely used diabetogenic agent in animal models of islet transplantation. However, the immunomodifying effects of STZ and the ensuing hyperglycemia on lymphocyte subsets, particularly on T regulatory cells (Tregs), remain poorly understood. RESEARCH DESIGN AND METHODS: This study evaluated how STZ-induced diabetes affects adaptive immunity and the consequences thereof on allograft rejection in murine models of islet and skin transplantation. The respective toxicity of STZ and hyperglycemia on lymphocyte subsets was tested in vitro. The effect of hyperglycemia was assessed independently of STZ in vivo by the removal of transplanted syngeneic islets, using an insulin pump, and with rat insulin promoter diphtheria toxin receptor transgenic mice. RESULTS: Early lymphopenia in both blood and spleen was demonstrated after STZ administration. Direct toxicity of STZ on lymphocytes, particularly on CD8(+) cells and B cells, was shown in vitro. Hyperglycemia also correlated with blood and spleen lymphopenia in vivo but was not lymphotoxic in vitro. Independently of hyperglycemia, STZ led to a relative increase of Tregs in vivo, with the latter retaining their suppressive capacity in vitro. The higher frequency of Tregs was associated with Treg proliferation in the blood, but not in the spleen, and higher blood levels of transforming growth factor-ß. Finally, STZ administration delayed islet and skin allograft rejection compared with naive mice. CONCLUSIONS: These data highlight the direct and indirect immunosuppressive effects of STZ and acute hyperglycemia, respectively. Thus, these results have important implications for the future development of tolerance-based protocols and their translation from the laboratory to the clinic.


Asunto(s)
Inmunidad Adaptativa/inmunología , Diabetes Mellitus Experimental/inmunología , Tolerancia Inmunológica/inmunología , Linfopenia/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Diabetes Mellitus Experimental/complicaciones , Linfopenia/etiología , Ratones , Ratas , Bazo/inmunología , Estreptozocina
19.
J Endocrinol ; 206(3): 297-306, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20566490

RESUMEN

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine produced by many cells and tissues including pancreatic beta-cells, liver, skeletal muscle, and adipocytes. This study investigates the potential role of MIF in carbohydrate homeostasis in a physiological setting outside of severe inflammation, utilizing Mif knockout (MIF-/-) mice. Compared with wild-type (WT) mice, MIF-/- mice had a lower body weight, from birth until 4 months of age, but subsequently gained weight faster, resulting in a higher body weight at 12 months of age. The lower weight in young mice was related to a higher energy expenditure, and the higher weight in older mice was related to an increased food intake and a higher fat mass. Fasting blood insulin level was higher in MIF-/- mice compared with WT mice at any age. After i.p. glucose injection, the elevation of blood insulin level was higher in MIF-/- mice compared with WT mice, at 2 months of age, but was lower in 12-month-old MIF-/- mice. As a result, the glucose clearance during intraperitoneal glucose tolerance tests was higher in MIF-/- mice compared with WT mice until 4 months of age, and was lower in 12-month-old MIF-/- mice. Insulin resistance was estimated (euglycemic-hyperinsulinemic clamp tests), and the phosphorylation activity of AKT was similar in MIF-/- mice and WT mice. In conclusion, this mouse model provides evidence for the role of MIF in the control of glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Homeostasis/fisiología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Western Blotting , Composición Corporal/fisiología , Peso Corporal/fisiología , Metabolismo Energético/fisiología , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Leptina/sangre , Lípidos/sangre , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas
20.
PLoS One ; 5(4): e10352, 2010 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-20436684

RESUMEN

BACKGROUND: Anti-CD154 (MR1) monoclonal antibody (mAb) and rapamycin (RAPA) treatment both improve survival of rat-to-mouse islet xenograft. The present study investigated the effect of combined RAPA/MR1 treatment on rat-to-mouse islet xenograft survival and analyzed the role of CD4(+)CD25(+)Foxp3(+) T regulatory cells (Treg) in the induction and maintenance of the ensuing tolerance. METHODOLOGY/PRINCIPAL FINDINGS: C57BL/6 mice were treated with MR1/RAPA and received additional monoclonal anti-IL2 mAb or anti CD25 mAb either early (0-28 d) or late (100-128 d) post-transplantation. Treg were characterised in the blood, spleen, draining lymph nodes and within the graft of tolerant and rejecting mice by flow cytometry and immunohistochemistry. Fourteen days of RAPA/MR1 combination therapy allowed indefinite islet graft survival in >80% of the mice. Additional administration of anti-IL-2 mAb or depleting anti-CD25 mAb at the time of transplantation resulted in rejection (100% and 89% respectively), whereas administration at 100 days post transplantation lead to lower rejection rates (25% and 40% respectively). Tolerant mice showed an increase of Treg within the graft and in draining lymph nodes early post transplantation, whereas 100 days post transplantation no significant increase of Treg was observed. Rejecting mice showed a transient increase of Treg in the xenograft and secondary lymphoid organs, which disappeared within 7 days after rejection. CONCLUSIONS/SIGNIFICANCES: These results suggest a critical role for Treg in the induction phase of tolerance early after islet xenotransplantation. These encouraging data support the need of developing further Treg therapy for overcoming the species barrier in xenotransplantation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Ligando de CD40/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Trasplante de Islotes Pancreáticos/inmunología , Sirolimus/farmacología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratas , Sirolimus/uso terapéutico , Factores de Tiempo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA