Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 294(52): 19877-19888, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31712309

RESUMEN

Bone morphogenetic protein 10 (BMP10) is a cardiac peptide growth factor belonging to the transforming growth factor ß superfamily that critically controls cardiovascular development, growth, and maturation. It has been shown that BMP10 elicits its intracellular signaling through a receptor complex of activin receptor-like kinase 1 with morphogenetic protein receptor type II or activin receptor type 2A. Previously, we generated and characterized a transgenic mouse line expressing BMP10 from the α-myosin heavy chain gene promoter and found that these mice have normal cardiac hypertrophic responses to both physiological and pathological stimuli. In this study, we report that these transgenic mice exhibit significantly reduced levels of cardiomyocyte apoptosis and cardiac fibrosis in response to a prolonged administration of the ß-adrenoreceptor agonist isoproterenol. We further confirmed this cardioprotective function with a newly generated conditional Bmp10 transgenic mouse line, in which Bmp10 was activated in adult hearts by tamoxifen. Moreover, the intraperitoneal administration of recombinant human BMP10 was found to effectively protect hearts from injury, suggesting potential therapeutic utility of using BMP10 to prevent heart failure. Gene profiling and biochemical analyses indicated that BMP10 activates the SMAD-mediated canonical pathway and, unexpectedly, also the signal transducer and activator of transcription 3 (STAT3)-mediated signaling pathway both in vivo and in vitro Additional findings further supported the notion that BMP10's cardioprotective function likely is due to its dual activation of SMAD- and STAT3-regulated signaling pathways, promoting cardiomyocyte survival and suppressing cardiac fibrosis.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Smad/metabolismo , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Morfogenéticas Óseas/genética , Matriz Extracelular/metabolismo , Corazón/efectos de los fármacos , Humanos , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos
2.
Acta Pharmacol Sin ; 41(11): 1416-1426, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32973325

RESUMEN

Immunotherapies for cancers may cause severe and life-threatening cardiotoxicities. The underlying mechanisms are complex and largely elusive. Currently, there are several ongoing clinical trials based on the use of activated invariant natural killer T (iNKT) cells. The potential cardiotoxicity commonly associated with this particular immunotherapy has yet been carefully evaluated. The present study aims to determine the effect of activated iNKT cells on normal and ß-adrenergic agonist (isoproterenol, ISO)-stimulated hearts. Mice were treated with iNKT stimulants, α-galactosylceramide (αGC) or its analog OCH, respectively, to determine their effect on ISO-induced cardiac injury. We showed that administration of αGC (activating both T helper type 1 (Th1)- and T helper type 2 (Th2)-liked iNKT cells) significantly accelerated the progressive cardiac injury, leading to enhanced cardiac hypertrophy and cardiac fibrosis with prominent increases in collagen deposition and TGF-ß1, IL-6, and alpha smooth muscle actin expression. In contrast to αGC, OCH (mainly activating Th2-liked iNKT cells) significantly attenuated the progression of cardiac injury and cardiac inflammation induced by repeated infusion of ISO. Flow cytometry analysis revealed that αGC promoted inflammatory macrophage infiltration in the heart, while OCH was able to restrain the infiltration. In vitro coculture of αGC- or OCH-pretreated primary peritoneal macrophages with primary cardiac fibroblasts confirmed the profibrotic effect of αGC and the antifibrotic effect of OCH. Our results demonstrate that activating both Th1- and Th2-liked iNKT cells is cardiotoxic, while activating Th2-liked iNKT cells is likely cardiac protective, which has implied key differences among subpopulations of iNKT cells in their response to cardiac pathological stimulation.


Asunto(s)
Cardiomegalia/etiología , Cardiotónicos/uso terapéutico , Galactosilceramidas/efectos adversos , Glucolípidos/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Células T Asesinas Naturales/efectos de los fármacos , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Citocinas/metabolismo , Fibrosis , Inflamación/prevención & control , Isoproterenol , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Células T Asesinas Naturales/clasificación
5.
Lab Invest ; 94(7): 706-15, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24862966

RESUMEN

Tumor-associated macrophages are a prominent component of lung cancer stroma and contribute to tumor progression. The protein V-set and Ig domain-containing 4 (VSIG4), a novel B7 family-related macrophage protein that has the capacity to inhibit T-cell activation, has a potential role in the development of lung cancer. In this study, 10 human non-small-cell lung cancer specimens were collected and immunohistochemically analyzed for VSIG4 expression. Results showed massive VSIG4(+) cell infiltration throughout the samples. Immunofluorescent double staining showed that VSIG4 was present on CD68(+) macrophages, but absent from CD3(+) T cells, CD31(+) endothelial cells, and CK-18(+) epithelial cells. Moreover, VSIG4 was coexpressed on B7-H1(+) and B7-H3(+) cells in these tumor specimens. Transfection of the VSIG4 gene into 293FT cells demonstrated that the VSIG4 signal could inhibit cocultured CD4(+) and CD8(+) T-cell proliferation and cytokine (IL-2 and IFN-γ) production in vitro. Interestingly, in a murine tumor model induced by Lewis lung carcinoma cell line, we found that tumors grown in VSIG4-deficient (VSIG4(-/-)) mice were significantly smaller than those found in wild-type littermates. All of these results demonstrate that macrophage-associated VSIG4 is an activator that facilitates lung carcinoma development. Specific targeting of VSIG4 may prove to be a novel, efficacious strategy for the treatment of this carcinoma.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Receptores de Complemento/biosíntesis , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos B7/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Células HEK293 , Humanos , Inmunohistoquímica , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Complemento/genética
6.
Gut ; 62(8): 1204-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22637698

RESUMEN

OBJECTIVES: Fulminant viral hepatitis (FH) remains a serious clinical problem for which the underlying pathogenesis remains unclear. The B and T lymphocyte attenuator (BTLA) is an immunoglobulin-domain-containing protein that has the capacity to maintain peripheral tolerance and limit immunopathological damage during immune responses. However, its precise role in FH has yet to be investigated. DESIGN: BTLA-deficient (BTLA-/-) mice and their wild-type littermates were infected with murine hepatitis virus strain-3 (MHV-3), and the levels of tissue damage, cell apoptosis, serum liver enzymes, fibrinogen-like protein 2 (FGL2) and cytokine production were measured and compared. Survival rate was studied after MHV-3 infection with or without adoptive transferring macrophages. RESULTS: FGL2 production, liver and spleen damage, and mortality were significantly reduced in BTLA-/- mice infected with MHV-3. This effect is due to rapid, TRAIL (TNF-related apoptosis-inducing ligand)-dependent apoptosis of MHV-3-infected macrophages in BTLA-/- mice. The early loss of macrophages resulted in reduced pathogenic tumour necrosis factor α (TNFα) and FGL2 levels and lower viral titres. The importance of TNFα in MHV-3-induced pathology was demonstrated by increased mortality in TNFα-treated MHV-3-infected BTLA-/- mice, whereas TNFα-/- mice were resistant to the infection. Moreover, adoptively transferring macrophages to BTLA-/- mice caused sensitisation, whereas blocking BTLA protected wild-type mice from virus-induced FH mortality. CONCLUSIONS: BTLA promotes the pathogenesis of virus-induced FH by enhancing macrophage viability and function. Targeting BTLA may be a novel strategy for the treatment of FH.


Asunto(s)
Infecciones por Coronavirus/inmunología , Hepatitis Viral Animal/inmunología , Macrófagos/inmunología , Virus de la Hepatitis Murina , Receptores Inmunológicos/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/prevención & control , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/prevención & control , Macrófagos/patología , Macrófagos/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
7.
Inflammation ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38913145

RESUMEN

It has recently become more recognized that renal diseases in adults can originate from adverse intrauterine (maternal) environmental exposures. Previously, we found that prenatal lipopolysaccharide (LPS) exposure can result in chronic renal inflammation, which leads to renal damage in older offspring rats. To test whether prenatal inflammatory exposure predisposes offspring to renal damage, a mouse model of oral adenine consumption-induced chronic kidney disease (CKD) was applied to offspring from prenatal LPS-treated mothers (offspring-pLPS) and age-matched control offspring of prenatal saline-treated mothers (offspring-pSaline). We found that offspring-pLPS mice presented with more severe renal collagen deposition and renal dysfunction after 4 weeks of adenine consumption than sex- and treatment-matched offspring-pSaline controls. To illustrate the underlying molecular mechanism, we subjected offspring-pLPS and offspring-pSaline kidneys to genome-wide transcriptomic analysis. Bioinformatic analysis of the sequencing data, together with further experimental confirmation, revealed a strong activation of the PERK-eIF2α-ATF4-mediated unfolded protein response (UPR) in offspring-pLPS kidneys, which likely contributed to the CKD predisposition seen in offspring-pLPS mice. More importantly, the specific eIF2α-ATF4 signaling inhibitor ISIRB was able to prevent adenine-induced CKD in the offspring-pLPS mice. Our findings suggest that the eIF2α-ATF4-mediated UPR, but not PERK, is likely the major disease-causing pathway in prenatal inflammatory exposure-induced CKD predisposition. Our study also suggests that targeting this signaling pathway is a potentially promising approach for CKD treatment.

8.
Inflammation ; 47(2): 789-806, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38446361

RESUMEN

Altered cardiac innate immunity is highly associated with the progression of cardiac disease states and heart failure. S100A8/A9 is an important component of damage-associated molecular patterns (DAMPs) that is critically involved in the pathogenesis of heart failure, thus considered a promising target for pharmacological intervention. In the current study, initially, we validated the role of S100A8/A9 in contributing to cardiac injury and heart failure via the overactivation of the ß-adrenergic pathway and tested the potential use of paquinimod as a pharmacological intervention of S100A8/A9 activation in preventing cardiac dysfunction, collagen deposition, inflammation, and immune cell infiltration in ß-adrenergic overactivation-mediated heart failure. This finding was further confirmed by the cardiomyocyte-specific silencing of S100A9 via the use of the adeno-associated virus (AAV) 9-mediated short hairpin RNA (shRNA) gene silencing system. Most importantly, in the assessment of the underlying cellular mechanism by which activated S100A8/A9 cause aggravated progression of cardiac fibrosis and heart failure, we discovered that the activated S100A8/A9 can promote fibroblast-macrophage interaction, independent of inflammation, which is likely a key mechanism leading to the enhanced collagen production. Our results revealed that targeting S100A9 provides dual beneficial effects, which is not only a strategy to counteract cardiac inflammation but also preclude cardiac fibroblast-macrophage interactions. The findings of this study also indicate that targeting S100A9 could be a promising strategy for addressing cardiac fibrosis, potentially leading to future drug development.


Asunto(s)
Calgranulina B , Miocitos Cardíacos , Animales , Ratones , Agonistas Adrenérgicos beta/farmacología , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Calgranulina B/genética , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Fibrosis , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Inflamación/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología
9.
Cell Physiol Biochem ; 32(4): 814-26, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080960

RESUMEN

BACKGROUND: Endothelial progenitor cells (EPCs) derived from the bone marrow (BM) play a key role in the homeostasis of vascular repair by enhanced reendothelialization. Panax notoginseng saponins (PNS), a highly valued traditional Chinese medicine, has been shown to reduce morbidity and mortality from coronary artery disease. The present research was designed to explore the contribution of progenitor cells to the progression of atherosclerotic plaques and the possible modulatory role of PNS in this process. METHODS: PNS (60 or 120 mg/kg via intraperitoneal injection) was administered over 8 weeks in apolipoprotein E knockout mice on an atherogenic diet. The sizes and histochemical alteration of atherosclerotic lesions and numbers of EPCs in BM and peripheral blood were analyzed. The expression of chemokine stromal cell-derived factor 1α (SDF-1α) and its receptor, CXCR4, was monitored as well. RESULTS: PNS significantly reduced the lesion area and intima-to-media ratio compared to vehicle treatment. PNS also augmented endothelialization and reduced the smooth muscle cell (SMCs) content of the lesions. The number of c-kit and sca-1 double-positive progenitor cells and flk-1 and sca-1 double-positive progenitor cells were significantly increased in the BM and the peripheral blood of the PNS-treated groups. PNS treatment increased the plasma levels of SDF-1α and SCF as well as the BM levels of matrix metalloproteinase-9 (MMP-9). Moreover, the mRNA levels of SDF-1α and protein levels of CXCR4 were both increased in the BM of mice treated with PNS, while SDF-1α expression decreased. CONCLUSION: PNS reduce the size of atherosclerotic plaques, and this effect appears to involve progenitor cell mobilization. SDF-1α-CXCR4 interactions and the possible modulatory role of PNS in this process may contribute to the increased progenitor cell mobilization.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/tratamiento farmacológico , Células Endoteliales/citología , Panax notoginseng/química , Saponinas/farmacología , Saponinas/uso terapéutico , Células Madre/efectos de los fármacos , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Citometría de Flujo , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saponinas/química , Células Madre/citología
10.
Pathol Int ; 62(10): 665-74, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23005593

RESUMEN

Hepatitis B virus (HBV) is a major public health problem, and HBV-related acute-on-chronic liver failure (HBV-ACLF) has an extremely poor prognosis due to a lack of effective treatments. B7-H3 and B7-H4 are two novel members of the B7 superfamily that are actively involved in regulating the pathogenesis of infectious diseases. However, the intrahepatic expression of both members in HBV-ACLF patients has yet to be described. In this study, we analyzed the expression of B7-H3 and B7-H4 in HBV-ACLF biopsies by immunohistochemistry. Our results showed that both members were observed in all HBV-ACLF samples, and their expression was chiefly observed on infiltrating inflammatory cells and the damaged bile ducts. Immunofluorescence double staining showed that B7-H4 was expressed chiefly on CD3(+) T cells, CD68(+) macrophages, CK-18(+) bile ducts, and CD31(+) endothelial cells, while B7-H3 was found on all cell types detected. The expression of the programmed death (PD)-1 ligands, PD-L1 and PD-L2, was also detected in these liver tissues and they were found to be co-expressed with B7-H3 and B7-H4. These results suggest that the B7-family signaling is most likely to affect the pathogenesis of this disease, and a clear understanding of their functional roles may further elucidate the disease process.


Asunto(s)
Antígenos B7/metabolismo , Enfermedad Hepática en Estado Terminal/metabolismo , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/metabolismo , Fallo Hepático Agudo/metabolismo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Adulto , Anciano , Antígeno B7-H1/metabolismo , Conductos Biliares , Biomarcadores/metabolismo , Biopsia , Progresión de la Enfermedad , Enfermedad Hepática en Estado Terminal/inmunología , Enfermedad Hepática en Estado Terminal/virología , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Humanos , Inmunohistoquímica , Hígado/metabolismo , Fallo Hepático Agudo/inmunología , Fallo Hepático Agudo/virología , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Linfocitos T/inmunología
11.
Eur J Pharmacol ; 927: 175022, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35569549

RESUMEN

Phillyrin, a well-known natural compound from the dried fruits of Forsythia suspensa (Thunb.) Vahl., has shown anti-inflammatory, antioxidant and anti-virus activities as well as renal protective effects on diabetic nephropathy. In this study, we investigated whether phillyrin attenuated cardiac hypertrophy induced by catecholamine in vivo and in vitro, and explored the underlying mechanisms. Cardiac hypertrophy was induced in C57BL/6 mice by subcutaneous injection of norepinephrine (NE, a key catecholamine), and in rat cardiomyoblasts (H9c2) by stimulation with NE in vitro. Our results showed that administration of phillyrin (100 mg/kg, i.p. for 15 days) significantly improved cardiac function, histopathological changes, cardiac hypertrophy and decreased the upregulated hypertrophic markers (ANP, BNP, and ß-MHC). Moreover, treatment with phillyrin obviously reduced the infiltration of the CD68 positive macrophages and the mRNA expression of proinflammatory genes (IL-1ß, IL-6, and TNF-α) in left ventricular tissue. In addition, treatment with phillyrin markedly inhibited the phosphorylation of p38 MAPK, ERK1/2, AKT, and NF-κB p65 in heart tissues. Furthermore, in NE-treated H9c2 cells, pretreatment with phillyrin clearly attenuated cardiomyocyte hypertrophy, reduced ROS production and inhibited the phosphorylation of p38 MAPK, ERK1/2, AKT, and NF-κB p65 in vitro. Collectively, our results demonstrate that phillyrin effectively alleviates NE-induced cardiac hypertrophy and inflammatory response by suppressing p38 MAPK/ERK1/2 and AKT/NF-κB signaling pathways.


Asunto(s)
FN-kappa B , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Glucósidos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Norepinefrina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Front Cell Dev Biol ; 9: 668659, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34222237

RESUMEN

RNA binding proteins (RBPs) have a broad biological and physiological function and are critical in regulating pre-mRNA posttranscriptional processing, intracellular migration, and mRNA stability. QKI, also known as Quaking, is a member of the signal transduction and activation of RNA (STAR) family, which also belongs to the heterogeneous nuclear ribonucleoprotein K- (hnRNP K-) homology domain protein family. There are three major alternatively spliced isoforms, QKI-5, QKI-6, and QKI-7, differing in carboxy-terminal domains. They share a common RNA binding property, but each isoform can regulate pre-mRNA splicing, transportation or stability differently in a unique cell type-specific manner. Previously, QKI has been known for its important role in contributing to neurological disorders. A series of recent work has further demonstrated that QKI has important roles in much broader biological systems, such as cardiovascular development, monocyte to macrophage differentiation, bone metabolism, and cancer progression. In this mini-review, we will focus on discussing the emerging roles of QKI in regulating cardiac and vascular development and function and its potential link to cardiovascular pathophysiology.

13.
Nat Commun ; 12(1): 89, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397958

RESUMEN

The RNA-binding protein QKI belongs to the hnRNP K-homology domain protein family, a well-known regulator of pre-mRNA alternative splicing and is associated with several neurodevelopmental disorders. Qki is found highly expressed in developing and adult hearts. By employing the human embryonic stem cell (hESC) to cardiomyocyte differentiation system and generating QKI-deficient hESCs (hESCs-QKIdel) using CRISPR/Cas9 gene editing technology, we analyze the physiological role of QKI in cardiomyocyte differentiation, maturation, and contractile function. hESCs-QKIdel largely maintain normal pluripotency and normal differentiation potential for the generation of early cardiogenic progenitors, but they fail to transition into functional cardiomyocytes. In this work, by using a series of transcriptomic, cell and biochemical analyses, and the Qki-deficient mouse model, we demonstrate that QKI is indispensable to cardiac sarcomerogenesis and cardiac function through its regulation of alternative splicing in genes involved in Z-disc formation and contractile physiology, suggesting that QKI is associated with the pathogenesis of certain forms of cardiomyopathies.


Asunto(s)
Empalme Alternativo/genética , Desarrollo de Músculos/genética , Contracción Miocárdica/genética , Precursores del ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Actinina/genética , Animales , Diferenciación Celular/genética , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Precursores del ARN/genética , Proteínas de Unión al ARN/genética , Transcriptoma/genética
14.
Front Physiol ; 10: 1102, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31507457

RESUMEN

Toll-like receptor 2 (TLR2), which recognizes several lipopeptides and transduces inflammatory signaling, promotes the pathogenesis of diet-induced dyslipidemia and obesity. TLR2-deficient mice were shown to have improved insulin sensitivity and reduced diet-induced metabolic syndrome. Previous studies demonstrated that prenatal lipopolysaccharide (LPS) exposure causes dyslipidemia accompanied by increased body weight and insulin resistance in offspring. To determine whether TLRs are involved in this complex abnormal phenotype, we analyzed TLR2 and TLR4 expression levels in adipose tissues from offspring with prenatal LPS-exposure (offspring-pLPS) and compared these levels to those of control offspring with prenatal saline-exposure (offspring-pSaline). TLR2 expression was specifically upregulated in the adipose tissue of offspring-pLPS mice. However, unexpectedly, TLR2-deficient offspring-pLPS mice not only presented with an abnormal phenotype comparable to that of wild-type offspring-pLPS mice but also exhibited significantly more severe hyperlipidemia. Our further analyses revealed a dramatic upregulation of TLR4 expression and overactivation of the TLR4/Myd88 signaling pathway in TLR2-deficient offspring-pLPS adipose tissue. Our finding suggests a compensatory genetic interaction between TLR2 and TLR4 in the context of prenatal inflammatory stimulation, and this interaction likely contributes to the prenatal inflammation-induced hyperlipidemia and lipid overload-induced obesity, thus providing a potential mechanism for the fetal origin of adult metabolic diseases.

15.
Diagn Pathol ; 9: 97, 2014 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-24886298

RESUMEN

BACKGROUND: Programmed death-1 (PD-1, Pdcd1)-deficient mice develop different types of autoimmune diseases depending on the mouse strain but its role in uterus development has not been reported. METHODS: In this study, the expression of PD-1 and its ligands, PD-L1 and PD-L2, in uterine tissues from aged WT mice in a 129svEv-Brd background was analyzed by immunohistochemistry and the uterine morphology between WT and PD-1-/- mice was compared by hematoxylin and eosin staining. RESULTS: The aged PD-1-/- female mice in a 129svEv-Brd rather than Balb/c background develop endometrial hyperplasia. H&E staining showed an increase in the number of glands, neovascularization and an extremely large luminal cavity in aged PD-1-/- uteri. Immunohistochemical assay showed that the expression of PD-1 was observed in glandular/luminal epithelium and cells infiltrating the stroma. Fluorescent double staining demonstrated that PD-1 expresses on CD68+ macrophages, CD3+ T cells, CD16+ monocytes, CD56+ NK cells and CK-18+ epithelial cells, respectively. Additionally, PD-1 co-expresses with vascular endothelial growth factor (VEGF), and PD-1 deficiency resulted in an accumulation of glandular/luminal epithelium derived VEGF, which accelerates the expression of the proliferation-associated protein, proliferating cell nuclear antigen (PCNA), and thus potentially lead to epithelial proliferation in aged PD-1-/- uteri. CONCLUSIONS: These findings showed that PD-1 deficiency augments luminal epithelial cell proliferation probably through induced VEGF secretion, suggesting PD-1 plays an important role in controlling the growth and differentiation of the uterine epithelium. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/5809067461223905.


Asunto(s)
Proliferación Celular , Hiperplasia Endometrial/metabolismo , Endometrio/metabolismo , Células Epiteliales/metabolismo , Receptor de Muerte Celular Programada 1/deficiencia , Animales , Antígeno B7-H1/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patología , Endometrio/irrigación sanguínea , Endometrio/patología , Células Epiteliales/patología , Femenino , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Noqueados , Neovascularización Patológica , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Immunol Lett ; 158(1-2): 25-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24286726

RESUMEN

TNF-α plays an essential role in the pathogenesis of fulminant virus hepatitis (FH) caused by infection with murine hepatitis virus strain-3 (MHV-3). However, the specific TNF-α receptors (TNFR) involved in this disease and how they mediate this effect are uncertain. Here, we showed that the expression of TNFR1 and TNFR2 in the liver and spleen was triggered by MHV-3. However, only TNFR1(-/-) mice were resistant to MHV-3 mediated FH, as displayed by a dramatic decrease in tissue necrosis and cell apoptosis in the infected spleens and livers from TNFR1(-/-) mice, as well as prolonged survival in these mice compared to wild type littermate controls. Mechanistically, TNFR1 deficiency directly impeded the serum and tissue levels of fibrinogen-like protein 2 (FGL2), a virus-induced procoagulant molecule that promotes cell apoptosis. Additionally, the expression of apoptosis-associated molecules, Fas and Fas ligand (FasL) in the infected organs from TNFR1(-/-) mice were also decreased. Moreover, the infiltration of neutrophils rather than Foxp3(+) regulatory T cells, which produce proinflammatory factors and FGL2 directly, into the infected liver and spleen tissues was also decreased in TNFR1(-/-) mice. These combined results indicate that signaling through TNFR1 plays an essential role in the pathogenesis of FH caused by MHV-3 infection, and interruption of this signaling pathway could be useful for clinical therapy.


Asunto(s)
Hepatitis Viral Animal/inmunología , Hígado/inmunología , Virus de la Hepatitis Murina/inmunología , Neutrófilos/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis/genética , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Fibrinógeno/metabolismo , Regulación de la Expresión Génica/genética , Hígado/patología , Hígado/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/genética , Neutrófilos/virología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transgenes/genética , Factor de Necrosis Tumoral alfa/inmunología , Receptor fas/genética , Receptor fas/metabolismo
17.
PLoS One ; 9(7): e102273, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25025169

RESUMEN

BACKGROUND: Prenatal exposure to Lipopolysaccharide (LPS) produces hypertension in adult offspring rats. The present study was to explore the effects of prenatal inflammation on morphological and functional changes in the aorta from offspring rats and to further assess its susceptibility to cardiovascular diseases. METHODS AND RESULTS: Pregnant rats were treated intraperitoneally on gestation Days 8, 10 and 12 with saline, LPS (0.79 mg/kg), or pyrrolidine dithiocarbamate (PDTC, 100 mg/kg)+LPS, respectively. Aortic ring reactivity and histopathological alteration were analyzed in offspring at the age of 12 weeks. The detections of connexin (Cx) 37, Cx40, Cx43, and Cx45, including immunofluorescent patterns, protein levels and mRNA expression in the aorta, were performed as well. Furthermore, the expressions of Nuclear factor (NF)-κB (p65), IκBα, phospho-IκBα and IκBß were determined. The results showed that prenatal LPS exposure leads to morphological abnormalities and impaired aortic reactivity in offspring. Prenatal LPS exposure also decreased the protein and mRNA expression of Cx37 in the aorta from offspring rats. NF-κB and phospho-IκBα levels were both increased, IκBα level, however, was decreased in the aorta of offspring from the maternal LPS exposure compared to the controls. Simultaneously, PDTC treatment markedly reversed the action of LPS. CONCLUSIONS: Decreased expression of Cx37 contributed to the aortic dysfunction of prenatal LPS exposure offspring, which should be associated with NF-κB activation.


Asunto(s)
Aorta/fisiopatología , Lipopolisacáridos/efectos adversos , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Aorta/metabolismo , Aorta/patología , Aorta/ultraestructura , Enfermedades Cardiovasculares/etiología , Conexinas/genética , Conexinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas I-kappa B , FN-kappa B/metabolismo , Fosforilación , Embarazo , Ratas
18.
J Mol Histol ; 44(2): 213-20, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23264111

RESUMEN

Signals from the T cell immunoglobulin and mucin-domain (TIM)-containing molecules have been demonstrated to be involved in regulating the progress of carcinoma. However, the expression and anatomical distribution of TIMs in Langerhans cell sarcoma (LCS), which is a rare malignancy derived from dendritic cells of the epidermis, has yet to be determined. In this study, the expression of TIM-1, TIM-3 and TIM-4 in LCS samples were detected by immunohistochemistry. Our results showed that these three molecules were found in LCS sections. At the cellular level, these molecules were found on the cell membrane and in the cytoplasm. Immunofluorescence double-staining demonstrated that these TIMs were co-expressed with Langerin, a potential biomarker for detecting LCS. In addition, TIM-1 was also expressed on CD68(+) macrophages and CK-18(+) epithelial cells, while TIM-3 and TIM-4 were expressed on all cell types investigated, including CD3(+)T cells, CD68(+) macrophages, CD11c(+) dendritic cells, CD16(+) NK Cells, CD31(+) endothelial cells and CK-18(+) epithelial cells. Interestingly, TIMs were also co-expressed with some members of the B7 superfamily, including B7-H1, B7-H3 and B7-H4 on sarcoma cells. Our results clearly showed the characteristic expression and anatomical distribution of TIMs in LCS, and a clear understanding of their functional roles may further elucidate the pathogenesis of this carcinoma and potentially contribute to the development of novel immunotherapeutic strategies.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sarcoma de Células de Langerhans/metabolismo , Antígenos B7/metabolismo , Proteínas de Ciclo Celular/genética , Expresión Génica , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Espacio Intracelular/metabolismo , Fenotipo , Transporte de Proteínas
19.
Inflammation ; 36(1): 110-20, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22895698

RESUMEN

Hepatitis B virus (HBV) infection is a major public health problem, and HBV-related acute-on-chronic liver failure (HBV-ACLF) has an extremely poor prognosis due to a lack of understanding of pathogenesis as well as a lack of effective treatments. Signals from the inhibitory receptor programmed death-1 (PD-1) have been demonstrated to be involved in regulating the pathogenesis of infectious diseases. However, the expression of PD-1 and its ligands in HBV-ACLF patients has yet to be evaluated. In this study, the expression of PD-1 and its ligands, PD-L1 and PD-L2, in liver biopsies from HBV-ACLF as well as chronic hepatitis B (CHB) patients were analyzed by immunohistochemistry. The results showed that all three molecules were observed in the HBV-ACLF samples and their levels were significantly higher than they were in CHB. Immunofluorescence double-staining showed that PD-1 was found on CD3(+), CD8(+) T cells, CD56(+) NK cells, CD68(+) macrophages, CK-18(+) epithelial cells, and CD16(+) monocytes. The PD-L1 expression was observed on all cell types detected and the PD-L2 was chiefly on CK-18(+) epithelial cells and CD31(+) endothelial cells. Interestingly, high levels of virus-induced procoagulant molecule fibrinogen-like protein 2 (FGL2) were observed in liver sections from HBV-ACLF, and PD-L1 and PD-L2 expression was also observed on FGL2(+) cells in these patients. Our combined results suggest that the expression of PD-L1 and PD-L2 may be biomarkers to identify and diagnose ACLF, and a clear understanding of their functional roles should further elucidate the pathogenesis of this disease.


Asunto(s)
Antígeno B7-H1/biosíntesis , Hepatitis B Crónica/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/biosíntesis , Apoptosis , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Enfermedad Hepática en Estado Terminal/metabolismo , Enfermedad Hepática en Estado Terminal/virología , Células Epiteliales/metabolismo , Fibrinógeno/biosíntesis , Virus de la Hepatitis B , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Humanos , Células Asesinas Naturales/metabolismo , Ligandos , Hígado/metabolismo , Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/virología , Macrófagos/metabolismo , Monocitos/metabolismo , Carga Viral
20.
Diagn Pathol ; 7: 142, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23067542

RESUMEN

OBJECTIVE: It has been demonstrated that signals from the inhibitory receptor B and T lymphocyte attenuator (BTLA) are involved in regulating the pathogenesis of infectious diseases. However, the expression and anatomical distribution of BTLA and its ligand, the herpes virus entry mediator (HVEM), have not yet been determined in cases of HBV-related acute-on-chronic liver failure (HBV-ACLF) patients. METHODS: In this study, the expression of BTLA and HVEM in liver tissues from HBV-ACLF, chronic hepatitis B (CHB) patients and healthy individuals was analyzed by immunohistochemistry. RESULTS: The results of this analysis demonstrated that both molecules were observed in the HBV-ACLF samples and that their expression was chiefly in the infiltrating inflammatory cells and the damaged bile ducts. However, they were absent in liver sections from CHB patients and healthy controls. Immunofluorescence double-staining indicated that BTLA was found on CK-18+ epithelial cells, CD31+ endothelial cells, CD68+ macrophages, CD56+ NK cells, CD16+ monocytes, CD3+ , CD8+ T cells, and Foxp3+ regulatory T cells (Treg). By contrast, HVEM expression was restricted to CK18+ epithelial cells and CD68+ macrophages. Moreover, the expression of several members of the B7 superfamily, including PD-L1, PD-L2, B7-H3 and B7-H4, was also detected in these liver tissues, and these proteins were co-expressed with HVEM. Interestingly, the expression of fibrinogen-like protein 2 (FGL2), a virus-induced procoagulant molecule, was also found in liver sections from HBV-ACLF, this molecule also co-expresses with BTLA and HVEM. CONCLUSIONS: These results suggest that BTLA-HVEM signaling is likely to affect the pathogenesis of HBV-ACLF, a clear understanding of the functional roles of these proteins should further elucidate the disease process. VIRTUAL SLIDES: The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/8080806838149123.


Asunto(s)
Enfermedad Hepática en Estado Terminal/metabolismo , Hepatitis B Crónica/complicaciones , Fallo Hepático Agudo/metabolismo , Hígado/química , Receptores Inmunológicos/análisis , Miembro 14 de Receptores del Factor de Necrosis Tumoral/análisis , Conductos Biliares Intrahepáticos/química , Biomarcadores/análisis , Biopsia , Estudios de Casos y Controles , Enfermedad Hepática en Estado Terminal/patología , Enfermedad Hepática en Estado Terminal/virología , Células Endoteliales/química , Células Epiteliales/química , Técnica del Anticuerpo Fluorescente , Hepatitis B Crónica/patología , Hepatitis B Crónica/virología , Humanos , Inmunohistoquímica , Ligandos , Hígado/patología , Hígado/virología , Fallo Hepático Agudo/patología , Fallo Hepático Agudo/virología , Macrófagos/química , Fenotipo , Linfocitos T/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA