Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 52(4): 700-715.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294409

RESUMEN

The omentum is a visceral adipose tissue rich in fat-associated lymphoid clusters (FALCs) that collects peritoneal contaminants and provides a first layer of immunological defense within the abdomen. Here, we investigated the mechanisms that mediate the capture of peritoneal contaminants during peritonitis. Single-cell RNA sequencing and spatial analysis of omental stromal cells revealed that the surface of FALCs were covered by CXCL1+ mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.


Asunto(s)
Apendicitis/inmunología , Linfocitos/inmunología , Neutrófilos/inmunología , Epiplón/inmunología , Peritonitis/inmunología , Células del Estroma/inmunología , Enfermedad Aguda , Animales , Apendicitis/genética , Apendicitis/microbiología , Comunicación Celular/inmunología , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Epitelio/inmunología , Epitelio/microbiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Trampas Extracelulares/inmunología , Femenino , Expresión Génica , Humanos , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Neutrófilos/microbiología , Epiplón/microbiología , Peritonitis/inducido químicamente , Peritonitis/genética , Peritonitis/microbiología , Arginina Deiminasa Proteína-Tipo 4/genética , Arginina Deiminasa Proteína-Tipo 4/inmunología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células del Estroma/microbiología , Técnicas de Cultivo de Tejidos , Zimosan/administración & dosificación
4.
Cell ; 132(3): 397-409, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18267072

RESUMEN

Adenoviruses are used extensively as gene transfer agents, both experimentally and clinically. However, targeting of liver cells by adenoviruses compromises their potential efficacy. In cell culture, the adenovirus serotype 5 fiber protein engages the coxsackievirus and adenovirus receptor (CAR) to bind cells. Paradoxically, following intravascular delivery, CAR is not used for liver transduction, implicating alternate pathways. Recently, we demonstrated that coagulation factor (F)X directly binds adenovirus leading to liver infection. Here, we show that FX binds to the Ad5 hexon, not fiber, via an interaction between the FX Gla domain and hypervariable regions of the hexon surface. Binding occurs in multiple human adenovirus serotypes. Liver infection by the FX-Ad5 complex is mediated through a heparin-binding exosite in the FX serine protease domain. This study reveals an unanticipated function for hexon in mediating liver gene transfer in vivo.


Asunto(s)
Adenovirus Humanos/fisiología , Proteínas de la Cápside/metabolismo , Factor X/metabolismo , Hígado/virología , Transducción Genética , Internalización del Virus , Adenovirus Humanos/química , Adenovirus Humanos/clasificación , Animales , Proteínas de la Cápside/química , Proteínas Portadoras/metabolismo , Microscopía por Crioelectrón , Factor X/química , Hepatocitos/virología , Humanos , Imagenología Tridimensional , Ratones , Ratones Transgénicos , Modelos Moleculares , Filogenia , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Resonancia por Plasmón de Superficie , Warfarina/farmacología
5.
Int J Mol Sci ; 25(1)2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38203544

RESUMEN

Heart failure and chronic kidney disease (CKD) share several mediators of cardiac pathological remodelling. Akin to heart failure, this remodelling sets in motion a vicious cycle of progressive pathological hypertrophy and myocardial dysfunction in CKD. Several decades of heart failure research have shown that beta blockade is a powerful tool in preventing cardiac remodelling and breaking this vicious cycle. This phenomenon remains hitherto untested in CKD. Therefore, we set out to test the hypothesis that beta blockade prevents cardiac pathological remodelling in experimental uremia. Wistar rats had subtotal nephrectomy or sham surgery and were followed up for 10 weeks. The animals were randomly allocated to the beta blocker metoprolol (10 mg/kg/day) or vehicle. In vivo and in vitro cardiac assessments were performed. Cardiac tissue was extracted, and protein expression was quantified using immunoblotting. Histological analyses were performed to quantify myocardial fibrosis. Beta blockade attenuated cardiac pathological remodelling in nephrectomised animals. The echocardiographic left ventricular mass and the heart weight to tibial length ratio were significantly lower in nephrectomised animals treated with metoprolol. Furthermore, beta blockade attenuated myocardial fibrosis associated with subtotal nephrectomy. In addition, the Ca++- calmodulin-dependent kinase II (CAMKII) pathway was shown to be activated in uremia and attenuated by beta blockade, offering a potential mechanism of action. In conclusion, beta blockade attenuated hypertrophic signalling pathways and ameliorated cardiac pathological remodelling in experimental uremia. The study provides a strong scientific rationale for repurposing beta blockers, a tried and tested treatment in heart failure, for the benefit of patients with CKD.


Asunto(s)
Insuficiencia Cardíaca , Insuficiencia Renal Crónica , Humanos , Ratas , Animales , Ratas Wistar , Metoprolol/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Hipertrofia , Fibrosis
6.
Circ Res ; 126(8): 988-1003, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32065054

RESUMEN

RATIONALE: Despite increasing understanding of the prognostic importance of vascular stiffening linked to perivascular fibrosis in hypertension, the molecular and cellular regulation of this process is poorly understood. OBJECTIVES: To study the functional role of microRNA-214 (miR-214) in the induction of perivascular fibrosis and endothelial dysfunction driving vascular stiffening. METHODS AND RESULTS: Out of 381 miRs screened in the perivascular tissues in response to Ang II (angiotensin II)-mediated hypertension, miR-214 showed the highest induction (8-fold, P=0.0001). MiR-214 induction was pronounced in perivascular and circulating T cells, but not in perivascular adipose tissue adipocytes. Global deletion of miR-214-/- prevented Ang II-induced periaortic fibrosis, Col1a1, Col3a1, Col5a1, and Tgfb1 expression, hydroxyproline accumulation, and vascular stiffening, without difference in blood pressure. Mechanistic studies revealed that miR-214-/- mice were protected against endothelial dysfunction, oxidative stress, and increased Nox2, all of which were induced by Ang II in WT mice. Ang II-induced recruitment of T cells into perivascular adipose tissue was abolished in miR-214-/- mice. Adoptive transfer of miR-214-/- T cells into RAG1-/- mice resulted in reduced perivascular fibrosis compared with the effect of WT T cells. Ang II induced hypertension caused significant change in the expression of 1380 T cell genes in WT, but only 51 in miR-214-/-. T cell activation, proliferation and chemotaxis pathways were differentially affected. MiR-214-/- prevented Ang II-induction of profibrotic T cell cytokines (IL-17, TNF-α, IL-9, and IFN-γ) and chemokine receptors (CCR1, CCR2, CCR4, CCR5, CCR6, and CXCR3). This manifested in reduced in vitro and in vivo T cell chemotaxis resulting in attenuation of profibrotic perivascular inflammation. Translationally, we show that miR-214 is increased in plasma of patients with hypertension and is directly correlated to pulse wave velocity as a measure of vascular stiffness. CONCLUSIONS: T-cell-derived miR-214 controls pathological perivascular fibrosis in hypertension mediated by T cell recruitment and local profibrotic cytokine release.


Asunto(s)
Endotelio Vascular/metabolismo , Hipertensión/genética , Hipertensión/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Linfocitos T/metabolismo , Animales , Endotelio Vascular/patología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Hipertensión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de la Onda del Pulso/métodos , Linfocitos T/patología , Transcriptoma/fisiología
7.
J Am Soc Nephrol ; 31(12): 2833-2854, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32978267

RESUMEN

BACKGROUND: Little is known about the roles of myeloid cell subsets in kidney injury and in the limited ability of the organ to repair itself. Characterizing these cells based only on surface markers using flow cytometry might not provide a full phenotypic picture. Defining these cells at the single-cell, transcriptomic level could reveal myeloid heterogeneity in the progression and regression of kidney disease. METHODS: Integrated droplet- and plate-based single-cell RNA sequencing were used in the murine, reversible, unilateral ureteric obstruction model to dissect the transcriptomic landscape at the single-cell level during renal injury and the resolution of fibrosis. Paired blood exchange tracked the fate of monocytes recruited to the injured kidney. RESULTS: A single-cell atlas of the kidney generated using transcriptomics revealed marked changes in the proportion and gene expression of renal cell types during injury and repair. Conventional flow cytometry markers would not have identified the 12 myeloid cell subsets. Monocytes recruited to the kidney early after injury rapidly adopt a proinflammatory, profibrotic phenotype that expresses Arg1, before transitioning to become Ccr2+ macrophages that accumulate in late injury. Conversely, a novel Mmp12+ macrophage subset acts during repair. CONCLUSIONS: Complementary technologies identified novel myeloid subtypes, based on transcriptomics in single cells, that represent therapeutic targets to inhibit progression or promote regression of kidney disease.


Asunto(s)
Enfermedades Renales/etiología , Enfermedades Renales/patología , Células Mieloides/fisiología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enfermedades Renales/metabolismo , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Obstrucción Ureteral/etiología
8.
Cell Commun Signal ; 17(1): 143, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31703702

RESUMEN

BACKGROUND: Excessive TGF-ß signalling has been shown to underlie pulmonary hypertension (PAH). Human pulmonary artery smooth muscle cells (HPASMCs) can release extracellular vesicles (EVs) but their contents and significance have not yet been studied. Here, we aimed to analyse the contents and biological relevance of HPASMC-EVs and their transport to human pulmonary arterial endothelial cells (HPAECs), as well as the potential alteration of these under pathological conditions. METHODS: We used low-input RNA-Seq to analyse the RNA cargoes sorted into released HPASMC-EVs under basal conditions. We additionally analysed the effects of excessive TGF-ß signalling, using TGF-ß1 and BMP4, in the transcriptome of HPASMCs and their EVs. We then, for the first time, optimised Cre-loxP technology for its use with primary cells in vitro, directly visualising HPASMC-to-HPAEC communication and protein markers on cells taking up EVs. Furthermore we could analyse alteration of this transport with excessive TGF-ß signalling, as well as by other cytokines involved in PAH: IL-1ß, TNF-α and VEGFA. RESULTS: We were able to detect transcripts from 2417 genes in HPASMC-EVs. Surprisingly, among the 759 enriched in HPASMC-EVs compared to their donor cells, we found Zeb1 and 2 TGF-ß superfamily ligands, GDF11 and TGF-ß3. Moreover, we identified 90 genes differentially expressed in EVs from cells treated with TGF-ß1 compared to EVs in basal conditions, including a subset involved in actin and ECM remodelling, among which were bHLHE40 and palladin. Finally, using Cre-loxP technology we showed cell-to-cell transfer and translation of HPASMC-EV Cre mRNA from HPASMC to HPAECs, effectively evidencing communication via EVs. Furthermore, we found increased number of smooth-muscle actin positive cells on HPAECs that took up HPASMC-EVs. The uptake and translation of mRNA was also higher in activated HPAECs, when stimulated with TGF-ß1 or IL-1ß. CONCLUSIONS: HPASMC-EVs are enriched in RNA transcripts that encode genes that could contribute to vascular remodelling and EndoMT during development and PAH, and TGF-ß1 up-regulates some that could enhance this effects. These EVs are functionally transported, increasingly taken up by activated HPAECs and contribute to EndoMT, suggesting a potential effect of HPASMC-EVs in TGF-ß signalling and other related processes during PAH development.


Asunto(s)
Vesículas Extracelulares/metabolismo , Hipertensión Pulmonar/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular , Proteínas Morfogenéticas Óseas/metabolismo , Endotelio Vascular/patología , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Interleucina-1beta/metabolismo , Fenotipo , Factor de Crecimiento Transformador beta3/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
9.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381574

RESUMEN

Human adenoviral serotype 5 (HAdV-5) vectors have predominantly hepatic tropism when delivered intravascularly, resulting in immune activation and toxicity. Coagulation factor X (FX) binding to HAdV-5 mediates liver transduction and provides protection from virion neutralization in mice. FX is dispensable for liver transduction in mice lacking IgM antibodies or complement, suggesting that alternative transduction pathways exist. To identify novel factor(s) mediating HAdV-5 FX-independent entry, we investigated HAdV-5 transduction in vitro in the presence of serum from immunocompetent C57BL/6 or immunocompromised mice lacking IgM antibodies (Rag 2-/- and NOD-scid-gamma [NSG]). Sera from all three mouse strains enhanced HAdV-5 transduction of A549 cells. While inhibition of HAdV-5-FX interaction with FX-binding protein (X-bp) inhibited transduction in the presence of C57BL/6 serum, it had negligible effect on the enhanced transduction observed in the presence of Rag 2-/- or NSG serum. Rag 2-/- serum also enhanced transduction of the FX binding-deficient HAdV-5HVR5*HVR7*E451Q (AdT*). Interestingly, Rag 2-/- serum enhanced HAdV-5 transduction in a FX-independent manner in CHO-CAR and SKOV3-CAR cells (CHO or SKOV3 cells transfected to stably express human coxsackievirus and adenovirus receptor [CAR]). Additionally, blockade of CAR with soluble HAdV-5 fiber knob inhibited mouse serum-enhanced transduction in A549 cells, suggesting a potential role for CAR. Transduction of HAdV-5 KO1 and HAdV-5/F35 (CAR binding deficient) in the presence of Rag 2-/- serum was equivalent to that of HAdV-5, indicating that direct interaction between HAdV-5 and CAR is not required. These data suggest that FX may protect HAdV-5 from neutralization but has minimal contribution to HAdV-5 transduction in the presence of immunocompromised mouse serum. Alternatively, transduction occurs via an unidentified mouse serum protein capable of bridging HAdV-5 to CAR.IMPORTANCE The intravascular administration of HAdV-5 vectors can result in acute liver toxicity, transaminitis, thrombocytopenia, and injury to the vascular endothelium, illustrating challenges yet to overcome for HAdV-5-mediated systemic gene therapy. The finding that CAR and potentially an unidentified factor present in mouse serum might be important mediators of HAdV-5 transduction highlights that a better understanding of the complex biology defining the interplay between adenovirus immune recognition and cellular uptake mechanisms is still required. These findings are important to inform future optimization and development of HAdV-5-based adenoviral vectors for gene therapy.


Asunto(s)
Adenovirus Humanos/metabolismo , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Vectores Genéticos , Suero/inmunología , Células A549 , Adenovirus Humanos/clasificación , Animales , Línea Celular , Línea Celular Tumoral , Factor X/metabolismo , Humanos , Inmunocompetencia , Huésped Inmunocomprometido , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Unión Proteica , Serogrupo , Tropismo Viral
10.
Clin Sci (Lond) ; 132(21): 2383-2385, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30425169

RESUMEN

Accurate biomarkers that both predict the progression to, and detect the early stages of chronic kidney disease (CKD) are lacking, resulting in difficulty in identifying individuals who could potentially benefit from targeted intervention. In a recent issue [Clinical Science (2018) 132, 2121-2133], Cui et al. examine the ability of urinary angiotensinogen (uAGT) to predict the progression of acute kidney injury (AKI) to CKD. They principally employ a murine ischaemia reperfusion injury model to study this and provide data from a small prospective study of patients with biopsy proven acute tubular necrosis. The authors suggest that uAGT is a dynamic marker of renal injury that could be used to predict the likelihood of structural recovery following AKI. Here we comment on their findings, exploring the clinical utility of uAGT as a biomarker to predict AKI to CKD transition and perhaps more controversially, to discuss whether the early renin-angiotensin system blockade following AKI represents a therapeutic target.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Angiotensinógeno , Animales , Biomarcadores , Humanos , Riñón , Ratones , Pronóstico , Estudios Prospectivos
11.
Nephrol Dial Transplant ; 32(10): 1617-1627, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27448672

RESUMEN

BACKGROUND: Hyperphosphataemia is an independent risk factor for accelerated cardiovascular disease in chronic kidney disease (CKD), although the mechanism for this is poorly understood. We investigated the effects of sustained exposure to a high-phosphate environment on endothelial function in cellular and preclinical models, as well as in human subjects. METHODS: Resistance vessels from rats and humans (± CKD) were incubated in a normal (1.18 mM) or high (2.5 mM) phosphate concentration solution and cells were cultured in normal- (0.5 mM) or high-phosphate (3 mM) concentration media. A single-blind crossover study was performed in healthy volunteers, receiving phosphate supplements or a phosphate binder (lanthanum), and endothelial function measured was by flow-mediated dilatation. RESULTS: Endothelium-dependent vasodilatation was impaired when resistance vessels were exposed to high phosphate; this could be reversed in the presence of a phosphodiesterase-5-inhibitor. Vessels from patients with CKD relaxed normally when incubated in normal-phosphate conditions, suggesting that the detrimental effects of phosphate may be reversible. Exposure to high-phosphate disrupted the whole nitric oxide pathway with reduced nitric oxide and cyclic guanosine monophosphate production and total and phospho endothelial nitric oxide synthase expression. In humans, endothelial function was reduced by chronic phosphate loading independent of serum phosphate, but was associated with higher urinary phosphate excretion and serum fibroblast growth factor 23. CONCLUSIONS: These directly detrimental effects of phosphate, independent of other factors in the uraemic environment, may explain the increased cardiovascular risk associated with phosphate in CKD.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Hiperfosfatemia/complicaciones , Óxido Nítrico/fisiología , Insuficiencia Renal Crónica/complicaciones , Animales , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/patología , Células Cultivadas , Estudios Cruzados , GMP Cíclico/metabolismo , Células Endoteliales/enzimología , Endotelio Vascular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Hiperfosfatemia/sangre , Hiperfosfatemia/patología , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatos/fisiología , Fosfatos/toxicidad , Ratas , Ratas Endogámicas WKY , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/patología , Factores de Riesgo , Transducción de Señal , Método Simple Ciego , Vasodilatación/efectos de los fármacos
12.
Nephrol Dial Transplant ; 36(3): 428-429, 2021 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31539062
13.
J Am Soc Nephrol ; 25(1): 65-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24158985

RESUMEN

Renal tubulointerstitial fibrosis is the common end point of progressive renal disease. MicroRNA (miR)-214 and miR-21 are upregulated in models of renal injury, but the function of miR-214 in this setting and the effect of its manipulation remain unknown. We assessed the effect of inhibiting miR-214 in an animal model of renal fibrosis. In mice, genetic deletion of miR-214 significantly attenuated interstitial fibrosis induced by unilateral ureteral obstruction (UUO). Treatment of wild-type mice with an anti-miR directed against miR-214 (anti-miR-214) before UUO resulted in similar antifibrotic effects, and in vivo biodistribution studies demonstrated that anti-miR-214 accumulated at the highest levels in the kidney. Notably, in vivo inhibition of canonical TGF-ß signaling did not alter the regulation of endogenous miR-214 or miR-21. Whereas miR-21 antagonism blocked Smad 2/3 activation, miR-214 antagonism did not, suggesting that miR-214 induces antifibrotic effects independent of Smad 2/3. Furthermore, TGF-ß blockade combined with miR-214 deletion afforded additional renal protection. These phenotypic effects of miR-214 depletion were mediated through broad regulation of the transcriptional response to injury, as evidenced by microarray analysis. In human kidney tissue, miR-214 was detected in cells of the glomerulus and tubules as well as in infiltrating immune cells in diseased tissue. These studies demonstrate that miR-214 functions to promote fibrosis in renal injury independent of TGF-ß signaling in vivo and that antagonism of miR-214 may represent a novel antifibrotic treatment in the kidney.


Asunto(s)
MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/prevención & control , Animales , Modelos Animales de Enfermedad , Fibrosis , Eliminación de Gen , Expresión Génica , Humanos , Imidazoles/farmacología , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinoxalinas/farmacología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Proteína Smad2/antagonistas & inhibidores , Proteína smad3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
14.
Am J Pathol ; 183(6): 1885-1896, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24103556

RESUMEN

Fibrosis pathophysiology is critically regulated by Smad 2- and Smad 3-mediated transforming growth factor-ß (TGF-ß) signaling. Disintegrin metalloproteases (Adam) can manipulate the signaling environment, however, the role and regulation of ADAMs in renal fibrosis remain unclear. TGF-ß stimulation of renal cells results in a significant up-regulation of Adams 10, 17, 12, and 19. The selective Smad2/3 inhibitor SB 525334 reversed these TGF-ß-induced changes. In vivo, using ureteral obstruction to model renal fibrosis, we observed increased Adams gene expression that was blocked by oral administration of SB 525334. Similar increases in Adam gene expression also occurred in preclinical models of hypertension-induced renal damage and glomerulonephritis. miRNAs are a recently discovered second level of regulation of gene expression. Analysis of 3' untranslated regions of Adam12 and Adam19 mRNAs showed multiple binding sites for miR-29a, miR-29b, and miR-29c. We show that miR-29 family expression is decreased after unilateral ureter obstruction and this significant decrease in miR-29 family expression was observed consistently in preclinical models of renal dysfunction and correlated with an increase in Adam12 and Adam19 expression. Exogenous overexpression of the miR-29 family blocked TGF-ß-mediated up-regulation of Adam12 and Adam19 gene expression. This study shows that Adams are involved in renal fibrosis and are regulated by canonical TGF-ß signaling and miR-29. Therefore, both Adams and the miR-29 family represent therapeutic targets for renal fibrosis.


Asunto(s)
Desintegrinas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Glomerulonefritis/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Desintegrinas/genética , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Glomerulonefritis/genética , Glomerulonefritis/patología , Imidazoles/farmacología , Masculino , Ratones , MicroARNs/genética , Quinoxalinas/farmacología , Ratas , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética
15.
Sci Rep ; 14(1): 9573, 2024 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-38670993

RESUMEN

P2X7 receptors mediate immune and endothelial cell responses to extracellular ATP. Acute pharmacological blockade increases renal blood flow and filtration rate, suggesting that receptor activation promotes tonic vasoconstriction. P2X7 expression is increased in kidney disease and blockade/knockout is renoprotective. We generated a P2X7 knockout rat on F344 background, hypothesising enhanced renal blood flow and protection from angiotensin-II-induced renal injury. CRISPR/Cas9 introduced an early stop codon into exon 2 of P2rx7, abolishing P2X7 protein in kidney and reducing P2rx7 mRNA abundance by ~ 60% in bone-marrow derived macrophages. The M1 polarisation response to lipopolysaccharide was unaffected but P2X7 receptor knockout suppressed ATP-induced IL-1ß release. In male knockout rats, acetylcholine-induced dilation of the renal artery ex vivo was diminished but not the response to nitroprusside. Renal function in male and female knockout rats was not different from wild-type. Finally, in male rats infused with angiotensin-II for 6 weeks, P2X7 knockout did not reduce albuminuria, tubular injury, renal macrophage accrual, and renal perivascular fibrosis. Contrary to our hypothesis, global P2X7 knockout had no impact on in vivo renal hemodynamics. Our study does not indicate a major role for P2X7 receptor activation in renal vascular injury.


Asunto(s)
Angiotensina II , Riñón , Ratas Endogámicas F344 , Receptores Purinérgicos P2X7 , Animales , Receptores Purinérgicos P2X7/metabolismo , Receptores Purinérgicos P2X7/genética , Masculino , Ratas , Riñón/metabolismo , Riñón/patología , Femenino , Técnicas de Inactivación de Genes , Macrófagos/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología
16.
Sci Adv ; 9(47): eadj4846, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-38000021

RESUMEN

Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.


Asunto(s)
Insuficiencia Cardíaca , Insuficiencia Renal Crónica , Ratones , Animales , Humanos , Factor de Necrosis Tumoral alfa/genética , Tóxinas Urémicas , Remodelación Ventricular , Insuficiencia Cardíaca/etiología
17.
Sci Transl Med ; 15(698): eabn0736, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37256934

RESUMEN

Progressive fibrosis is a feature of aging and chronic tissue injury in multiple organs, including the kidney and heart. Glioma-associated oncogene 1 expressing (Gli1+) cells are a major source of activated fibroblasts in multiple organs, but the links between injury, inflammation, and Gli1+ cell expansion and tissue fibrosis remain incompletely understood. We demonstrated that leukocyte-derived tumor necrosis factor (TNF) promoted Gli1+ cell proliferation and cardiorenal fibrosis through induction and release of Indian Hedgehog (IHH) from renal epithelial cells. Using single-cell-resolution transcriptomic analysis, we identified an "inflammatory" proximal tubular epithelial (iPT) population contributing to TNF- and nuclear factor κB (NF-κB)-induced IHH production in vivo. TNF-induced Ubiquitin D (Ubd) expression was observed in human proximal tubular cells in vitro and during murine and human renal disease and aging. Studies using pharmacological and conditional genetic ablation of TNF-induced IHH signaling revealed that IHH activated canonical Hedgehog signaling in Gli1+ cells, which led to their activation, proliferation, and fibrosis within the injured and aging kidney and heart. These changes were inhibited in mice by Ihh deletion in Pax8-expressing cells or by pharmacological blockade of TNF, NF-κB, or Gli1 signaling. Increased amounts of circulating IHH were associated with loss of renal function and higher rates of cardiovascular disease in patients with chronic kidney disease. Thus, IHH connects leukocyte activation to Gli1+ cell expansion and represents a potential target for therapies to inhibit inflammation-induced fibrosis.


Asunto(s)
Proteínas Hedgehog , Insuficiencia Renal Crónica , Animales , Humanos , Ratones , Fibrosis , Proteínas Hedgehog/metabolismo , Inflamación , FN-kappa B , Factores de Necrosis Tumoral , Proteína con Dedos de Zinc GLI1
18.
Am J Pathol ; 179(2): 661-72, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21704009

RESUMEN

Transforming growth factor (TGF)-ß is one of the main fibrogenic cytokines that drives the pathophysiology of progressive renal scarring. MicroRNAs (miRNAs) are endogenous non-coding RNAs that post-transcriptionally regulate gene expression. We examined the role of TGF-ß-induced expression of miR-21, miRNAs in cell culture models and miRNA expression in relevant models of renal disease. In vitro, TGF-ß changed expression of miR-21, miR-214, and miR-145 in rat mesangial cells (CRL-2753) and miR-214, miR-21, miR-30c, miR-200b, and miR-200c during induction of epithelial-mesenchymal transition in rat tubular epithelial cells (NRK52E). miR-214 expression was robustly modulated in both cell types, whereas in tubular epithelial cells miR-21 was increased and miR-200b and miR-200c were decreased by 58% and 48%, respectively, in response to TGF-ß. TGF-ß receptor-1 was found to be a target of miR-200b/c and was down-regulated after overexpression of miR-200c. To assess the differential expression of these miRNAs in vivo, we used the anti-Thy1.1 mesangial glomerulonephritis model and the unilateral ureteral obstruction model in which TGF-ß plays a role and also a genetic model of hypertension, the stroke-prone spontaneously hypertensive rat with and without salt loading. The expressions of miR-214 and miR-21 were significantly increased in all in vivo models, showing a possible miRNA signature of renal damage despite differing causes.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Animales , Modelos Animales de Enfermedad , Glomerulonefritis/metabolismo , Hipertensión/patología , Riñón/lesiones , Riñón/metabolismo , Glomérulos Renales/metabolismo , Túbulos Renales/metabolismo , Masculino , Ratas , Ratas Endogámicas WKY , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Uréter/patología
19.
Blood ; 116(15): 2656-64, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20610817

RESUMEN

A major limitation for adenoviral transduction in vivo is the profound liver tropism of adenovirus type 5 (Ad5). Recently, we demonstrated that coagulation factor X (FX) binds to Ad5-hexon protein at high affinity to mediate hepatocyte transduction after intravascular delivery. We developed novel genetically FX-binding ablated Ad5 vectors with lower liver transduction. Here, we demonstrate that FX-binding ablated Ad5 predominantly localize to the liver and spleen 1 hour after injection; however, they had highly reduced liver transduction in both control and macrophage-depleted mice compared with Ad5. At high doses in macrophage-depleted mice, FX-binding ablated vectors transduced the spleen more efficiently than Ad5. Immunohistochemical studies demonstrated transgene colocalization with CD11c(+), ER-TR7(+), and MAdCAM-1(+) cells in the splenic marginal zone. Systemic inflammatory profiles were broadly similar between FX-binding ablated Ad5 and Ad5 at low and intermediate doses, although higher levels of several inflammatory proteins were observed at the highest dose of FX-binding ablated Ad5. Subsequently, we generated a FX-binding ablated virus containing a high affinity Ad35 fiber that mediated a significant improvement in lung/liver ratio in macrophage-depleted CD46(+) mice compared with controls. Therefore, this study documents the biodistribution and reports the retargeting capacity of FX binding-ablated Ad5 vectors in vitro and in vivo.


Asunto(s)
Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Proteínas de la Cápside/metabolismo , Factor X/metabolismo , Vectores Genéticos , Adenovirus Humanos/clasificación , Animales , Proteínas de la Cápside/genética , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Hígado/metabolismo , Hígado/virología , Pulmón/metabolismo , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serotipificación , Bazo/metabolismo , Bazo/virología , Factores de Tiempo , Distribución Tisular , Transducción Genética , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
20.
Adv Drug Deliv Rev ; 182: 114045, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34767865

RESUMEN

The global prevalence of diabetes mellitus was estimated to be 463 million people in 2019 and is predicted to rise to 700 million by 2045. The associated financial and societal costs of this burgeoning epidemic demand an understanding of the pathology of this disease, and its complications, that will inform treatment to enable improved patient outcomes. Nearly two decades after the sequencing of the human genome, the significance of noncoding RNA expression is still being assessed. The family of functional noncoding RNAs known as microRNAs regulates the expression of most genes encoded by the human genome. Altered microRNA expression profiles have been observed both in diabetes and in diabetic complications. These transcripts therefore have significant potential and novelty as targets for therapy, therapeutic agents and biomarkers.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/fisiopatología , Portadores de Fármacos , MicroARNs/farmacología , MicroARNs/uso terapéutico , Biomarcadores , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/fisiopatología , Fibrosis/tratamiento farmacológico , Fibrosis/fisiopatología , Humanos , Hipoglucemiantes/farmacología , Inflamación/metabolismo , MicroARNs/administración & dosificación , Sistema de Administración de Fármacos con Nanopartículas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA