Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(34): e2211370119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969740

RESUMEN

Sepsis, defined as organ dysfunction caused by a dysregulated host-response to infection, is characterized by immunosuppression. The vasopressor norepinephrine is widely used to treat low blood pressure in sepsis but exacerbates immunosuppression. An alternative vasopressor is angiotensin-II, a peptide hormone of the renin-angiotensin system (RAS), which displays complex immunomodulatory properties that remain unexplored in severe infection. In a murine cecal ligation and puncture (CLP) model of sepsis, we found alterations in the surface levels of RAS proteins on innate leukocytes in peritoneum and spleen. Angiotensin-II treatment induced biphasic, angiotensin-II type 1 receptor (AT1R)-dependent modulation of the systemic inflammatory response and decreased bacterial counts in both the blood and peritoneal compartments, which did not occur with norepinephrine treatment. The effect of angiotensin-II was preserved when treatment was delivered remote from the primary site of infection. At an independent laboratory, angiotensin-II treatment was compared in LysM-Cre AT1aR-/- (Myeloid-AT1a-) mice, which selectively do not express AT1R on myeloid-derived leukocytes, and littermate controls (Myeloid-AT1a+). Angiotensin-II treatment significantly reduced post-CLP bacteremia in Myeloid-AT1a+ mice but not in Myeloid-AT1a- mice, indicating that the AT1R-dependent effect of angiotensin-II on bacterial clearance was mediated through myeloid-lineage cells. Ex vivo, angiotensin-II increased post-CLP monocyte phagocytosis and ROS production after lipopolysaccharide stimulation. These data identify a mechanism by which angiotensin-II enhances the myeloid innate immune response during severe systemic infection and highlight a potential role for angiotensin-II to augment immune responses in sepsis.


Asunto(s)
Angiotensina II , Bacteriemia/inmunología , Células Mieloides/metabolismo , Sepsis/inmunología , Angiotensina II/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Receptor de Angiotensina Tipo 1 , Sepsis/metabolismo , Transducción de Señal
2.
Kidney Int ; 105(4): 661-663, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38519230

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) has long been considered a genetic renal disorder, but emerging evidence suggests that the immune microenvironment within the kidney plays a pivotal role in disease progression and severity. In recent years, the previously obscure cytokine interleukin-37 has proved a strong inhibitor of innate immunity in multiple disease models. However, its role in ADPKD has not received scrutiny. In a provocative study published in the current issue, Zylberberg et al. show that interleukin-37 activates interferon signaling in renal macrophages, which inhibits ADPKD initiation. This finding identifies interleukin-37 as a potential viable immunomodulatory therapy for ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Humanos , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón , Citocinas , Progresión de la Enfermedad , Interleucinas
3.
Am J Kidney Dis ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38484869

RESUMEN

RATIONALE & OBJECTIVE: Although functional impairment is common among older adults with chronic kidney disease (CKD), functional reserve before an acute health event and physical resilience after the event have not been characterized in this population. The purpose of this study was to identify distinct patterns of physical function before and after an acute health event among older veterans with stage 4 CKD. STUDY DESIGN: Prospective cohort study. SETTING & PARTICIPANTS: National sample of veterans≥70 years of age with an estimated glomerular filtration rate (eGFR) of<30mL/min/1.73m2 who had an acute care encounter (emergency department visit or hospitalization) during the follow-up period (n = 272). PREDICTORS: Demographic characteristics, eGFR, basic and instrumental activities of daily living (ADL/IADL) difficulty, symptom burden, cognition, depressive symptoms, social support. OUTCOME: Function measured using the life-space mobility assessment obtained by telephone survey before and after an acute care encounter. ANALYTICAL APPROACH: General growth mixture models to identify classes of functional trajectories. Calculation of percentages for demographic characteristics and means for eGFR, ADL/IADL difficulty, symptom burden, cognition, depressive symptoms, and social support by trajectory class. RESULTS: Four trajectory classes were identified and characterized by different levels of life-space mobility before (reserve) and change in life-space mobility after (resilience) an acute care encounter: (1) low reserve, low resilience (n=91), (2) high reserve, high resilience (n=23), (3) moderate reserve, moderate resilience (n=89), and (4) high reserve, low resilience (n=69). Mean levels of ADL/IADL difficulty, symptom burden, cognition, and depressive symptoms, but not demographic characteristics, eGFR, or social support, differed by trajectory class. LIMITATIONS: Veteran cohort was primarily male. CONCLUSIONS: Among older adults with stage 4 CKD, physical function trajectories before and after an acute health event vary. Integrating reserve and resilience into care for this population may be useful for anticipating changes in function and developing tailored treatment plans.

4.
J Am Soc Nephrol ; 34(10): 1629-1646, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37545036

RESUMEN

SIGNIFICANCE STATEMENT: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. However, blockade of IL-1 signaling in AKI has not consistently demonstrated kidney protection. The current murine experiments show that IL-1R1 activation in the proximal tubule exacerbates toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorates AKI by restoring VEGFA-dependent endothelial cell viability. Using this information, future delivery strategies can maximize the protective effects of blocking IL-1R1 while mitigating unwanted actions of IL-1R1 manipulation. BACKGROUND: Activation of the type 1 IL-1 receptor (IL-1R1) triggers a critical innate immune signaling cascade that contributes to the pathogenesis of AKI. IL-1R1 is expressed on some myeloid cell populations and on multiple kidney cell lineages, including tubular and endothelial cells. Pharmacological inhibition of the IL-1R1 does not consistently protect the kidney from injury, suggesting there may be complex, cell-specific effects of IL-1R1 stimulation in AKI. METHODS: To examine expression of IL-1 and IL-1R1 in intrinsic renal versus infiltrating immune cell populations during AKI, we analyzed single-cell RNA sequencing (scRNA-seq) data from kidney tissues of humans with AKI and mice with acute aristolochic acid exposure. We then investigated cell-specific contributions of renal IL-1R1 signaling to AKI using scRNA-seq, RNA microarray, and pharmacological interventions in mice with IL-1R1 deletion restricted to the proximal tubule or endothelium. RESULTS: scRNA-seq analyses demonstrated robust IL-1 expression in myeloid cell populations and low-level IL-1R1 expression in kidney parenchymal cells during toxin-induced AKI. Our genetic studies showed that IL-1R1 activation in the proximal tubule exacerbated toxin-induced AKI and cell death through local suppression of apolipoprotein M. By contrast, IL-1R1 activation in endothelial cells ameliorated aristolochic acid-induced AKI by restoring VEGFA-dependent endothelial cell viability and density. CONCLUSIONS: These data highlight opposing cell-specific effects of IL-1 receptor signaling on AKI after toxin exposure. Disrupting pathways activated by IL-1R1 in the tubule, while preserving those triggered by IL-1R1 activation on endothelial cells, may afford renoprotection exceeding that of global IL-1R1 inhibition while mitigating unwanted actions of IL-1R1 blockade.


Asunto(s)
Lesión Renal Aguda , Receptores de Interleucina-1 , Humanos , Ratones , Animales , Receptores de Interleucina-1/genética , Apolipoproteínas M , Células Endoteliales/metabolismo , Lesión Renal Aguda/patología , Ratones Noqueados , Interleucina-1 , Endotelio/metabolismo , Ratones Endogámicos C57BL
5.
Kidney Int ; 103(3): 514-528, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36334787

RESUMEN

The most common cause of acute kidney injury (AKI) in critically ill patients is sepsis. Kidney macrophages consist of both F4/80hi and CD11bhi cells. The role of macrophage subpopulations in septic AKI pathogenesis remains unclear. As F4/80hi macrophages are reported to contribute to immunomodulation following injury, we hypothesized that selective depletion of F4/80hi macrophages would worsen septic AKI. F4/80hi macrophages were depleted via diphtheria toxin injection in CD11cCre(+)/CX3CR1dtr/wt (F4/80 MKO mice) compared to CD11cCre(-)/CX3CR1dtr/wt (F4/80 MWT) mice. F4/80 MWT and F4/80 MKO mice were subjected to sham or cecal ligation and puncture to induce sepsis. Compared to F4/80 MWT mice, F4/80 MKO mice displayed worsened septic AKI at 24 hours as measured by serum creatinine and histologic injury scoring. Kidneys from F4/80 MKO mice elaborated higher kidney interleukin-6 levels. Mechanistically, single cell RNA sequencing identified a macrophage-endothelial cell immunoregulatory axis that underlies interleukin-6 expression. F4/80hi macrophages expressed interleukin-1 receptor antagonist and limited interleukin-6 expression in endothelial cells. In turn, anti-interleukin-6 therapy ameliorated septic AKI in F4/80 MKO mice. Thus, F4/80hi macrophages express interleukin-1 receptor antagonist and constrain interleukin-6 generation from endothelial cells to limit septic AKI, representing a targetable cellular crosstalk in septic AKI. These findings are particularly relevant owing to the efficacy of anti-interleukin-6 therapies during COVID-19 infection, a disease associated with high rates of AKI and endothelial dysfunction.


Asunto(s)
Lesión Renal Aguda , COVID-19 , Sepsis , Ratones , Animales , Células Endoteliales/patología , COVID-19/complicaciones , Lesión Renal Aguda/patología , Riñón/patología , Macrófagos/metabolismo , Interleucina-6/metabolismo , Sepsis/complicaciones , Receptores de Interleucina-1/metabolismo , Ratones Endogámicos C57BL
6.
Am J Physiol Renal Physiol ; 322(2): F164-F174, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894725

RESUMEN

Interleukin (IL)-1 receptor type 1 (IL-1R1) activation triggers a proinflammatory signaling cascade that can exacerbate kidney injury. However, the functions of podocyte IL-1R1 in glomerular disease remain unclear. To study the role of IL-1R1 signaling in podocytes, we selectively ablated podocyte IL-1R1 in mice (PKO mice). We then subjected PKO mice and wild-type controls to two glomerular injury models: nephrotoxic serum (NTS)- and adriamycin-induced nephropathy. Surprisingly, we found that IL-1R1 activation in podocytes limited albuminuria and podocyte injury during NTS- and adriamycin-induced nephropathy. Moreover, deletion of IL-1R1 in podocytes drove podocyte apoptosis and glomerular injury through diminishing Akt activation. Activation of Akt signaling abrogated the differences in albuminuria and podocyte injury between wild-type and PKO mice during NTS. Thus, IL-1R1 signaling in podocytes limits susceptibility to glomerular injury via an Akt-dependent signaling pathway. These data identify an unexpected protective role for IL-1R1 signaling in podocytes in the pathogenesis of glomerular disease.NEW & NOTEWORTHY The present study establishes that activation of the receptor for interleukin-1 limits susceptibility to damage to the kidney glomerulus in preclinical mouse models by stimulating Akt signaling cascades inside the podocyte.


Asunto(s)
Glomerulonefritis/metabolismo , Podocitos/metabolismo , Proteinuria/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Doxorrubicina , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Glomerulonefritis/prevención & control , Humanos , Interleucina-1beta/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones de la Cepa 129 , Ratones Noqueados , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/inducido químicamente , Proteinuria/patología , Proteinuria/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Tipo I de Interleucina-1/agonistas , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal
7.
BMC Nephrol ; 23(1): 418, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-36585609

RESUMEN

BACKGROUND: The purpose of this manuscript is to introduce reserve and resilience as novel concepts in chronic kidney disease (CKD) research and present baseline data from a unique prospective cohort study designed to characterize recovery from functional decline after a health event. METHODS: The Physical REsilience Prediction in Advanced REnal Disease (PREPARED) study recruited a national, prospective cohort of Veterans ≥70 years old with an estimated glomerular filtration rate (eGFR) < 30 ml/min/1.73 m2, prior nephrology care, and at high risk for hospitalization. Electronic health record data were paired with telephone surveys. Self-reported measures of reserve included physical, psychological, and cognitive capacity and environmental resources. We calculated counts (frequencies) and medians (25th, 75th percentiles) for baseline measures of reserve. The study's longitudinal follow-up of physical function every 8 weeks or following an acute care encounter, which will be used to define resilience, is ongoing. RESULTS: Participants had a median (25th, 75th percentile) age of 76.3 (72.8, 81.4) years and eGFR of 23.4 (18.2, 28.8) ml/min/1.73 m2; 23.3% were Black, and 97.4% were male, 91.6% had hypertension, 67.4% had diabetes mellitus, 46.0% had coronary heart disease, and 39.8% had heart failure. Baseline measures of physical, psychological, and cognitive domains showed low reserve on average, but with wide ranges. CONCLUSIONS: Despite similar levels of kidney function, older adults participating in PREPARED had a wide range of measures of reserve in other health domains. Non-renal measures of reserve may be important indicators of capacity of CKD patients to recover after acute care encounters.


Asunto(s)
Insuficiencia Renal Crónica , Humanos , Masculino , Anciano , Femenino , Estudios Prospectivos , Estudios de Cohortes , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/epidemiología , Riñón , Tasa de Filtración Glomerular , Progresión de la Enfermedad
8.
Annu Rev Physiol ; 80: 283-307, 2018 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-29144825

RESUMEN

The link between inappropriate salt retention in the kidney and hypertension is well recognized. However, growing evidence suggests that the immune system can play surprising roles in sodium homeostasis, such that the study of inflammatory cells and their secreted effectors has provided important insights into salt sensitivity. As part of the innate immune system, myeloid cells have diverse roles in blood pressure regulation, ranging from prohypertensive actions in the kidney, vasculature, and brain, to effects in the skin that attenuate blood pressure elevation. In parallel, T lymphocyte subsets, as key constituents of the adaptive immune compartment, have variable effects on renal sodium handling and the hypertensive response, accruing from the functions of the cytokines that they produce. Conversely, salt can directly modulate the phenotypes of myeloid and T cells, illustrating bidirectional regulatory mechanisms through which sodium and the immune system coordinately impact blood pressure. This review details the complex interplay between myeloid cells, T cells, and salt in the pathogenesis of essential hypertension.


Asunto(s)
Inmunidad Adaptativa/fisiología , Presión Sanguínea/fisiología , Hipertensión/fisiopatología , Riñón/fisiopatología , Monocitos/inmunología , Cloruro de Sodio/metabolismo , Linfocitos T/inmunología , Animales , Citocinas/metabolismo , Humanos , Hipertensión/inmunología , Hipertensión/metabolismo , Riñón/inmunología , Riñón/metabolismo , Monocitos/metabolismo , Linfocitos T/metabolismo
9.
Circ Res ; 125(12): 1055-1066, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31630621

RESUMEN

RATIONALE: The ubiquitin-editing protein A20 in dendritic cells (DCs) suppresses NF-κB (nuclear factor-κB) signaling and constrains DC-mediated T-cell stimulation, but the role of A20 in modulating the hypertensive response requires elucidation. OBJECTIVE: Here, we tested the hypothesis that A20 in CD11c-expressing myeloid cells mitigates Ang II (angiotensin II)-induced hypertension by limiting renal T-cell activation. METHODS AND RESULTS: Mice with heterozygous deletion of A20 in CD11c-expressing myeloid cells (DC ACT[Cd11c-Cre+A20flox/wt]) have spontaneous DC activation but have normal baseline blood pressures. In response to low-dose chronic Ang II infusion, DC ACT mice compared with WT (wild type) controls had an exaggerated hypertensive response and augmented proportions of CD62LloCD44hi effector memory T lymphocytes in the kidney lymph node. After 10 days of Ang II, DC ACT kidneys had increased numbers of memory effector CD8+, but not CD4+ T cells, compared with WTs. Moreover, the expressions of TNF-α (tumor necrosis factor-α) and IFN-γ (interferon-γ) were upregulated in the DC ACT renal CD8+ T cells but not CD4+ T cells. Saline challenge testing revealed enhanced renal fluid retention in the DC ACT mice. DC ACT kidneys showed augmented protein expression of γ-epithelial sodium channel and NHE3 (sodium-hydrogen antiporter 3). DC ACT mice also had greater reductions in renal blood flow following acute injections with Ang II and enhanced oxidant stress in the vasculature as evidenced by higher circulating levels of malondialdehyde compared with WT controls. To directly test whether enhanced T-cell activation in the DC ACT cohort was responsible for their exaggerated hypertensive response, we chronically infused Ang II into lymphocyte-deficient DC ACT Rag1 (recombination activating protein 1)-deficient (Rag1-/-) mice and WT (Cd11c-Cre-A20flox/wt) Rag1-/- controls. The difference in blood pressure elevation accruing from DC activation was abrogated on the Rag1-/- strain. CONCLUSIONS: Following stimulation of the renin-angiotensin system, A20 suppresses DC activation and thereby mitigates T-cell-dependent blood pressure elevation.


Asunto(s)
Células Dendríticas/metabolismo , Hipertensión/metabolismo , Riñón/metabolismo , Células Mieloides/metabolismo , Linfocitos T/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia , Animales , Células Cultivadas , Células Dendríticas/inmunología , Hipertensión/inmunología , Hipertensión/prevención & control , Riñón/citología , Riñón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/inmunología , Linfocitos T/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología
10.
Acta Pharmacol Sin ; 42(12): 2106-2119, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33658705

RESUMEN

Incomplete recovery from episodes of acute kidney injury (AKI) can predispose patients to develop chronic kidney disease (CKD). Although hypoxia-inducible factor-1α (HIF-1α) is a master regulator of the response to hypoxia/ischemia, the role of HIF-1α in CKD progression following incomplete recovery from AKI is poorly understood. Here, we investigated this issue using moderate and severe ischemia/reperfusion injury (I/RI) mouse models. We found that the outcomes of AKI were highly associated with the time course of tubular HIF-1α expression. Sustained activation of HIF-1α, accompanied by the development of renal fibrotic lesions, was found in kidneys with severe AKI. The AKI to CKD progression was markedly ameliorated when PX-478 (a specific HIF-1α inhibitor, 5 mg· kg-1·d-1, i.p.) was administered starting on day 5 after severe I/RI for 10 consecutive days. Furthermore, we demonstrated that HIF-1α C-terminal transcriptional activation domain (C-TAD) transcriptionally stimulated KLF5, which promoted progression of CKD following severe AKI. The effect of HIF-1α C-TAD activation on promoting AKI to CKD progression was also confirmed in in vivo and in vitro studies. Moreover, we revealed that activation of HIF-1α C-TAD resulted in the loss of FIH-1, which was the key factor governing HIF-1α-driven AKI to CKD progression. Overexpression of FIH-1 inhibited HIF-1α C-TAD and prevented AKI to CKD progression. Thus, FIH-1-modulated HIF-1α C-TAD activation was the key mechanism of AKI to CKD progression by transcriptionally regulating KLF5 pathway. Our results provide new insights into the role of HIF-1α in AKI to CKD progression and also the potential therapeutic strategy for the prevention of renal diseases progression.


Asunto(s)
Lesión Renal Aguda/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Oxigenasas de Función Mixta/metabolismo , Insuficiencia Renal Crónica/etiología , Transducción de Señal/efectos de los fármacos , Lesión Renal Aguda/patología , Animales , Progresión de la Enfermedad , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Riñón/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Compuestos de Mostaza/uso terapéutico , Fenilpropionatos/uso terapéutico , Dominios Proteicos , Insuficiencia Renal Crónica/patología , Regulación hacia Arriba/fisiología
11.
Am J Physiol Renal Physiol ; 318(1): F107-F116, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31736350

RESUMEN

Nephrotoxic serum nephritis (NTN) models immune-mediated human glomerulonephritis and culminates in kidney inflammation and fibrosis, a process regulated by T lymphocytes. TNF-α is a key proinflammatory cytokine that contributes to diverse forms of renal injury. Therefore, we posited that TNF-α from T lymphocytes may contribute to NTN pathogenesis. Here, mice with T cell-specific deletion of TNF-α (TNF TKO) and wild-type (WT) control mice were subjected to the NTN model. At 14 days after NTN, kidney injury and fibrosis were increased in kidneys from TNF TKO mice compared with WT mice. PD1+CD4+ T cell numbers and mRNA levels of IL-17A were elevated in NTN kidneys of TNF TKO mice, suggesting that augmented local T helper 17 lymphocyte responses in the TNF TKO kidney may exaggerate renal injury and fibrosis. In turn, we found increased accumulation of neutrophils in TNF TKO kidneys during NTN. We conclude that TNF-α production in T lymphocytes mitigates NTN-induced kidney injury and fibrosis by inhibiting renal T helper 17 lymphocyte responses and infiltration of neutrophils.


Asunto(s)
Fibrosis/metabolismo , Glomerulonefritis/metabolismo , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/patología , Glomerulonefritis/genética , Glomerulonefritis/patología , Interleucina-17/genética , Interleucina-17/metabolismo , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Noqueados , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/genética
12.
Kidney Int ; 97(4): 651-653, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32200858

RESUMEN

Chronic kidney disease features chronic inflammation and fibrosis, both of which contribute to and are exacerbated by arterial hypertension. The contribution of immune responses to renal sodium retention has received intense scrutiny. In this regard, the article by Furusho et al. details a mechanism wherein intrarenal TNFα augments salt-sensitive hypertension during CKD via activation of the WNK1-SPAKNCC phosphorylation cascade.


Asunto(s)
Hipertensión , Insuficiencia Renal Crónica , Citocinas , Humanos , Riñón , Nefronas , Fosforilación , Factor de Necrosis Tumoral alfa
13.
Kidney Int ; 97(1): 119-129, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31685313

RESUMEN

Tubulointerstitial disease in the kidney culminates in renal fibrosis that portents organ failure. Twist1, a basic helix-loop-helix protein 38 transcription factor, regulates several essential biological functions, but inappropriate Twist1 activity in the kidney epithelium can trigger kidney fibrogenesis and chronic kidney disease. By contrast, Twist1 in circulating myeloid cells may constrain inflammatory injury by attenuating cytokine generation. To dissect the effects of Twist1 in kidney tubular versus immune cells on renal inflammation following toxin-induced renal injury, we subjected mice with selective deletion of Twist1 in renal epithelial cells or macrophages to aristolochic acid-induced chronic kidney disease. Ablation of Twist1 in the distal nephron attenuated kidney damage, interstitial fibrosis, and renal inflammation after aristolochic acid exposure. However, macrophage-specific deletion of Twist1 did not impact the development of aristolochic acid-induced nephropathy. In vitro studies confirmed that Twist1 in renal tubular cells underpins their susceptibility to apoptosis and propensity to generate pro-fibrotic mediators in response to aristolochic acid. Moreover, co-culture studies revealed that Twist1 in renal epithelia augmented the recruitment and activation of pro-inflammatory CD64+ macrophages. Thus, Twist1 in the distal nephron rather than in infiltrating macrophages propagates chronic inflammation and fibrogenesis during aristolochic acid-induced nephropathy.


Asunto(s)
Túbulos Renales Distales/patología , Macrófagos/inmunología , Nefritis Intersticial/inmunología , Insuficiencia Renal Crónica/inmunología , Proteína 1 Relacionada con Twist/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Ácidos Aristolóquicos/toxicidad , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Fibrosis , Técnicas de Silenciamiento del Gen , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Túbulos Renales Distales/citología , Túbulos Renales Distales/inmunología , Túbulos Renales Distales/metabolismo , Lipocalina 2/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Nefritis Intersticial/inducido químicamente , Nefritis Intersticial/patología , Cultivo Primario de Células , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/patología , Proteína 1 Relacionada con Twist/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
14.
Am J Pathol ; 189(5): 981-988, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31000207

RESUMEN

Most forms of chronic kidney disease culminate in renal fibrosis that heralds organ failure. In contrast to the protective effects of globally blocking type 1 angiotensin (AT1) receptors throughout the body, activating AT1 receptors directly on immune cells may serve protective functions. However, the effects of stimulating the T-cell AT1 receptor on the progression of renal fibrosis remain unknown. In this study, mice with T-cell-specific deletion of the dominant murine AT1 receptor isoform Lck-Cre Agtraflox/flox [total knockout (TKO)] and wild-type (WT) controls were subjected to the unilateral ureteral obstruction model of kidney fibrosis. Compared with WT controls, obstructed kidneys from TKO mice at day 14 had increased collagen 1 deposition. CD4+ T cells, CD11b+Ly6Chi myeloid cells, and mRNA levels of Th1 inflammatory cytokines are elevated in obstructed TKO kidneys, suggesting that augmented Th1 responses in the TKO mice may exaggerate renal fibrosis by driving proinflammatory macrophage differentiation. In turn, T-bet deficient (T-bet knockout) mice lacking Th1 responses have attenuated collagen deposition after unilateral ureteral obstruction. We conclude that activating the AT1 receptor on T cells mitigates renal fibrogenesis by inhibiting Th1 differentiation and renal accumulation of profibrotic macrophages.


Asunto(s)
Fibrosis/prevención & control , Inflamación/prevención & control , Enfermedades Renales/prevención & control , Receptor de Angiotensina Tipo 1/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis/inmunología , Fibrosis/metabolismo , Fibrosis/patología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Enfermedades Renales/inmunología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Noqueados , Linfocitos T/patología
15.
Curr Opin Nephrol Hypertens ; 29(3): 286-292, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32235271

RESUMEN

PURPOSE OF REVIEW: Macrophages play an important role in regulating homeostasis, kidney injury, repair, and tissue fibrogenesis. The present review will discuss recent advances that explore the novel subsets and functions of macrophage in the pathogenesis of kidney damage and hypertension. RECENT FINDINGS: Macrophages differentiate into a variety of subsets in microenvironment-dependent manner. Although the M1/M2 nomenclature is still applied in considering the pro-inflammatory versus anti-inflammatory effects of macrophages in kidney injury, novel, and accurate macrophage phenotypes are defined by flow cytometric markers and single-cell RNA signatures. Studies exploring the crosstalk between macrophages and other cells are rapidly advancing with the additional recognition of exosome trafficking between cells. Using murine conditional mutants, actions of macrophage can be defined more precisely than in bone marrow transfer models. Some studies revealed the opposing effects of the same protein in renal parenchymal cells and macrophages, highlighting a need for the development of cell-specific immune therapies for translation. SUMMARY: Macrophage-targeted therapies hold potential for limiting kidney injury and hypertension. To realize this potential, future studies will be required to understand precise mechanisms in macrophage polarization, crosstalk, proliferation, and maturation in the setting of renal disease.


Asunto(s)
Enfermedades Renales/etiología , Macrófagos/fisiología , Lesión Renal Aguda/etiología , Animales , Polaridad Celular , Humanos , Hipertensión/etiología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/fisiopatología , Macrófagos/efectos de los fármacos , Ratones , Insuficiencia Renal Crónica/etiología
16.
Curr Opin Nephrol Hypertens ; 29(5): 515-522, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32701602

RESUMEN

PURPOSE OF REVIEW: Inflammatory processes play a critical role in the pathogenesis of hypertension. Innate and adaptive immune responses participate in blood pressure (BP) elevation and end-organ damage. In this review, we discuss recent studies illustrating mechanisms through which immune cells and cytokines regulate BP via their actions in the kidney. RECENT FINDINGS: Cells of the innate immune system, including monocytes, neutrophils, and dendritic cells, can all promote BP elevation via effects on kidney function. These innate immune cells can directly impact oxidative stress and cytokine generation in the kidney and/or present antigens to lymphocytes for the engagement of the adaptive immune system. Once activated by dendritic cells, effector memory T cells accumulate in the hypertensive kidney and facilitate renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha (TNF-α), interleukin-17a (IL-17a), and interferon-gamma (IFN-γ), each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. B cells, regulate blood pressure via vasopressin receptor 2 (V2R)-dependent effects on fluid transport in the kidney. SUMMARY: Immune cells of the innate and adaptive immune systems drive sodium retention and blood pressure elevation in part by altering renal solute transport.


Asunto(s)
Hipertensión/inmunología , Riñón/fisiopatología , Inmunidad Adaptativa , Animales , Citocinas/fisiología , Células Dendríticas/inmunología , Humanos , Hipertensión/etiología , Inmunidad Innata
17.
J Am Soc Nephrol ; 30(10): 1925-1938, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31337692

RESUMEN

BACKGROUND: Polarized macrophage populations can orchestrate both inflammation of the kidney and tissue repair during CKD. Proinflammatory M1 macrophages initiate kidney injury, but mechanisms through which persistent M1-dependent kidney damage culminates in fibrosis require elucidation. Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor that suppresses inflammatory signals, is an essential regulator of macrophage polarization in adipose tissues, but the effect of myeloid KLF4 on CKD progression is unknown. METHODS: We used conditional mutant mice lacking KLF4 or TNFα (KLF4's downstream effector) selectively in myeloid cells to investigate macrophage KLF4's role in modulating CKD progression in two models of CKD that feature robust macrophage accumulation, nephrotoxic serum nephritis, and unilateral ureteral obstruction. RESULTS: In these murine CKD models, KLF4 deficiency in macrophages infiltrating the kidney augmented their M1 polarization and exacerbated glomerular matrix deposition and tubular epithelial damage. During the induced injury in these models, macrophage-specific KLF4 deletion also exacerbated kidney fibrosis, with increased levels of collagen 1 and α-smooth muscle actin in the injured kidney. CD11b+Ly6Chi myeloid cells isolated from injured kidneys expressed higher levels of TNFα mRNA versus wild-type controls. In turn, mice bearing macrophage-specific deletion of TNFα exhibited decreased glomerular and tubular damage and attenuated kidney fibrosis in the models. Moreover, treatment with the TNF receptor-1 inhibitor R-7050 during nephrotoxic serum nephritis reduced damage, fibrosis, and necroptosis in wild-type mice and mice with KLF4-deficient macrophages, and abrogated the differences between the two groups in these parameters. CONCLUSIONS: These data indicate that macrophage KLF4 ameliorates CKD by mitigating TNF-dependent injury and fibrosis.


Asunto(s)
Enfermedades Renales/etiología , Riñón/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Macrófagos/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Fibrosis/etiología , Factor 4 Similar a Kruppel , Masculino , Ratones , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
18.
J Am Soc Nephrol ; 30(9): 1674-1685, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315922

RESUMEN

BACKGROUND: Following an acute insult, macrophages regulate renal fibrogenesis through the release of various factors that either encourage the synthesis of extracellular matrix synthesis or the degradation of matrix via endocytosis, proteolysis, or both. However, the roles of infiltrating versus resident myeloid cells in these opposing processes require elucidation. The transcription factor Twist1 controls diverse essential cellular functions through induction of several downstream targets, including matrix metalloproteinases (MMPs). In macrophages, Twist1 can influence patterns of cytokine generation, but the role of macrophage Twist1 in renal fibrogenesis remains undefined. METHODS: To study Twist1 functions in different macrophage subsets during kidney scar formation, we used two conditional mutant mouse models in which Twist1 was selectively ablated either in infiltrating, inflammatory macrophages or in resident tissue macrophages. We assessed fibrosis-related parameters, matrix metallopeptidase 13 (MMP13, or collagen 3, which catalyzes collagen degradation), inflammatory cytokines, and other factors in these Twist1-deficient mice compared with wild-type controls after subjecting the animals to unilateral ureteral obstruction. We also treated wild-type and Twist1-deficient mice with an MMP13 inhibitor after unilateral ureteral obstruction. RESULTS: Twist1 in infiltrating inflammatory macrophages but not in resident macrophages limited kidney fibrosis after ureteral obstruction by driving extracellular matrix degradation. Moreover, deletion of Twist1 in infiltrating macrophages attenuated the expression of MMP13 in CD11b+Ly6Clo myeloid cells. Inhibition of MMP13 abrogated the protection from renal fibrosis afforded by macrophage Twist1. CONCLUSIONS: Twist1 in infiltrating myeloid cells mitigates interstitial matrix accumulation in the injured kidney by promoting MMP13 production, which drives extracellular matrix degradation. These data highlight the complex cell-specific actions of Twist1 in the pathogenesis of kidney fibrosis.


Asunto(s)
Matriz Extracelular/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Riñón/patología , Macrófagos/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Actinas/metabolismo , Animales , Benzofuranos/farmacología , Receptor 1 de Quimiocinas CX3C/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica , Hidroxiprolina/metabolismo , Enfermedades Renales/etiología , Enfermedades Renales/patología , Macrófagos Peritoneales/metabolismo , Masculino , Metaloproteinasa 13 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Morfolinas/farmacología , Células Mieloides/enzimología , Proteína 1 Relacionada con Twist/genética , Obstrucción Ureteral/complicaciones
19.
Am J Physiol Renal Physiol ; 316(2): F351-F359, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30516426

RESUMEN

Acute kidney injury (AKI) is a common cause of morbidity and mortality in hospitalized patients. Nevertheless, there is limited ability to diagnose AKI in its earliest stages through the collection of structural and functional information. Magnetic resonance imaging (MRI) is increasingly being used to provide structural and functional data that characterize the injured kidney. Dynamic contrast-enhanced (DCE) MRI is an imaging modality with robust spatial and temporal resolution; however, its ability to detect changes in kidney function following AKI has not been determined. We hypothesized that DCE MRI would detect a prolongation in contrast transit time following toxin-induced AKI earlier than commonly used serum and tissue biomarkers. To test our hypothesis, we injected mice with either vehicle or cisplatin (30 mg/kg) and performed DCE MRI at multiple time points. We found that commonly used kidney injury biomarkers, including creatinine, blood urea nitrogen, and neutrophil gelatinase-associated lipocalin, did not rise until day 2 following cisplatin. Tissue levels of the proinflammatory cytokines and chemokines, tumor necrosis factor-α, interleukin (IL)-1ß, IL-1α, IL-6, C-C motif chemokine ligand 2, and C-X-C motif chemokine ligand 2 similarly did not upregulate until day 2 following cisplatin. However, the time to peak intensity of contrast in the renal collecting system was already prolonged at day 1 following cisplatin compared with vehicle-treated mice. This intensity change mirrored changes in kidney injury as measured by histological analysis and in transporter expression in the proximal tubule. Taken together, DCE MRI is a promising preclinical imaging modality that is useful for assessing functional capacity of the kidney in the earliest stages following AKI.


Asunto(s)
Lesión Renal Aguda/diagnóstico por imagen , Medios de Contraste/administración & dosificación , Gadolinio DTPA/administración & dosificación , Riñón/diagnóstico por imagen , Imagen por Resonancia Magnética , Lesión Renal Aguda/sangre , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Animales , Biomarcadores/sangre , Nitrógeno de la Urea Sanguínea , Cisplatino , Creatinina/sangre , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Diagnóstico Precoz , Femenino , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Ratones de la Cepa 129 , Valor Predictivo de las Pruebas , Factores de Tiempo
20.
Am J Physiol Renal Physiol ; 317(2): F221-F228, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31141402

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is associated with increased chemokines, cytokines, and growth factors in the diseased kidney. We found that both isoforms of IL-1, IL-1α and IL-1ß, were upregulated in ADPKD tissues. Here, we used a unique murine ADPKD model with selective deletion of polycystin-1 (pkd1) in the kidney (KPKD1) to study the role of IL-1 signaling in ADPKD progression. In KPKD mice, genetic deletion of the IL-1 receptor [IL-1 receptor (IL-1R) knockout (KO)] prolongs survival and attenuates cyst volume. Compared with IL-1R wild-type KPKD1 kidneys, IL-1R KO KPKD1 kidneys have upregulated TNF-α gene expression, with consequent elevations in markers for TNF-dependent regulated necrosis. We further observed that regulated necrosis was increased in ADPKD tissues from both humans and mice. To confirm that enhanced necroptosis is protective in ADPKD, we treated KPKD1 mice with an inhibitor of regulated necrosis (Nec-1). Regulated necrosis suppression augments kidney weights, suggesting that regulated necrosis is required to limit kidney growth in ADPKD. Thus, IL-1R activation drives ADPKD progression by paradoxically limiting regulated necrosis.


Asunto(s)
Riñón/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Riñón/patología , Ratones Noqueados , Necroptosis , Necrosis , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA