Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Nature ; 543(7643): 108-112, 2017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28225753

RESUMO

Gaucher disease is caused by mutations in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA1 mutations drive extensive accumulation of glucosylceramide (GC) in multiple innate and adaptive immune cells in the spleen, liver, lung and bone marrow, often leading to chronic inflammation. The mechanisms that connect excess GC to tissue inflammation remain unknown. Here we show that activation of complement C5a and C5a receptor 1 (C5aR1) controls GC accumulation and the inflammatory response in experimental and clinical Gaucher disease. Marked local and systemic complement activation occurred in GCase-deficient mice or after pharmacological inhibition of GCase and was associated with GC storage, tissue inflammation and proinflammatory cytokine production. Whereas all GCase-inhibited mice died within 4-5 weeks, mice deficient in both GCase and C5aR1, and wild-type mice in which GCase and C5aR were pharmacologically inhibited, were protected from these adverse effects and consequently survived. In mice and humans, GCase deficiency was associated with strong formation of complement-activating GC-specific IgG autoantibodies, leading to complement activation and C5a generation. Subsequent C5aR1 activation controlled UDP-glucose ceramide glucosyltransferase production, thereby tipping the balance between GC formation and degradation. Thus, extensive GC storage induces complement-activating IgG autoantibodies that drive a pathway of C5a generation and C5aR1 activation that fuels a cycle of cellular GC accumulation, innate and adaptive immune cell recruitment and activation in Gaucher disease. As enzyme replacement and substrate reduction therapies are expensive and still associated with inflammation, increased risk of cancer and Parkinson disease, targeting C5aR1 may serve as a treatment option for patients with Gaucher disease and, possibly, other lysosomal storage diseases.


Assuntos
Proteínas do Sistema Complemento/imunologia , Doença de Gaucher/imunologia , Doença de Gaucher/patologia , Glucosilceramidas/imunologia , Glucosilceramidas/metabolismo , Inflamação/imunologia , Inflamação/patologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Autoanticorpos/imunologia , Ativação do Complemento , Complemento C5a/biossíntese , Complemento C5a/imunologia , Proteínas do Sistema Complemento/biossíntese , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Doença de Gaucher/metabolismo , Doença de Gaucher/prevenção & controle , Glucosilceramidase/antagonistas & inibidores , Glucosilceramidase/deficiência , Glucosilceramidase/genética , Glucosiltransferases/biossíntese , Glucosiltransferases/metabolismo , Humanos , Imunoglobulina G/imunologia , Inflamação/metabolismo , Inflamação/prevenção & controle , Masculino , Camundongos , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia
2.
J Allergy Clin Immunol ; 150(5): 1154-1167, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35792218

RESUMO

BACKGROUND: Hyperinflammation is a life-threatening condition associated with various clinical disorders characterized by excessive immune activation and tissue damage. Multiple cytokines promote the development of hyperinflammation; however, the contribution of IL-10 remains unclear despite emerging speculations for a pathological role. Clinical observations from hemophagocytic lymphohistiocytosis (HLH), a prototypical hyperinflammatory disease, suggest that IL-18 and IL-10 may collectively promote the onset of a hyperinflammatory state. OBJECTIVE: We aimed to investigate the collaborative roles of IL-10 and IL-18 in hyperinflammation. METHODS: A comprehensive plasma cytokine profile for 87 secondary HLH patients was first depicted and analyzed. We then investigated the systemic and cellular effects of coelevated IL-10 and IL-18 in a transgenic mouse model and cultured macrophages. Single-cell RNA sequencing was performed on the monocytes/macrophages isolated from secondary HLH patients to explore the clinical relevance of IL-10/IL-18-mediated cellular signatures. The therapeutic efficacy of IL-10 blockade was tested in HLH mouse models. RESULTS: Excessive circulating IL-10 and IL-18 triggered a lethal hyperinflammatory disease recapitulating HLH-like phenotypes in mice, driving peripheral lymphopenia and a striking shift toward enhanced myelopoiesis in the bone marrow. IL-10 and IL-18 polarized cultured macrophages to a distinct proinflammatory state with pronounced expression of myeloid cell-recruiting chemokines. Transcriptional characterization suggested the IL-10/IL-18-mediated cellular features were clinically relevant with HLH, showing enhanced granzyme expression and proteasome activation in macrophages. IL-10 blockade protected against the lethal disease in HLH mouse models. CONCLUSION: Coelevated IL-10 and IL-18 are sufficient to drive HLH-like hyperinflammatory syndrome, and blocking IL-10 is protective in HLH models.


Assuntos
Interleucina-10 , Interleucina-18 , Linfo-Histiocitose Hemofagocítica , Mielopoese , Animais , Camundongos , Modelos Animais de Doenças , Linfo-Histiocitose Hemofagocítica/patologia
3.
Nature ; 517(7535): 501-4, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25363774

RESUMO

Immunoglobulins protect against disease to a considerable extent by activating complement and stimulatory immunoglobulin crystallizable fragment receptors (Ig FcRs), and aggregating microbial pathogens. Yet IgG1, the predominant murine serum Ig isotype, cannot activate complement by the classical pathway, binds more avidly to an inhibitory than to stimulatory FcRs, and has limited ability to aggregate pathogens. In these regards, it resembles human IgG4 (ref. 4). We hypothesized that limited ability to activate effector mechanisms might protect against immune complex immunopathology. Here we show that IgG1-deficient (γ1(-)) mice, immunized with a potent antigen, develop lethal renal disease soon after they begin to produce antigen-specific antibody, whereas similarly immunized wild-type mice remain healthy. Surprisingly, renal disease in this model is complement and FcR independent and results from immune complex precipitation in glomerular capillaries, as in some cryoglobulinaemic humans. IgG3, which self-associates to form large immune complexes, accounts for more than 97% of the mouse Ig in this cryoglobulin; furthermore, glomerular disease develops when mice are injected with IgG3 anti-trinitrophenyl (TNP) monoclonal antibody followed by a TNP-labelled protein. Renal disease is prevented in both active and passive immunization models by antigen-specific IgG1; other isotypes are less potent at preventing disease. These observations demonstrate the adaptive significance of Ig isotypes that poorly activate effector mechanisms, reveal an immune-complex-dependent, complement- and FcR-independent nephrotoxic mechanism, and suggest that isotypes that poorly activate effector mechanisms may be useful for inhibiting immune complex immunopathology.


Assuntos
Crioglobulinemia/complicações , Glomerulonefrite/etiologia , Glomerulonefrite/prevenção & controle , Imunoglobulina G/imunologia , Animais , Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/imunologia , Antígenos/imunologia , Ligação Competitiva , Proteínas do Sistema Complemento , Crioglobulinemia/imunologia , Crioglobulinemia/patologia , Modelos Animais de Doenças , Feminino , Glomerulonefrite/imunologia , Glomerulonefrite/patologia , Cabras , Masculino , Camundongos , Receptores de IgG , Solubilidade , Trinitrobenzenos/imunologia
4.
Proc Natl Acad Sci U S A ; 115(23): 5998-6003, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784808

RESUMO

Nephrogenesis concludes by the 36th week of gestation in humans and by the third day of postnatal life in mice. Extending the nephrogenic period may reduce the onset of adult renal and cardiovascular disease associated with low nephron numbers. We conditionally deleted either Mtor or Tsc1 (coding for hamartin, an inhibitor of Mtor) in renal progenitor cells. Loss of one Mtor allele caused a reduction in nephron numbers; complete deletion led to severe paucity of glomeruli in the kidney resulting in early death after birth. By contrast, loss of one Tsc1 allele from renal progenitors resulted in a 25% increase in nephron endowment with no adverse effects. Increased progenitor engraftment rates ex vivo relative to controls correlated with prolonged nephrogenesis through the fourth postnatal day. Complete loss of both Tsc1 alleles in renal progenitors led to a lethal tubular lesion. The hamartin phenotypes are not dependent on the inhibitory effect of TSC on the Mtor complex but are dependent on Raptor.


Assuntos
Néfrons , Organogênese/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Néfrons/química , Néfrons/citologia , Néfrons/crescimento & desenvolvimento , Néfrons/fisiologia , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa
5.
J Neurooncol ; 149(3): 511-522, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33034839

RESUMO

PURPOSE: Cyclin-dependent kinase-retinoblastoma (CDK-RB) pathway is dysregulated in some diffuse intrinsic pontine gliomas (DIPG). We evaluated safety, feasibility, and early efficacy of the CDK4/6-inhibitor ribociclib, administered following radiotherapy in newly-diagnosed DIPG patients. METHODS: Following radiotherapy, eligible patients received ribociclib in 28-day cycles (350 mg/m2; 21 days on/7 days off). Feasibility endpoints included tolerability for at least 6 courses, and a less than 2-week delay in restarting therapy after 1 dose reduction. Early efficacy was measured by 1-year and median overall survival (OS). Patient/parent-by-proxy reported outcomes measurement information system (PROMIS) assessments were completed prospectively. RESULTS: The study included 10 evaluable patients, 9 DIPG and 1 diffuse midline glioma (DMG)-all 3.7 to 19.8 years of age. The median number of courses was 8 (range 3-14). Three patients required dose reduction for grade-4 neutropenia, and 1 discontinued therapy for hematological toxicity following course 4. The most common grade-3/4 toxicity was myelosuppression. After 2 courses, MRI evaluations in 4 patients revealed increased necrotic volume, associated with new neurological symptoms in 3 patients. The 1-year and median OS for DIPG was 89% and 16.1 months (range 10-30), respectively; the DMG patient died at 6 months post-diagnosis. Five patients donated brain tissue and tumor; 3 were RB+ . CONCLUSIONS: Ribociclib administered following radiotherapy is feasible in DIPG and DMG. Increased tumor necrosis may represent a treatment effect. These data warrant further prospective volumetric analyses of tumors with necrosis. Feasibility and stabilization findings support further investigation of ribociclib in combination therapies. TRIAL REGISTRATION: NCT02607124.


Assuntos
Aminopiridinas/uso terapêutico , Neoplasias do Tronco Encefálico/terapia , Quimiorradioterapia/métodos , Glioma Pontino Intrínseco Difuso/terapia , Purinas/uso terapêutico , Adolescente , Adulto , Aminopiridinas/farmacocinética , Neoplasias do Tronco Encefálico/patologia , Criança , Pré-Escolar , Glioma Pontino Intrínseco Difuso/patologia , Feminino , Seguimentos , Humanos , Masculino , Dose Máxima Tolerável , Prognóstico , Purinas/farmacocinética , Distribuição Tecidual , Adulto Jovem
6.
Rheumatology (Oxford) ; 58(2): 321-330, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30285245

RESUMO

Objectives: We used an unbiased proteomics approach to identify candidate urine biomarkers (CUBMs) predictive of LN chronicity and pursued their validation in a larger cohort. Methods: In this cross-sectional pilot study, we selected urine collected at kidney biopsy from 20 children with varying levels of LN damage (discovery cohort) and performed proteomic analysis using isobaric tags for relative and absolute quantification (iTRAQ). We identified differentially excreted proteins based on degree of LN chronicity and sought to distinguish markers exhibiting different relative expression patterns using hierarchically clustered log10-normalized relative abundance data with linked and distinct functions by biological network analyses. For each CUBM, we performed specific ELISAs on urine from a validation cohort (n = 41) and analysis of variance to detect differences between LN chronicity, with LN activity adjustment. We evaluated for CUBM expression in LN biopsies with immunohistochemistry. Results: iTRAQ detected 112 proteins in urine from the discovery cohort, 51 quantifiable in all replicates. Simple analysis of variance revealed four differentially expressed, chronicity-correlated proteins (P-values < 0.05). Further correlation and network analyses led to selection of seven CUBMs for LN chronicity. In the validation cohort, none of the CUBMs distinguished LN chronicity degree; however, urine SERPINA3 demonstrated a moderate positive correlation with LN histological activity. Immunohistochemistry further demonstrated SERPINA3 staining in proximal tubular epithelial and endothelial cells. Conclusion: We identified SERPINA3, a known inhibitor of neutrophil cathepsin G and angiotensin II production, as a potential urine biomarker to help quantify LN activity.


Assuntos
Nefrite Lúpica/diagnóstico , Serpinas/urina , Adolescente , Adulto , Biomarcadores/urina , Biópsia , Criança , Estudos Transversais , Feminino , Humanos , Rim/patologia , Nefrite Lúpica/complicações , Nefrite Lúpica/patologia , Masculino , Projetos Piloto , Proteinúria/diagnóstico , Proteinúria/etiologia , Proteômica/métodos , Adulto Jovem
7.
FASEB J ; 32(4): 2073-2085, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203592

RESUMO

Myotonic dystrophy type 1 (DM1) is a progressive neuromuscular disease caused by expanded CUG repeats, which misregulate RNA metabolism through several RNA-binding proteins, including CUG-binding protein/CUGBP1 elav-like factor 1 (CUGBP1/CELF1) and muscleblind 1 protein. Mutant CUG repeats elevate CUGBP1 and alter CUGBP1 activity via a glycogen synthase kinase 3ß (GSK3ß)-cyclin D3-cyclin D-dependent kinase 4 (CDK4) signaling pathway. Inhibition of GSK3ß corrects abnormal activity of CUGBP1 in DM1 mice [human skeletal actin mRNA, containing long repeats ( HSALR) model]. Here, we show that the inhibition of GSK3ß in young HSALR mice prevents development of DM1 muscle pathology. Skeletal muscle in 1-yr-old HSALR mice, treated at 1.5 mo for 6 wk with the inhibitors of GSK3, exhibits high fiber density, corrected atrophy, normal fiber size, with reduced central nuclei and normalized grip strength. Because CUG-GSK3ß-cyclin D3-CDK4 converts the active form of CUGBP1 into a form of translational repressor, we examined the contribution of CUGBP1 in myogenesis using Celf1 knockout mice. We found that a loss of CUGBP1 disrupts myogenesis, affecting genes that regulate differentiation and the extracellular matrix. Proteins of those pathways are also misregulated in young HSALR mice and in muscle biopsies of patients with congenital DM1. These findings suggest that the correction of GSK3ß-CUGBP1 pathway in young HSALR mice might have a positive effect on the myogenesis over time.-Wei, C., Stock, L., Valanejad, L., Zalewski, Z. A., Karns, R., Puymirat, J., Nelson, D., Witte, D., Woodgett, J., Timchenko, N. A., Timchenko, L. Correction of GSK3ß at young age prevents muscle pathology in mice with myotonic dystrophy type 1.


Assuntos
Inibidores Enzimáticos/farmacologia , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Distrofia Miotônica/tratamento farmacológico , Animais , Proteínas CELF1/genética , Células Cultivadas , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Masculino , Camundongos , Desenvolvimento Muscular , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/metabolismo , Distrofia Miotônica/prevenção & controle , Tiadiazóis/farmacologia , Tiadiazóis/uso terapêutico
8.
Am J Med Genet A ; 179(6): 1010-1014, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30895720

RESUMO

Orofaciodigital syndrome type I and X-linked recessive Joubert syndrome are known ciliopathic disorders that are caused by pathogenic variants in OFD1 gene. Endocrine system involvement with these conditions is not well described. We present the first report of a newborn male with a novel hemizygous variant in OFD1 gene c.515T>C, (p.Leu172Pro) resulting in X-linked Joubert syndrome and orofaciodigital features with complete pituitary gland aplasia and subsequent severe hypoplasia of peripheral endocrine glands. This clinical report expands the phenotypic spectrum of endocrine system involvement in OFD1-related disorders and suggests that OFD1 gene may be related to pituitary gland development.


Assuntos
Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Cerebelo/anormalidades , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/genética , Genes Ligados ao Cromossomo X , Doenças Renais Císticas/diagnóstico , Doenças Renais Císticas/genética , Mutação , Fenótipo , Proteínas/genética , Retina/anormalidades , Alelos , Genótipo , Humanos , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Síndromes Orofaciodigitais/diagnóstico , Síndromes Orofaciodigitais/genética , Linhagem , Hipófise/anormalidades , Radiografia , Sequenciamento do Exoma
9.
Pediatr Nephrol ; 34(1): 117-128, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30159624

RESUMO

OBJECTIVES: To delineate urine biomarkers that reflect kidney structural damage and predict renal functional decline in pediatric lupus nephritis (LN). METHODS: In this prospective study, we evaluated kidney biopsies and urine samples of 89 patients with pediatric LN. Urinary levels of 10 biomarkers [adiponectin, ceruloplasmin, kidney injury molecule-1, monocyte chemotactic protein-1, neutrophil gelatinase-associated lipocalin, osteopontin, transforming growth factor-ß (TGFß), vitamin-D binding protein, liver fatty acid binding protein (LFABP), and transferrin] were measured. Regression analysis was used to identify individual and combinations of biomarkers that determine LN damage status [NIH-chronicity index (NIH-CI) score ≤ 1 vs. ≥ 2] both individually and in combination, and biomarker levels were compared for patients with vs. without renal functional decline, i.e., a 20% reduction of the glomerular filtration rate (GFR) within 12 months of a kidney biopsy. RESULTS: Adiponectin, LFABP, and osteopontin levels differed significantly with select histological damage features considered in the NIH-CI. The GFR was associated with NIH-CI scores [Pearson correlation coefficient (r) = - 0.49; p < 0.0001] but not proteinuria (r = 0.20; p > 0.05). Similar to the GFR [area under the ROC curve (AUC) = 0.72; p < 0.01], combinations of osteopontin and adiponectin levels showed moderate accuracy [AUC = 0.75; p = 0.003] in discriminating patients by LN damage status. Renal functional decline occurred more commonly with continuously higher levels of the biomarkers, especially of TGFß, transferrin, and LFABP. CONCLUSION: In combination, urinary levels of adiponectin and osteopontin predict chronic LN damage with similar accuracy as the GFR. Ongoing LN activity as reflected by high levels of LN activity biomarkers heralds renal functional decline. KEY MESSAGES: • Levels of osteopontin and adiponectin measured at the time of kidney biopsy are good predictors of histological damage with lupus nephritis. • Only about 20% of children with substantial kidney damage from lupus nephritis will have an abnormally low urine creatinine clearance. • Continuously high levels of biomarkers reflecting lupus nephritis activity are risk factors of declining renal function.


Assuntos
Falência Renal Crônica/diagnóstico , Rim/fisiopatologia , Nefrite Lúpica/fisiopatologia , Adiponectina/urina , Adolescente , Área Sob a Curva , Biomarcadores/urina , Biópsia , Criança , Progressão da Doença , Feminino , Humanos , Rim/patologia , Falência Renal Crônica/fisiopatologia , Falência Renal Crônica/urina , Testes de Função Renal/métodos , Estudos Longitudinais , Nefrite Lúpica/patologia , Nefrite Lúpica/urina , Masculino , Osteopontina/urina , Prognóstico , Estudos Prospectivos
10.
Dev Dyn ; 247(3): 451-461, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28390160

RESUMO

This article focuses on the role of Rho family GTPases, particularly Rac1 and Rac1b in TGF-ß-induced epithelial-mesenchymal transition (EMT) and EMT-associated responses such as cell migration, invasion, and metastasis in cancer. EMT is considered a prerequisite for cells to adopt a motile and invasive phenotype and eventually become metastatic. A major regulator of EMT and metastasis in cancer is TGF-ß, and its specific functions on tumor cells are mediated beside Smad proteins and mitogen-activated protein kinases (MAPKs) by small GTPases of the Rho/Rac1 family. Available data point to extensive signaling crosstalk between TGF-ß and various Rho GTPases, and in particular a synergistic role of Rho and Rac1 during EMT and cell motility in normal and neoplastic epithelial cells. In contrast, the Rac1-related isoform, Rac1b, emerges as an endogenous inhibitor of Rac1 in TGF-ß signaling, at least in pancreatic carcinoma cells. Given the tumor-promoting role of TGF-ß in late-stage carcinomas and the intimate crosstalk of Rho/Rac1/Rac1b and TGF-ß signaling in various tumor cell responses, targeting specific Rho GTPases may allow for selective interference with prooncogenic TGF-ß responses to aid in anticancer treatments. Developmental Dynamics 247:451-461, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Neoplasias/patologia , Fator de Crescimento Transformador beta/fisiologia , Movimento Celular , Receptor Cross-Talk , Fator de Crescimento Transformador beta/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
11.
Mol Pharmacol ; 92(5): 519-532, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28842394

RESUMO

Transforming growth factor-ß (TGF-ß), serine proteinases such as trypsin, and proteinase-activated receptor 2 (PAR2) promote tumor development by stimulating invasion and metastasis. Previously, we found that in cancer cells derived from pancreatic ductal adenocarcinoma (PDAC) PAR2 protein is necessary for TGF-ß1-dependent cell motility. Here, we show in the same cells that, conversely, the type I TGF-ß receptor activin receptor-like kinase 5 is dispensable for trypsin and PAR2 activating peptide (PAR2-AP)-induced migration. To reveal whether Gq-calcium signaling is a prerequisite for PAR2 to enhance TGF-ß signaling, we investigated the effects of PAR2-APs, PAR2 mutation and PAR2 inhibitors on TGF-ß1-induced migration, reporter gene activity, and Smad activation. Stimulation of cells with PAR2-AP alone failed to enhance basal or TGF-ß1-induced C-terminal phosphorylation of Smad3, Smad-dependent activity of a luciferase reporter gene, and cell migration. Consistently, in complementary loss of function studies, abrogation of the PAR2-Gq-calcium signaling arm failed to suppress TGF-ß1-induced cell migration, reporter gene activity, and Smad3 activation. Together, our findings suggest that the calcium-regulating motif is not required for PAR2 to synergize with TGF-ß1 to promote cell motility. Additional experiments in PDAC cells revealed that PAR2 and TGF-ß1 synergy may involve TGF-ß1 induction of enzymes that cause autocrine cleavage/activation of PAR2, possibly through a biased signaling function. Our results suggest that although reducing PAR2 protein expression may potentially block TGF-ß's prooncogenic function, inhibiting PAR2-Gq-calcium signaling alone would not be sufficient to achieve this effect.


Assuntos
Sinalização do Cálcio/fisiologia , Movimento Celular/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células HEK293 , Humanos , Oligopeptídeos/farmacologia , Receptor PAR-2 , Receptor do Fator de Crescimento Transformador beta Tipo I
12.
Carcinogenesis ; 38(7): 738-747, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28535186

RESUMO

The development of hepatoblastoma (HBL) is associated with failure of hepatic stem cells (HSC) to differentiate into hepatocytes. Despite intensive investigations, mechanisms of the failure of HSC to differentiate are not known. We found that oncogene Gankyrin (Gank) is involved in the inhibition of differentiation of HSC via triggering degradation of tumor suppressor proteins (TSPs) Rb, p53, C/EBPα and HNF4α. Our data show that the activation of a repressor of Gank, farnesoid X receptor, FXR, after initiation of liver cancer by Diethylnitrosamine (DEN) prevents the development of liver cancer by inhibiting Gank and rescuing tumor suppressor proteins. We next analyzed FXR-Gank-Tumor suppressor pathways in a large cohort of HBL patients which include 6 controls and 53 HBL samples. Systemic analysis of these samples and RNA-Seq approach revealed that the FXR-Gank axis is activated; markers of hepatic stem cells are dramatically elevated and hepatocyte markers are reduced in HBL samples. In the course of these studies, we found that RNA binding protein CUGBP1 is a new tumor suppressor protein which is reduced in all HBL samples. Therefore, we generated CUGBP1 KO mice and examined HBL signatures in the liver of these mice. Micro-array studies revealed that the HBL-specific molecular signature is developed in livers of CUGBP1 KO mice at very early ages. Thus, we conclude that FXR-Gank-TSPs-Stem cells pathway is a key determinant of liver cancer in animal models and in pediatric liver cancer. Our data provide a strong basis for development of FXR-Gank-based therapy for treatment of patients with hepatoblastoma.


Assuntos
Proteínas CELF1/genética , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas/genética , Receptores Citoplasmáticos e Nucleares/genética , Animais , Proteínas CELF1/biossíntese , Diferenciação Celular/genética , Linhagem Celular Tumoral , Dietilnitrosamina/toxicidade , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hepatoblastoma/induzido quimicamente , Hepatoblastoma/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Estadiamento de Neoplasias , Pediatria , Receptores Citoplasmáticos e Nucleares/biossíntese
13.
Int J Mol Sci ; 18(11)2017 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-29165389

RESUMO

The G protein-coupled receptor proteinase-activated receptor 2 (PAR2) has been implicated in various aspects of cellular physiology including inflammation, obesity and cancer. In cancer, it usually acts as a driver of cancer progression in various tumor types by promoting invasion and metastasis in response to activation by serine proteinases. Recently, we discovered another mode through which PAR2 may enhance tumorigenesis: crosstalk with transforming growth factor-ß (TGF-ß) signaling to promote TGF-ß1-induced cell migration/invasion and invasion-associated gene expression in ductal pancreatic adenocarcinoma (PDAC) cells. In this chapter, we review what is known about the cellular TGF-ß responses and signaling pathways affected by PAR2 expression, the signaling activities of PAR2 required for promoting TGF-ß signaling, and the potential molecular mechanism(s) that underlie(s) the TGF-ß signaling-promoting effect. Since PAR2 is activated through various serine proteinases and biased agonists, it may couple TGF-ß signaling to a diverse range of other physiological processes that may or may not predispose cells to cancer development such as local inflammation, systemic coagulation and pathogen infection.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Receptor PAR-2/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Humanos , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor PAR-2/química , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
14.
Int J Mol Sci ; 18(12)2017 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-29261154

RESUMO

BACKGROUND: Recently, the expression of proteinase-activated receptor 2 (PAR2) has been shown to be essential for activin receptor-like kinase 5 (ALK5)/SMAD-mediated signaling and cell migration by transforming growth factor (TGF)-ß1. However, it is not known whether activation of non-SMAD TGF-ß signaling (e.g., RAS-RAF-MEK-extracellular signal-regulated kinase (ERK) signaling) is required for cell migration and whether it is also dependent on PAR2. METHODS: RNA interference was used to deplete cells of PAR2, followed by xCELLigence technology to measure cell migration, phospho-immunoblotting to assess ERK1/2 activation, and co-immunoprecipitation to detect a PAR2-ALK5 physical interaction. RESULTS: Inhibition of ERK signaling with the MEK inhibitor U0126 blunted the ability of TGF-ß1 to induce migration in pancreatic cancer Panc1 cells. ERK activation in response to PAR2 agonistic peptide (PAR2-AP) was strong and rapid, while it was moderate and delayed in response to TGF-ß1. Basal and TGF-ß1-dependent ERK, but not SMAD activation, was blocked by U0126 in Panc1 and other cell types indicating that ERK activation is downstream or independent of SMAD signaling. Moreover, cellular depletion of PAR2 in HaCaT cells strongly inhibited TGF-ß1-induced ERK activation, while the biased PAR2 agonist GB88 at 10 and 100 µM potentiated TGF-ß1-dependent ERK activation and cell migration. Finally, we provide evidence for a physical interaction between PAR2 and ALK5. Our data show that both PAR2-AP- and TGF-ß1-induced cell migration depend on ERK activation, that PAR2 expression is crucial for TGF-ß1-induced ERK activation, and that the functional cooperation of PAR2 and TGF-ß1 involves a physical interaction between PAR2 and ALK5.


Assuntos
Movimento Celular , Sistema de Sinalização das MAP Quinases , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular Tumoral , Humanos , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor PAR-2 , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
15.
Hum Mol Genet ; 23(15): 3943-57, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24599400

RESUMO

Gaucher disease, a prevalent lysosomal storage disease (LSD), is caused by insufficient activity of acid ß-glucosidase (GCase) and the resultant glucosylceramide (GC)/glucosylsphingosine (GS) accumulation in visceral organs (Type 1) and the central nervous system (Types 2 and 3). Recent clinical and genetic studies implicate a pathogenic link between Gaucher and neurodegenerative diseases. The aggregation and inclusion bodies of α-synuclein with ubiquitin are present in the brains of Gaucher disease patients and mouse models. Indirect evidence of ß-amyloid pathology promoting α-synuclein fibrillation supports these pathogenic proteins as a common feature in neurodegenerative diseases. Here, multiple proteins are implicated in the pathogenesis of chronic neuronopathic Gaucher disease (nGD). Immunohistochemical and biochemical analyses showed significant amounts of ß-amyloid and amyloid precursor protein (APP) aggregates in the cortex, hippocampus, stratum and substantia nigra of the nGD mice. APP aggregates were in neuronal cells and colocalized with α-synuclein signals. A majority of APP co-localized with the mitochondrial markers TOM40 and Cox IV; a small portion co-localized with the autophagy proteins, P62/LC3, and the lysosomal marker, LAMP1. In cultured wild-type brain cortical neural cells, the GCase-irreversible inhibitor, conduritol B epoxide (CBE), reproduced the APP/α-synuclein aggregation and the accumulation of GC/GS. Ultrastructural studies showed numerous larger-sized and electron-dense mitochondria in nGD cerebral cortical neural cells. Significant reductions of mitochondrial adenosine triphosphate production and oxygen consumption (28-40%) were detected in nGD brains and in CBE-treated neural cells. These studies implicate defective GCase function and GC/GS accumulation as risk factors for mitochondrial dysfunction and the multi-proteinopathies (α-synuclein-, APP- and Aß-aggregates) in nGD.


Assuntos
Doença de Gaucher/genética , Regulação da Expressão Gênica , Mitocôndrias/metabolismo , Neurônios/metabolismo , beta-Glucosidase/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/metabolismo , Doença de Gaucher/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inositol/análogos & derivados , Inositol/farmacologia , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neurônios/patologia , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Agregação Patológica de Proteínas , Substância Negra/metabolismo , Substância Negra/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , beta-Glucosidase/antagonistas & inibidores , beta-Glucosidase/metabolismo
16.
Genome Res ; 23(1): 23-33, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23034409

RESUMO

An unanticipated and tremendous amount of the noncoding sequence of the human genome is transcribed. Long noncoding RNAs (lncRNAs) constitute a significant fraction of non-protein-coding transcripts; however, their functions remain enigmatic. We demonstrate that deletions of a small noncoding differentially methylated region at 16q24.1, including lncRNA genes, cause a lethal lung developmental disorder, alveolar capillary dysplasia with misalignment of pulmonary veins (ACD/MPV), with parent-of-origin effects. We identify overlapping deletions 250 kb upstream of FOXF1 in nine patients with ACD/MPV that arose de novo specifically on the maternally inherited chromosome and delete lung-specific lncRNA genes. These deletions define a distant cis-regulatory region that harbors, besides lncRNA genes, also a differentially methylated CpG island, binds GLI2 depending on the methylation status of this CpG island, and physically interacts with and up-regulates the FOXF1 promoter. We suggest that lung-transcribed 16q24.1 lncRNAs may contribute to long-range regulation of FOXF1 by GLI2 and other transcription factors. Perturbation of lncRNA-mediated chromatin interactions may, in general, be responsible for position effect phenomena and potentially cause many disorders of human development.


Assuntos
Variações do Número de Cópias de DNA , Metilação de DNA , Síndrome da Persistência do Padrão de Circulação Fetal/genética , RNA Longo não Codificante/genética , Cromatina/metabolismo , Cromossomos Humanos Par 16/genética , Ilhas de CpG , Elementos Facilitadores Genéticos , Evolução Fatal , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Impressão Genômica , Células HEK293 , Humanos , Recém-Nascido , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Nucleares/metabolismo , Síndrome da Persistência do Padrão de Circulação Fetal/diagnóstico , Regiões Promotoras Genéticas , RNA Longo não Codificante/metabolismo , Deleção de Sequência , Transcrição Gênica , Proteína Gli2 com Dedos de Zinco
17.
Cancer Cell ; 13(2): 105-16, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18242511

RESUMO

Neurofibromatosis type 1 (Nf1) mutation predisposes to benign peripheral nerve (glial) tumors called neurofibromas. The point(s) in development when Nf1 loss promotes neurofibroma formation are unknown. We show that inactivation of Nf1 in the glial lineage in vitro at embryonic day 12.5 + 1, but not earlier (neural crest) or later (mature Schwann cell), results in colony-forming cells capable of multilineage differentiation. In vivo, inactivation of Nf1 using a DhhCre driver beginning at E12.5 elicits plexiform neurofibromas, dermal neurofibromas, and pigmentation. Tumor Schwann cells uniquely show biallelic Nf1 inactivation. Peripheral nerve and tumors contain transiently proliferating Schwann cells that lose axonal contact, providing insight into early neurofibroma formation. We suggest that timing of Nf1 mutation is critical for neurofibroma formation.


Assuntos
Proteínas Hedgehog/metabolismo , Neurofibroma Plexiforme/patologia , Neurofibromina 1/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Pigmentação , Animais , Axônios/metabolismo , Axônios/patologia , Proliferação de Células , Perda do Embrião , Embrião de Mamíferos/citologia , Gânglios Espinais/citologia , Integrases/metabolismo , Camundongos , Modelos Biológicos , Neurofibroma Plexiforme/ultraestrutura , Neuroglia/citologia , Neuroglia/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Receptor de Fator de Crescimento Neural/metabolismo , Recombinação Genética , Células de Schwann/patologia , Células de Schwann/ultraestrutura , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo
19.
Hum Mol Genet ; 22(12): 2435-50, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23446636

RESUMO

Individual saposin A (A-/-) and saposin B (B-/-)-deficient mice show unique phenotypes caused by insufficient degradation of myelin-related glycosphingolipids (GSLs): galactosylceramide and galactosylsphingosine and sulfatide, respectively. To gain insight into the interrelated functions of saposins A and B, combined saposin AB-deficient mice (AB-/-) were created by knock-in point mutations into the saposins A and B domains on the prosaposin locus. Saposin A and B proteins were undetectable in AB-/- mice, whereas prosaposin, saposin C and saposin D were expressed near wild-type (WT) levels. AB-/- mice developed neuromotor deterioration at >61 days and exhibited abnormal locomotor activity and enhanced tremor. AB-/- mice (~96 days) lived longer than A-/- mice (~85 days), but shorter than B-/- mice (~644 days). Storage materials were observed in Schwann cells and neuronal processes by electron microscopy. Accumulation of p62 and increased levels of LC3-II were detected in the brainstem suggesting altered autophagy. GSL analyses by (liquid chromatography) LC/MS identified substantial increases in lactosylceramide in AB-/- mouse livers. Sulfatide accumulated, but galactosylceramide remained at WT levels, in the AB-/- mouse brains and kidneys. Brain galactosylsphingosine in AB-/- mice was ~68% of that in A-/- mice. These findings indicate that combined saposins A and B deficiencies attenuated GalCer-ß-galactosylceramidase and GM1-ß-galactosidase functions in the degradation of lactosylceramide preferentially in the liver. Blocking sulfatide degradation from the saposin B deficiency diminished galactosylceramide accumulation in the brain and kidney and galctosylsphingosine in the brain. These analyses of AB-/- mice continue to delineate the tissue differential interactions of saposins in GSL metabolism.


Assuntos
Glicoesfingolipídeos/metabolismo , Doenças do Sistema Nervoso/metabolismo , Saposinas/deficiência , Animais , Encéfalo/metabolismo , Feminino , Galactosilceramidase/metabolismo , Humanos , Rim/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/psicologia , Especificidade de Órgãos , Fenótipo , Saposinas/genética , beta-Galactosidase/metabolismo
20.
Mol Genet Metab ; 114(2): 233-241, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25219293

RESUMO

A Caucasian male with Gaucher disease type 3, treated with continuous enzyme therapy (ET) for 11 years, experienced progressive mesenteric and retroperitoneal lymphadenopathy, lung disease, and neurological involvement leading to death at an age of 12.5 years. Autopsy showed significant pathology of the brain, lymph nodes, and lungs. Liver and spleen glucosylceramide (GluCer) and glucosylsphingosine (GluS) levels were nearly normal and storage cells were cleared. Clusters of macrophages and very elevated GluCer and GluS levels were in the lungs, and brain parenchymal and perivascular regions. Compared to normal brain GluCer (GC 18:0), GluCer species with long fatty acid acyl chains were increased in the patient's brain. This profile was similar to that in the patient's lungs, suggesting that these lipids were present in brain perivascular macrophages. In the patient's brain, generalized astrogliosis, and enhanced LC3, ubiquitin, and Tau signals were identified in the regions surrounding macrophage clusters, indicating proinflammation, altered autophagy, and neurodegeneration. These findings highlight the altered phenotypes resulting from increased longevity due to ET, as well as those in poorly accessible compartments of brain and lung, which manifested progressive disease involvement despite ET.


Assuntos
Encéfalo/patologia , Doença de Gaucher/tratamento farmacológico , Pulmão/patologia , Linfonodos/patologia , Adolescente , Progressão da Doença , Terapia de Reposição de Enzimas , Seguimentos , Doença de Gaucher/patologia , Doença de Gaucher/fisiopatologia , Glucosilceramidas/análise , Glicoesfingolipídeos/análise , Humanos , Lipídeos/análise , Lipídeos/líquido cefalorraquidiano , Fígado/química , Fígado/patologia , Longevidade , Pulmão/química , Macrófagos/química , Masculino , Fenótipo , Psicosina/análogos & derivados , Psicosina/análise , Baço/química , Baço/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa