Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Am J Pathol ; 187(7): 1537-1550, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28495592

RESUMEN

Peritoneal dialysis (PD) is a life-saving form of renal replacement therapy for those with end-stage kidney disease. Mesothelial cells (MCs) line the peritoneal cavity and help define peritoneal response to treatment-associated injury, a major reason for treatment failure. miRNAs are important regulators, but their roles in peritoneal fibrosis are largely unknown. In this study, miR-21 was one of the most abundant miRNAs in primary MCs, and was up-regulated by the profibrotic cytokine transforming growth factor-ß1 and in PD effluent-derived MCs exhibiting mesenchymal phenotypic change. Increased miR-21 was found in peritoneal membrane biopsy specimens from PD patients compared to healthy controls (PD biocompatible, 5.86×, P = 0.0001; PD conventional, 7.09×, P < 0.0001, n = 11 per group). In PD effluent from a cohort of 230 patients, miR-21 was higher in those receiving the therapy long-term compared to new starters (n = 230, miR-21 3.26×, P = 0.001) and associated with icodextrin use (R = 0.52; 95% CI, 0.20-0.84), peritonitis count (R = 0.16; 95% CI, 0.03-0.29), and dialysate cytokines. miR-21 down-regulated programmed cell death 4 and programmed cell death 4 protein was decreased in peritoneal membrane biopsy specimens from PD patients compared to healthy controls. New miR-21 targets were identified that may be important during PD fibrogenesis. These data identify miR-21 as an important effector of fibrosis in the peritoneal membrane, and a promising biomarker in the dialysis effluent for membrane change in patients receiving PD.


Asunto(s)
Regulación de la Expresión Génica , Fallo Renal Crónico/terapia , MicroARNs/genética , Fibrosis Peritoneal/genética , Peritonitis/genética , Biomarcadores/análisis , Células Cultivadas , Estudios de Cohortes , Regulación hacia Abajo , Células Epiteliales/metabolismo , Epitelio/metabolismo , Glucanos/uso terapéutico , Glucosa/uso terapéutico , Humanos , Icodextrina , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Diálisis Peritoneal , Fibrosis Peritoneal/metabolismo , Peritoneo/metabolismo , Peritonitis/metabolismo , Insuficiencia del Tratamiento , Regulación hacia Arriba
2.
J Immunol ; 197(6): 2195-207, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27527598

RESUMEN

The antimicrobial responsiveness and function of unconventional human T cells are poorly understood, with only limited access to relevant specimens from sites of infection. Peritonitis is a common and serious complication in individuals with end-stage kidney disease receiving peritoneal dialysis. By analyzing local and systemic immune responses in peritoneal dialysis patients presenting with acute bacterial peritonitis and monitoring individuals before and during defined infectious episodes, our data show that Vγ9/Vδ2(+) γδ T cells and mucosal-associated invariant T cells accumulate at the site of infection with organisms producing (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate and vitamin B2, respectively. Such unconventional human T cells are major producers of IFN-γ and TNF-α in response to these ligands that are shared by many microbial pathogens and affect the cells lining the peritoneal cavity by triggering local inflammation and inducing tissue remodeling with consequences for peritoneal membrane integrity. Our data uncover a crucial role for Vγ9/Vδ2 T cells and mucosal-associated invariant T cells in bacterial infection and suggest that they represent a useful predictive marker for important clinical outcomes, which may inform future stratification and patient management. These findings are likely to be applicable to other acute infections where local activation of unconventional T cells contributes to the antimicrobial inflammatory response.


Asunto(s)
Infecciones Bacterianas/inmunología , Linfocitos T/fisiología , Infecciones Bacterianas/patología , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos , Interferón gamma/biosíntesis , Ligandos , Infiltración Neutrófila , Peritonitis/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
3.
Sci Signal ; 17(840): eadn8376, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861613

RESUMEN

Uveal melanoma (UM) is the deadliest form of eye cancer in adults. Inactivating mutations and/or loss of expression of the gene encoding BRCA1-associated protein 1 (BAP1) in UM tumors are associated with an increased risk of metastasis. To investigate the mechanisms underlying this risk, we explored the functional consequences of BAP1 deficiency. UM cell lines expressing mutant BAP1 grew more slowly than those expressing wild-type BAP1 in culture and in vivo. The ability of BAP1 reconstitution to restore cell proliferation in BAP1-deficient cells required its deubiquitylase activity. Proteomic analysis showed that BAP1-deficient cells had decreased phosphorylation of ribosomal S6 and its upstream regulator, p70S6K1, compared with both wild-type and BAP1 reconstituted cells. In turn, expression of p70S6K1 increased S6 phosphorylation and proliferation of BAP1-deficient UM cells. Consistent with these findings, BAP1 mutant primary UM tumors expressed lower amounts of p70S6K1 target genes, and S6 phosphorylation was decreased in BAP1 mutant patient-derived xenografts (PDXs), which grew more slowly than wild-type PDXs in the liver (the main metastatic site of UM) in mice. BAP1-deficient UM cells were also more resistant to amino acid starvation, which was associated with diminished phosphorylation of S6. These studies demonstrate that BAP1 deficiency slows the proliferation of UM cells through regulation of S6 phosphorylation. These characteristics may be associated with metastasis by ensuring survival during amino acid starvation.


Asunto(s)
Proliferación Celular , Melanoma , Transducción de Señal , Proteínas Supresoras de Tumor , Ubiquitina Tiolesterasa , Neoplasias de la Úvea , Animales , Humanos , Ratones , Línea Celular Tumoral , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Mutación , Fosforilación , Proteína S6 Ribosómica/metabolismo , Proteína S6 Ribosómica/genética , Estrés Fisiológico , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología , Femenino
4.
Mol Cancer Ther ; 22(1): 63-74, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36223548

RESUMEN

Uveal melanoma (UM) is the most common intraocular tumor in adults, and up to 50% of patients develop metastatic disease, which remains uncurable. Because patients with metastatic UM have an average survival of less than 1 year after diagnosis, there is an urgent need to develop new treatment strategies. Although activating mutations in Gαq or Gα11 proteins are major drivers of pathogenesis, the therapeutic intervention of downstream Gαq/11 targets has been unsuccessful in treating UM, possibly due to alternative signaling pathways and/or resistance mechanisms. Activation of the insulin-like growth factor 1 (IGF1) signaling pathway promotes cell growth, metastasis, and drug resistance in many types of cancers, including UM, where expression of the IGF1 receptor (IGF1R) correlates with a poor prognosis. In this article, we show that direct inhibition of Gαq/11 by the cyclic depsipeptide YM-254890 in combination with inhibition of IGF1R by linsitinib cooperatively inhibits downstream signaling and proliferation of UM cells. We further demonstrate that a 2-week combination treatment of 0.3 to 0.4 mg/kg of YM-254890 administered by intraperitoneal injection and 25 to 40 mg/kg linsitinib administered by oral gavage effectively inhibits the growth of metastatic UM tumors in immunodeficient NOD scid gamma (NSG) mice and identifies the IGF1 pathway as a potential resistance mechanism in response to Gαq/11 inhibition in UM. These data suggest that the combination of Gαq/11 and IGF1R inhibition provides a promising therapeutic strategy to treat metastatic UM.


Asunto(s)
Melanoma , Neoplasias de la Úvea , Ratones , Animales , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Transducción de Señal , Neoplasias de la Úvea/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Línea Celular Tumoral
5.
Sci Rep ; 11(1): 16499, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34389752

RESUMEN

Acute kidney injury (AKI) is a global clinical problem characterised by a sudden decline in renal function and mortality as high as 60%. Current AKI biomarkers have limited ability to classify disease progression and identify underlying pathological mechanisms. Here we hypothesised that alterations in urinary microRNA profiles could predict AKI recovery/nonrecovery after 90 days, and that injury-specific changes would signify microRNA mediators of AKI pathology. Comparison of urinary microRNA profiles from AKI patients with controls detected significant injury-specific increases in miR-21, miR-126 and miR-141 (p < 0.05) and decreases in miR-192 (p < 0.001) and miR-204 (p < 0.05). Expression of miR-141 increased in renal proximal tubular epithelial cells (PTECs) under oxidative stress in vitro and unilateral ischaemic reperfusion injury in vivo. Forced miR-141 expression in the presence of H2O2 increased PTEC death and decreased cell viability. Of nine messenger RNA targets with two or more miR-141 3'-untranslated region binding sites, we confirmed protein tyrosine phosphatase receptor type G (PTPRG) as a direct miR-141 target in PTECs. PTPRG-specific siRNA knockdown under oxidative stress increased PTEC death and decreased cell viability. In conclusion, we detected significant alterations in five urinary microRNAs following AKI, and identified proximal tubular cell PTPRG as a putative novel therapeutic target.


Asunto(s)
Lesión Renal Aguda/metabolismo , MicroARNs/metabolismo , Animales , Estudios de Casos y Controles , Muerte Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Túbulos Renales Proximales/metabolismo , Masculino , MicroARNs/orina , Persona de Mediana Edad , Estrés Oxidativo , Ratas , Ratas Endogámicas Lew
6.
Mech Ageing Dev ; 164: 37-40, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28373051

RESUMEN

Mesothelial cell (MC) senescence contributes to malignancy and tissue fibrosis. The role of telomere erosion in MC senescence remains controversial, with evidence for both telomere-dependent and telomere-independent mechanisms reported. Single telomere length analysis revealed considerable telomere length heterogeneity in freshly isolated human peritoneal MCs, reflecting a heterogeneous proliferative history and providing high-resolution evidence for telomere-dependent senescence. By contrast the attenuated replicative lifespan, lack of telomere erosion and induction of p16 expression in in vitro-aged cells was consistent with stress-induced senescence. Given the potential pathophysiological impact of senescence in mesothelial tissues, high-resolution MC telomere length analysis may provide clinically useful information.


Asunto(s)
Senescencia Celular/fisiología , Células Epiteliales/metabolismo , Peritoneo/metabolismo , Homeostasis del Telómero/fisiología , Telómero/metabolismo , Células Epiteliales/citología , Epitelio/metabolismo , Humanos , Peritoneo/citología
7.
Perit Dial Int ; 36(4): 367-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26493752

RESUMEN

Long-term peritoneal dialysis (PD) often results in the development of peritoneal fibrosis. In many other fibrosing diseases, monocytes enter the fibrotic lesion and differentiate into fibroblast-like cells called fibrocytes. We find that peritoneal tissue from short-term PD patients contains few fibrocytes, while fibrocytes are readily observed in the peritoneal membrane of long-term PD patients. The PD fluid Dianeal (Baxter Healthcare Corporation, Deerfield, IL, USA) contains dextrose, a number of electrolytes including sodium chloride, and sodium lactate. We find that PD fluid potentiates human fibrocyte differentiation in vitro and implicates sodium lactate in this potentiation. The plasma protein serum amyloid P (SAP) inhibits fibrocyte differentiation. Peritoneal dialysis fluid and sodium chloride decrease the ability of human SAP to inhibit human fibrocyte differentiation in vitro Together, these results suggest that PD fluid contributes to the development of peritoneal fibrosis by potentiating fibrocyte differentiation.


Asunto(s)
Soluciones para Diálisis/farmacología , Fibroblastos/efectos de los fármacos , Fallo Renal Crónico/patología , Fallo Renal Crónico/terapia , Leucocitos Mononucleares/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Adulto , Diferenciación Celular/efectos de los fármacos , Fibroblastos/citología , Humanos , Leucocitos Mononucleares/citología , Fibrosis Peritoneal/etiología , Componente Amiloide P Sérico/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
8.
Biomed Res Int ; 2015: 929806, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26495316

RESUMEN

Preservation of peritoneal cavity homeostasis and peritoneal membrane function is critical for long-term peritoneal dialysis (PD) treatment. Several microRNAs (miRNAs) have been implicated in the regulation of key molecular pathways driving peritoneal membrane alterations leading to PD failure. miRNAs regulate the expression of the majority of protein coding genes in the human genome, thereby affecting most biochemical pathways implicated in cellular homeostasis. In this review, we report published findings on miRNAs and PD therapy, with emphasis on evidence for changes in peritoneal miRNA expression during long-term PD treatment. Recent work indicates that PD effluent- (PDE-) derived cells change their miRNA expression throughout the course of PD therapy, contributing to the loss of peritoneal cavity homeostasis and peritoneal membrane function. Changes in miRNA expression profiles will alter regulation of key molecular pathways, with the potential to cause profound effects on peritoneal cavity homeostasis during PD treatment. However, research to date has mainly adopted a literature-based miRNA-candidate methodology drawing conclusions from modest numbers of patient-derived samples. Therefore, the study of miRNA expression during PD therapy remains a promising field of research to understand the mechanisms involved in basic peritoneal cell homeostasis and PD failure.


Asunto(s)
Homeostasis/efectos de los fármacos , MicroARNs/metabolismo , Cavidad Peritoneal/fisiopatología , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Animales , Soluciones para Diálisis/efectos adversos , Regulación de la Expresión Génica , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA