Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
J Am Soc Nephrol ; 30(5): 726-736, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31000567

RESUMEN

Senescent cells have undergone permanent growth arrest, adopt an altered secretory phenotype, and accumulate in the kidney and other organs with ageing and injury. Senescence has diverse physiologic roles and experimental studies support its importance in nephrogenesis, successful tissue repair, and in opposing malignant transformation. However, recent murine studies have shown that depletion of chronically senescent cells extends healthy lifespan and delays age-associated disease-implicating senescence and the senescence-associated secretory phenotype as drivers of organ dysfunction. Great interest is therefore focused on the manipulation of senescence as a novel therapeutic target in kidney disease. In this review, we examine current knowledge and areas of ongoing uncertainty regarding senescence in the human kidney and experimental models. We summarize evidence supporting the role of senescence in normal kidney development and homeostasis but also senescence-induced maladaptive repair, renal fibrosis, and transplant failure. Recent studies using senescent cell manipulation and depletion as novel therapies to treat renal disease are discussed, and we explore unanswered questions for future research.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Insuficiencia Renal Crónica/patología , Envejecimiento/genética , Animales , Humanos , Riñón/patología , Riñón/fisiopatología , Ratones , Modelos Animales , Pronóstico , Insuficiencia Renal Crónica/mortalidad , Insuficiencia Renal Crónica/terapia
3.
J Am Soc Nephrol ; 28(2): 407-420, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28143966

RESUMEN

Individuals age >65 years old are the fastest expanding population demographic throughout the developed world. Consequently, more aged patients than before are receiving diagnoses of impaired renal function and nephrosclerosis-age-associated histologic changes in the kidneys. Recent studies have shown that the aged kidney undergoes a range of structural changes and has altered transcriptomic, hemodynamic, and physiologic behavior at rest and in response to renal insults. These changes impair the ability of the kidney to withstand and recover from injury, contributing to the high susceptibility of the aged population to AKI and their increased propensity to develop subsequent progressive CKD. In this review, we examine these features of the aged kidney and explore the various validated and putative pathways contributing to the changes observed with aging in both experimental animal models and humans. We also discuss the potential for additional study to increase understanding of the aged kidney and lead to novel therapeutic strategies.


Asunto(s)
Envejecimiento , Riñón/fisiología , Investigación Biomédica/tendencias , Ciclo Celular , Hipoxia de la Célula , Predicción , Humanos , Riñón/citología , Riñón/patología , Riñón/fisiopatología , Masculino , Estrés Oxidativo , Insuficiencia Renal Crónica/etiología , Transducción de Señal
4.
Curr Opin Nephrol Hypertens ; 25(3): 194-202, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27023838

RESUMEN

PURPOSE OF REVIEW: The kidney mediates the excretion or conservation of water and electrolytes in the face of changing fluid and salt intake and losses. To ultrafilter and reabsorb the exact quantities of free water and salts to maintain euvolemia a range of endocrine, paracrine, and hormonal signaling systems have evolved linking the tubules, capillaries, glomeruli, arterioles, and other intrinsic cells of the kidney. Our understanding of these systems remains incomplete. RECENT FINDINGS: Recent work has provided new insights into the workings of the communication pathways between tubular segments and the glomeruli and vasculature, with novel therapeutic agents in development. Particular progress has also been made in the visualization of tubuloglomerular feedback. SUMMARY: The review summarizes our current understanding of pathway functions in health and disease, as well as future therapeutic options to protect the healthy and injured kidney.


Asunto(s)
Capilares/metabolismo , Homeostasis/fisiología , Enfermedades Renales/sangre , Glomérulos Renales/irrigación sanguínea , Túbulos Renales/irrigación sanguínea , Humanos , Hipertensión/fisiopatología
6.
Nephron Clin Pract ; 128(1-2): 29-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25358798

RESUMEN

BACKGROUND/AIMS: Acute kidney injury (AKI) following cardiac surgery is a complication associated with high rates of morbidity and mortality. We compared staging systems for the diagnosis of AKI after cardiac surgery, and assessed pre-operative factors predictive of post-operative AKI. METHODS: Clinical data, surgical risk scores, procedure and clinical outcome were obtained on all 4,651 patients undergoing cardiac surgery to the Royal Infirmary of Edinburgh between April 2006 and March 2011, of whom 4,572 had sufficient measurements of creatinine before and after surgery to permit inclusion and analysis. The presence of AKI was assessed using the AKIN and RIFLE criteria. RESULTS: By AKIN criteria, 12.4% of the studied population developed AKI versus 6.5% by RIFLE criteria. Any post-operation AKI was associated with increased mortality from 2.2 to 13.5% (relative risk 7.0, p < 0.001), and increased inpatient stay from a median of 7 (IQR 4) to 9 (IQR 11) days (p < 0.05). Patients identified by AKIN, but not RIFLE, had a mean peak creatinine rise of 34% from baseline and had a significantly lower mortality compared to RIFLE-'Risk' AKI (mortality 6.1 vs. 9.7%; p < 0.05). Pre-operative creatinine, diabetes, NYHA Class IV dyspnoea and EuroSCORE-1 (a surgical risk score) all predicted subsequent AKI on multivariate analysis. EuroSCORE-1 outperformed any single demographic factor in predicting post-operative AKI risk, equating to an 8% increase in relative risk for each additional point. CONCLUSION: AKI after cardiac surgery is associated with delayed discharge and high mortality rates. The AKIN and RIFLE criteria identify patients at a range of AKI severity levels suitable for trial recruitment. The utility of EuroSCORE as a risk stratification tool to identify high AKI-risk subjects for prospective intervention merits further study.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/etiología , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Anciano , Femenino , Humanos , Masculino , Pronóstico , Estudios Prospectivos , Riesgo , Índice de Severidad de la Enfermedad
7.
JAMA ; 321(9): 901, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30835302
8.
Kidney360 ; 5(1): 142-151, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38049936

RESUMEN

Senescent cells accumulate in the kidney with aging, after acute and chronic injuries, and are present in increased numbers in deteriorating kidney transplants. Senescent cells have undergone permanent cell cycle arrest and release many proinflammatory cytokines/chemokines and profibrotic factors: the senescence-associated secretory phenotype. Recent work from several groups including our own has shown that senescent cells play a causative role in progression of kidney disease. Experimental evidence also indicates that targeting senescent cells has potential to alter the renal regenerative response, reducing progressive fibrosis and improving functional recovery after injury. Research and clinical interest is focused on understanding how accumulating chronic senescent cells link acute injury to progressive fibrosis, dysfunction, and mortality in human CKD. In this review, we outline current protocols for the identification of how senescent cells are identified in vitro and in vivo . We discuss the proposed mechanisms of actions of first-generation senolytic and senomorphic agents, such as ABT-263 (navitoclax) which targets the BCL2 family of survival factors, and senomorphic agents such as metformin which targets aspects of the senescence-associated secretory phenotype. We also review that emerging technologies, such as nanocarriers, are now being developed to have safer delivery systems for senolytics, greater specificity, fewer off-target effects, and less toxicity. Other methods of senescent cell elimination being developed target various immune evasion tactics displayed by these cells. By understanding the role of senescence in kidney homeostasis and disease, developing new, targeted compounds and the tools to allow their efficacy to be charted noninvasively, it should become possible for senolytic treatments to move from the bench to bedside.


Asunto(s)
Senescencia Celular , Insuficiencia Renal Crónica , Humanos , Senescencia Celular/fisiología , Senoterapéuticos , Envejecimiento/genética , Insuficiencia Renal Crónica/terapia , Fibrosis
9.
Am J Respir Crit Care Med ; 186(6): 514-24, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22822022

RESUMEN

RATIONALE: Acute lung injury (ALI) is an important cause of morbidity and mortality, with no currently effective pharmacological therapies. Neutrophils have been specifically implicated in the pathogenesis of ALI, and there has been significant research into the mechanisms of early neutrophil recruitment, but those controlling the later phases of neutrophil emigration that characterize disease are poorly understood. OBJECTIVES: To determine the influence of peripheral blood monocytes (PBMs) in established ALI. METHODS: In a murine model of LPS-induced ALI, three separate models of conditional monocyte ablation were used: systemic liposomal clodronate (sLC), inducible depletion using CD11b diphtheria toxin receptor (CD11b DTR) transgenic mice, and antibody-dependent ablation of CCR2(hi) monocytes. MEASUREMENTS AND MAIN RESULTS: PBMs play a critical role in regulating neutrophil emigration in established murine LPS-induced lung injury. Gr1(hi) and Gr1(lo) PBM subpopulations contribute to this process. PBM depletion is associated with a significant reduction in measures of lung injury. The specificity of PBM depletion was demonstrated by replenishment studies in which the effects were reversed by systemic PBM infusion but not by systemic or local pulmonary infusion of mature macrophages or lymphocytes. CONCLUSIONS: These results suggest that PBMs, or the mechanisms by which they influence pulmonary neutrophil emigration, could represent therapeutic targets in established ALI.


Asunto(s)
Lesión Pulmonar Aguda/patología , Movimiento Celular/inmunología , Macrófagos/citología , Monocitos/citología , Neutrófilos/citología , Lesión Pulmonar Aguda/fisiopatología , Análisis de Varianza , Animales , Líquido del Lavado Bronquioalveolar/citología , Movimiento Celular/fisiología , Ácido Clodrónico/farmacología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inmunohistoquímica , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/inmunología , Neutrófilos/fisiología , Distribución Aleatoria , Estadísticas no Paramétricas
10.
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
11.
Kidney Int ; 82(8): 928-33, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22673886

RESUMEN

The role of resident renal mononuclear phagocytes in acute kidney injury is controversial with experimental data suggesting both deleterious and protective functions. To help resolve this, we used mice transgenic for the human diphtheria toxin receptor under the control of the CD11b promoter and treated them with diphtheria toxin, or liposomal clodronate, or both to deplete monocyte/mononuclear phagocytes prior to renal ischemia/reperfusion injury. Although either system effectively depleted circulating monocytes and resident mononuclear phagocytes, depletion was most marked in diphtheria toxin-treated mice. Despite this, diphtheria toxin treatment did not protect from renal ischemia. In contrast, mice treated with clodronate exhibited reduced renal failure and acute tubular necrosis, suggesting key differences between these depletion strategies. Clodronate did not deplete CD206-positive renal macrophages and, unlike diphtheria toxin, left resident CD11c-positive cells unscathed while inducing dramatic apoptosis in hepatic and splenic mononuclear phagocyte populations. Abolition of the protected phenotype by administration of diphtheria toxin to clodronate-treated mice suggested that the protective effect of clodronate resulted from the presence of a cytoprotective intrarenal population of mononuclear phagocytes sensitive to diphtheria toxin-mediated ablation.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Ácido Clodrónico/farmacología , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Lesión Renal Aguda/patología , Animales , Antígeno CD11b/genética , Antígeno CD11c/metabolismo , Toxina Diftérica/farmacología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Isquemia/tratamiento farmacológico , Isquemia/patología , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Riñón/patología , Lectinas Tipo C/metabolismo , Macrófagos/patología , Macrófagos/fisiología , Masculino , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Transgénicos , Monocitos/patología , Monocitos/fisiología , Regiones Promotoras Genéticas , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/genética , Daño por Reperfusión/patología
12.
JCI Insight ; 7(22)2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36509292

RESUMEN

Progressive fibrosis and maladaptive organ repair result in significant morbidity and millions of premature deaths annually. Senescent cells accumulate with aging and after injury and are implicated in organ fibrosis, but the mechanisms by which senescence influences repair are poorly understood. Using 2 murine models of injury and repair, we show that obstructive injury generated senescent epithelia, which persisted after resolution of the original injury, promoted ongoing fibrosis, and impeded adaptive repair. Depletion of senescent cells with ABT-263 reduced fibrosis in reversed ureteric obstruction and after renal ischemia/reperfusion injury. We validated these findings in humans, showing that senescence and fibrosis persisted after relieved renal obstruction. We next characterized senescent epithelia in murine renal injury using single-cell RNA-Seq. We extended our classification to human kidney and liver disease and identified conserved profibrotic proteins, which we validated in vitro and in human disease. We demonstrated that increased levels of protein disulfide isomerase family A member 3 (PDIA3) augmented TGF-ß-mediated fibroblast activation. Inhibition of PDIA3 in vivo significantly reduced kidney fibrosis during ongoing renal injury and as such represented a new potential therapeutic pathway. Analysis of the signaling pathways of senescent epithelia connected senescence to organ fibrosis, permitting rational design of antifibrotic therapies.


Asunto(s)
Senescencia Celular , Riñón , Ratones , Humanos , Animales , Senescencia Celular/fisiología , Fibrosis , Riñón/patología , Epitelio , Análisis de la Célula Individual
13.
Sci Transl Med ; 14(674): eabj4375, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36475903

RESUMEN

Liver transplantation is the only curative option for patients with end-stage liver disease. Despite improvements in surgical techniques, nonanastomotic strictures (characterized by the progressive loss of biliary tract architecture) continue to occur after liver transplantation, negatively affecting liver function and frequently leading to graft loss and retransplantation. To study the biological effects of organ preservation before liver transplantation, we generated murine models that recapitulate liver procurement and static cold storage. In these models, we explored the response of cholangiocytes and hepatocytes to cold storage, focusing on responses that affect liver regeneration, including DNA damage, apoptosis, and cellular senescence. We show that biliary senescence was induced during organ retrieval and exacerbated during static cold storage, resulting in impaired biliary regeneration. We identified decoy receptor 2 (DCR2)-dependent responses in cholangiocytes and hepatocytes, which differentially affected the outcome of those populations during cold storage. Moreover, CRISPR-mediated DCR2 knockdown in vitro increased cholangiocyte proliferation and decreased cellular senescence but had the opposite effect in hepatocytes. Using the p21KO model to inhibit senescence onset, we showed that biliary tract architecture was better preserved during cold storage. Similar results were achieved by administering senolytic ABT737 to mice before procurement. Last, we perfused senolytics into discarded human donor livers and showed that biliary architecture and regenerative capacities were better preserved. Our results indicate that cholangiocytes are susceptible to senescence and identify the use of senolytics and the combination of senotherapies and machine-perfusion preservation to prevent this phenotype and reduce the incidence of biliary injury after transplantation.


Asunto(s)
Sistema Biliar , Humanos , Ratones , Animales , Constricción Patológica , Senescencia Celular
14.
Kidney Int ; 80(4): 329-31, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21799503

RESUMEN

Garcia et al. report the utility of pharmacological activation of the adenosine A(2A) receptor (A(2A)R) in preserving renal function, reversing fibrosis, and reducing macrophage infiltration and inflammatory activation in rat nephrotoxic nephritis. The role of A(2A)R activation in determining outcome in renal inflammation is discussed.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Adenosina/análogos & derivados , Antiinflamatorios/farmacología , Glomerulonefritis/prevención & control , Riñón/efectos de los fármacos , Fenetilaminas/farmacología , Receptor de Adenosina A2A/efectos de los fármacos , Adenosina/farmacología , Animales , Masculino
15.
Kidney Int ; 79(9): 966-76, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21248714

RESUMEN

Aging is thought to be associated with a higher susceptibility to renal ischemia-reperfusion injury (IRI). To study whether defective induction of hemeoxygenase-1 (HO-1, a protective and anti-inflammatory enzyme) might contribute to this, we found that while 12-month-old mice had similar baseline renal function and HO-1 expression, the induction of HO-1 usually seen in ischemia-reperfusion was reduced. This was also associated with worsened renal function and acute tubular necrosis in the aged compared with young mice. In the older mice, heme arginate (HA) induced HO-1 in the cortex and medulla, significantly improved renal function, and reduced tissue injury. Cellular HO-1 induction in the medulla in response to injury or HA treatment was found to be interstitial rather than epithelial, as evidenced by its colocalization with macrophage markers. In vitro, HA treatment of primary macrophages resulted in marked HO-1 induction without impairment of classical activation pathways. Macrophage depletion, caused by diphtheria toxin treatment of 12-month-old CD11b-DTR transgenic animals, resulted in the loss of interstitial HO-1-positive cells and reversal of the protective phenotype of HA treatment. Thus, failure of HO-1 induction following renal IRI worsens structural and functional injury in older mice and represents a therapeutic target in the elderly. Hence, HO-1-positive renal macrophages mediate HA-induced protection in IRI.


Asunto(s)
Lesión Renal Aguda/enzimología , Envejecimiento , Hemo-Oxigenasa 1/fisiología , Macrófagos/enzimología , Proteínas de la Membrana/fisiología , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/prevención & control , Animales , Arginina/uso terapéutico , Células Cultivadas , Hemo/uso terapéutico , Riñón/enzimología , Ratones , Daño por Reperfusión/enzimología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/prevención & control
16.
Mol Ther ; 18(9): 1706-13, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20551909

RESUMEN

Acute kidney injury has a high mortality and lacks specific therapies, with ischemia/reperfusion injury (IRI) being the predominant cause. Macrophages (M phi) have been used successfully in cell therapy to deliver targeted therapeutic genes in models of inflammatory kidney disease. Heme oxygenase-1 (HO-1) catalyzes heme breakdown and has important cytoprotective functions. We hypothesized that administration of M phi modified to overexpress HO-1 would protect from renal IRI. Using an adenoviral construct (Ad-HO-1), HO-1 was overexpressed in primary bone marrow-derived M phi (BMDM). In vitro Ad-HO-1 M phi showed an anti-inflammatory phenotype with increased phagocytosis of apoptotic cells (ACs) and increased interleukin (IL)-10 but reduced TNF-alpha and nitric oxide (NO) following lipopolysaccharide/interferon-gamma (IFN gamma) stimulation compared to control transduced or unmodified M phi. In vivo, intravenously (IV) injected M phi homed preferentially to the post-IRI kidney compared to uninjured control following experimental IRI. At 24 hours postinjury, despite equivalent levels of tubular necrosis, apoptosis, and capillary density between groups, the injection of Ad-HO-1 M phi resulted in preserved renal function (serum creatinine reduced by 46%), and reduced microvascular platelet deposition. These data demonstrate that genetically modified M phi improve the outcomes in IRI when administered after the establishment of structural injury, raising the prospect of targeted cell therapy to support the function of the acutely injured kidney.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Hemo-Oxigenasa 1/metabolismo , Macrófagos/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/terapia , Adenoviridae/genética , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Técnica del Anticuerpo Fluorescente , Hemo-Oxigenasa 1/genética , Inmunohistoquímica , Inyecciones Intravenosas , Interferón gamma/farmacología , Interleucina-10/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Masculino , Ratones , Daño por Reperfusión/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Front Immunol ; 12: 700790, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220864

RESUMEN

In this review, we examine senescent cells and the overlap between the direct biological impact of senescence and the indirect impact senescence has via its effects on other cell types, particularly the macrophage. The canonical roles of macrophages in cell clearance and in other physiological functions are discussed with reference to their functions in diseases of the kidney and other organs. We also explore the translational potential of different approaches based around the macrophage in future interventions to target senescent cells, with the goal of preventing or reversing pathologies driven or contributed to in part by senescent cell load in vivo.


Asunto(s)
Envejecimiento/patología , Senescencia Celular/fisiología , Fibrosis/patología , Macrófagos , Envejecimiento/inmunología , Animales , Fibrosis/inmunología , Humanos , Riñón/patología
18.
Stem Cells Transl Med ; 10(8): 1232-1248, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33951342

RESUMEN

The renal mesenchyme contains heterogeneous cells, including interstitial fibroblasts and pericytes, with key roles in wound healing. Although healing is impaired in aged kidneys, the effect of age and injury on the mesenchyme remains poorly understood. We characterized renal mesenchymal cell heterogeneity in young vs old animals and after ischemia-reperfusion-injury (IRI) using multiplex immunolabeling and single cell transcriptomics. Expression patterns of perivascular cell markers (α-SMA, CD146, NG2, PDGFR-α, and PDGFR-ß) correlated with their interstitial location. PDGFR-α and PDGFR-ß co-expression labeled renal myofibroblasts more efficiently than the current standard marker α-SMA, and CD146 was a superior murine renal pericyte marker. Three renal mesenchymal subtypes; pericytes, fibroblasts, and myofibroblasts, were recapitulated with data from two independently performed single cell transcriptomic analyzes of murine kidneys, the first dataset an aging cohort and the second dataset injured kidneys following IRI. Mesenchymal cells segregated into subtypes with distinct patterns of expression with aging and following injury. Baseline uninjured old kidneys resembled post-ischemic young kidneys, with this phenotype further exaggerated following IRI. These studies demonstrate that age modulates renal perivascular/interstitial cell marker expression and transcriptome at baseline and in response to injury and provide tools for the histological and transcriptomic analysis of renal mesenchymal cells, paving the way for more accurate classification of renal mesenchymal cell heterogeneity and identification of age-specific pathways and targets.


Asunto(s)
Riñón , Daño por Reperfusión , Anciano , Envejecimiento , Animales , Fibrosis , Humanos , Isquemia/metabolismo , Riñón/patología , Ratones , Microvasos , Miofibroblastos/metabolismo , Pericitos/metabolismo , Daño por Reperfusión/metabolismo
19.
Sci Transl Med ; 13(594)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011625

RESUMEN

The ability of the kidney to regenerate successfully after injury is lost with advancing age, chronic kidney disease, and after irradiation. The factors responsible for this reduced regenerative capacity remain incompletely understood, with increasing interest in a potential role for cellular senescence in determining outcomes after injury. Here, we demonstrated correlations between senescent cell load and functional loss in human aging and chronic kidney diseases including radiation nephropathy. We dissected the causative role of senescence in the augmented fibrosis occurring after injury in aged and irradiated murine kidneys. In vitro studies on human proximal tubular epithelial cells and in vivo mouse studies demonstrated that senescent renal epithelial cells produced multiple components of the senescence-associated secretory phenotype including transforming growth factor ß1, induced fibrosis, and inhibited tubular proliferative capacity after injury. Treatment of aged and irradiated mice with the B cell lymphoma 2/w/xL inhibitor ABT-263 reduced senescent cell numbers and restored a regenerative phenotype in the kidneys with increased tubular proliferation, improved function, and reduced fibrosis after subsequent ischemia-reperfusion injury. Senescent cells are key determinants of renal regenerative capacity in mice and represent emerging treatment targets to protect aging and vulnerable kidneys in man.


Asunto(s)
Senescencia Celular , Daño por Reperfusión , Animales , Fibrosis , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Regeneración , Daño por Reperfusión/patología
20.
Nephron Exp Nephrol ; 115(3): e33-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20424481

RESUMEN

Degradation by the inducible enzyme hemeoxygenase-1 (HO-1) is the principal route of mammalian heme metabolism. The resultant generation of free iron, carbon monoxide and biliverdin results in myriad actions including promoting cell survival, circulatory integrity and immunomodulation. This review examines the evidence from both human studies and work performed in experimental models implicating the intrinsic heme-HO-1 pathway as important in determining both the susceptibility and severity of acute kidney injury. Additional work using chemical inducers of HO-1 has demonstrated the efficacy of strategies to upregulate enzyme activity in ameliorating the severity of experimental ischaemia-reperfusion injury whilst genetic ablation of HO-1 or pharmacological inhibition of HO-1 activity results in an augmented injury phenotype. There remain a multitude of candidate pathways to account for the therapeutic efficacy of HO-1 induction. Although this may reflect a truly multifactorial mechanism of action, the identification of the relative contribution of key components such as carbon monoxide generation remains critical to allow the rational design of agents for translational application in human disease.


Asunto(s)
Lesión Renal Aguda/metabolismo , Hemo-Oxigenasa 1/metabolismo , Daño por Reperfusión/enzimología , Lesión Renal Aguda/etiología , Animales , Apoptosis/efectos de los fármacos , Biliverdina/metabolismo , Monóxido de Carbono/fisiología , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Humanos , Riñón/enzimología , Riñón/inmunología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA