Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Kidney Int ; 103(3): 514-528, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36334787

RESUMO

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.


Assuntos
Injúria Renal Aguda , COVID-19 , Sepse , Camundongos , Animais , Células Endoteliais/patologia , COVID-19/complicações , Injúria Renal Aguda/patologia , Rim/patologia , Macrófagos/metabolismo , Interleucina-6/metabolismo , Sepse/complicações , Receptores de Interleucina-1/metabolismo , Camundongos Endogâmicos C57BL
3.
Compr Physiol ; 12(3): 4087-4101, 2022 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-35950656

RESUMO

The immune response plays a critical role in the pathogenesis of hypertension, and immune cell populations can promote blood pressure elevation via actions in the kidney. Among these cell lineages, dendritic cells (DCs), the most potent antigen-presenting cells, play a central role in regulating immune response during hypertension and kidney disease. DCs have different subtypes, and renal DCs are comprised of the CD103+ CD11b- and CD103- CD11b+ subsets. DCs become mature and express costimulatory molecules on their surface once they encounter antigen. Isolevuglandin-modified proteins function as antigens to activate DCs and trigger them to stimulate T cells. Activated T cells accumulate in the hypertensive kidney, release effector cytokines, promote renal oxidative stress, and promote renal salt and water retention. Individual subsets of activated T cells can secrete tumor necrosis factor-alpha, interleukin-17A, and interferon-gamma, each of which has augmented the elevation of blood pressure in hypertensive models by enhancing renal sodium transport. Fms-like tyrosine kinase 3 ligand-dependent classical DCs are required to sustain the full hypertensive response, but C-X3 -C chemokine receptor 1 positive DCs do not regulate blood pressure. Excess sodium enters the DC through transporters to activate DCs, whereas the ubiquitin editor A20 in dendritic cells constrains blood pressure elevation by limiting T cell activation. By contrast, activation of the salt sensing kinase, serum/glucocorticoid kinase 1 in DCs exacerbates salt-sensitive hypertension. This article discusses recent studies illustrating mechanisms through which DC-T cell interactions modulate levels of pro-hypertensive mediators to regulate blood pressure via actions in the kidney. © 2022 American Physiological Society. Compr Physiol 12:1-15, 2022.


Assuntos
Hipertensão , Pressão Sanguínea/fisiologia , Células Dendríticas/metabolismo , Humanos , Rim/metabolismo , Sódio/metabolismo
4.
Hypertension ; 79(6): 1227-1236, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35430875

RESUMO

BACKGROUND: Type 1 angiotensin (AT1) receptors are expressed on immune cells, and we previously found that bone marrow-derived AT1 receptors protect against Ang (angiotensin) II-induced hypertension. CD11c is expressed on myeloid cells derived from the bone marrow, including dendritic cells (DCs) that activate T lymphocytes. Here, we examined the role of AT1 receptors on CD11c+ cells in hypertension pathogenesis. METHODS: Mice lacking the dominant murine AT1 receptor isoform, AT1a, on CD11c+ cells (dendritic cell [DC] AT1aR knockout [KO]) and wild-type (WT) littermates were subjected to Ang II-induced hypertension. Blood pressures were measured by radiotelemetry. RESULTS: DC AT1aR KO mice had exaggerated hypertensive responses to chronic Ang II infusion with enhanced renal accumulation of effector memory T cells and CD40+ DCs. CCL5 (C-C motif chemokine ligand 5) recruits T cells into injured tissues, and CCR7 (C-C motif chemokine receptor 7) facilitates DC and T cell interactions in the kidney lymph node to allow T cell activation. DCs from the hypertensive DC AT1aR KO kidneys expressed higher levels of CCL5 and CCR7. mRNA expressions for CCR7 and tumor necrosis factor-α were increased in CD4+ T cells from the renal lymph nodes of DC AT1aR KO mice. During the second week of Ang II infusion when blood pressures between groups diverged, DC AT1aR KO mice excreted less sodium than WTs. Expressions for epithelial sodium channel subunits were increased in DC AT1aR KO kidneys. CONCLUSIONS: Following activation of the renin angiotensin system, AT1aR stimulation on DCs suppresses renal DC maturation and T cell activation with consequent protection from sodium retention and blood pressure elevation.


Assuntos
Hipertensão , Receptor Tipo 1 de Angiotensina , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Células Dendríticas/metabolismo , Hipertensão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores CCR7/metabolismo , Sódio/metabolismo , Linfócitos T/metabolismo
5.
Am J Physiol Renal Physiol ; 322(2): F164-F174, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34894725

RESUMO

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.


Assuntos
Glomerulonefrite/metabolismo , Podócitos/metabolismo , Proteinúria/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Doxorrubicina , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/patologia , Glomerulonefrite/prevenção & controle , Humanos , Interleucina-1beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos da Linhagem 129 , Camundongos Knockout , Podócitos/efeitos dos fármacos , Podócitos/patologia , Proteinúria/induzido quimicamente , Proteinúria/patologia , Proteinúria/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Tipo I de Interleucina-1/agonistas , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais
6.
JCI Insight ; 6(15)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34369383

RESUMO

The transcription factor Twist1 regulates several processes that could impact kidney disease progression, including epithelial cell differentiation and inflammatory cytokine induction. Podocytes are specialized epithelia that exhibit features of immune cells and could therefore mediate unique effects of Twist1 on glomerular disease. To study Twist1 functions in podocytes during proteinuric kidney disease, we employed a conditional mutant mouse in which Twist1 was selectively ablated in podocytes (Twist1-PKO). Deletion of Twist1 in podocytes augmented proteinuria, podocyte injury, and foot process effacement in glomerular injury models. Twist1 in podocytes constrained renal accumulation of monocytes/macrophages and glomerular expression of CCL2 and the macrophage cytokine TNF-α after injury. Deletion of TNF-α selectively from podocytes had no impact on the progression of proteinuric nephropathy. By contrast, the inhibition of CCL2 abrogated the exaggeration in proteinuria and podocyte injury accruing from podocyte Twist1 deletion. Collectively, Twist1 in podocytes mitigated urine albumin excretion and podocyte injury in proteinuric kidney diseases by limiting CCL2 induction that drove monocyte/macrophage infiltration into injured glomeruli. Myeloid cells, rather than podocytes, further promoted podocyte injury and glomerular disease by secreting TNF-α. These data highlight the capacity of Twist1 in the podocyte to mitigate glomerular injury by curtailing the local myeloid immune response.


Assuntos
Quimiocina CCL2/metabolismo , Células Mieloides/imunologia , Podócitos/metabolismo , Insuficiência Renal Crônica , Fator de Necrose Tumoral alfa/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Diferenciação Celular , Inativação Gênica , Imunidade/imunologia , Glomérulos Renais/imunologia , Glomérulos Renais/lesões , Glomérulos Renais/metabolismo , Macrófagos , Camundongos , Proteinúria/metabolismo , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
7.
Innate Immun ; 27(1): 50-60, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33241978

RESUMO

Classical collectins (surfactant protein A and D) play a significant role in innate immunity and host defence in uropathogenic Escherichia coli (UPEC)-induced urinary tract infection (UTI). However, the functions of collectin-11 (CL-11) with respect to UPEC and UTI remain largely unexplored. This study aimed to investigate the effect of CL-11 on UPEC and its role in UTI. We further examined its modulatory effect on inflammatory reactions in proximal tubular epithelial cells (PTECs). The present study provides evidence for the effect of CL-11 on the growth, agglutination, binding, epithelial adhesion and invasion of UPEC. We found increased basal levels of phosphorylated p38 MAPK and human cytokine homologue (keratinocyte-derived chemokine) expression in CL-11 knockdown PTECs. Furthermore, signal regulatory protein α blockade reversed the increased basal levels of inflammation associated with CL-11 knockdown in PTECs. Additionally, CL-11 knockdown effectively inhibited UPEC-induced p38 MAPK phosphorylation and cytokine production in PTECs. These were further inhibited by CD91 blockade. We conclude that CL-11 functions as a mediator of innate immunity via direct antibacterial roles as well as dual modulatory roles in UPEC-induced inflammatory responses during UTI. Thus, the study findings suggest a possible function for CL-11 in defence against UTI.


Assuntos
Colectinas/genética , Infecções por Escherichia coli/genética , Imunidade Inata/genética , Infecções Urinárias/genética , Animais , Atividade Bactericida do Sangue , Adesão Celular , Citocinas/genética , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Técnicas de Silenciamento de Genes , Túbulos Renais Proximais/imunologia , Túbulos Renais Proximais/microbiologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Infecções Urinárias/microbiologia , Infecções Urinárias/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética
8.
Curr Opin Nephrol Hypertens ; 29(5): 515-522, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32701602

RESUMO

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.


Assuntos
Hipertensão/imunologia , Rim/fisiopatologia , Imunidade Adaptativa , Animais , Citocinas/fisiologia , Células Dendríticas/imunologia , Humanos , Hipertensão/etiologia , Imunidade Inata
9.
Am J Physiol Renal Physiol ; 318(1): F107-F116, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31736350

RESUMO

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.


Assuntos
Fibrose/metabolismo , Glomerulonefrite/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Modelos Animais de Doenças , Fibrose/genética , Fibrose/patologia , Glomerulonefrite/genética , Glomerulonefrite/patologia , Interleucina-17/genética , Interleucina-17/metabolismo , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Knockout , Linfócitos T/patologia , Fator de Necrose Tumoral alfa/genética
10.
Kidney Int ; 97(1): 119-129, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31685313

RESUMO

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.


Assuntos
Túbulos Renais Distais/patologia , Macrófagos/imunologia , Nefrite Intersticial/imunologia , Insuficiência Renal Crônica/imunologia , Proteína 1 Relacionada a Twist/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Ácidos Aristolóquicos/toxicidade , Técnicas de Cocultura , Modelos Animais de Doenças , Células Epiteliais , Feminino , Fibrose , Técnicas de Silenciamento de Genes , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Humanos , Túbulos Renais Distais/citologia , Túbulos Renais Distais/imunologia , Túbulos Renais Distais/metabolismo , Lipocalina-2/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Nefrite Intersticial/induzido quimicamente , Nefrite Intersticial/patologia , Cultura Primária de Células , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/patologia , Proteína 1 Relacionada a Twist/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
11.
Circ Res ; 125(12): 1055-1066, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31630621

RESUMO

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.


Assuntos
Células Dendríticas/metabolismo , Hipertensão/metabolismo , Rim/metabolismo , Células Mieloides/metabolismo , Linfócitos T/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/deficiência , Animais , Células Cultivadas , Células Dendríticas/imunologia , Hipertensão/imunologia , Hipertensão/prevenção & controle , Rim/citologia , Rim/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/imunologia , Linfócitos T/imunologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/imunologia
12.
Kidney Int ; 96(6): 1308-1319, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31585741

RESUMO

Wnt/ß-catenin signaling is essential in the pathogenesis of renal fibrosis. We previously reported inhibition of the Wnt O-acyl transferase porcupine, required for Wnt secretion, dramatically attenuates kidney fibrosis in the murine unilateral ureteral obstruction model. Here, we investigated the tissue-specific contributions of porcupine to renal fibrosis and inflammation in ureteral obstruction using mice with porcupine deletion restricted to the kidney tubular epithelium or infiltrating myeloid cells. Obstruction of the ureter induced the renal mRNA expression of porcupine and downstream targets, ß-catenin, T-cell factor, and lymphoid enhancer factor in wild type mice. Renal tubular specific deficiency of porcupine reduced the expression of collagen I and other fibrosis markers in the obstructed kidney. Moreover, kidneys from obstructed mice with tubule-specific porcupine deficiency had reduced macrophage accumulation with attenuated expression of myeloid cytokine and chemokine mRNA. In co-culture with activated macrophages, renal tubular cells from tubular-specific porcupine knockout mice had blunted induction of fibrosis mediators compared with wild type renal tubular cells. In contrast, macrophages from macrophage-specific porcupine deficient mice in co-culture with wild type renal tubular cells had markedly enhanced expression of pro-fibrotic cytokines compared to wild type macrophages. Consequently, porcupine deletion specifically within macrophages augmented renal scar formation following ureteral obstruction. Thus, our experiments suggest a benefit of interrupting Wnt secretion specifically within the kidney epithelium while preserving Wnt O-acylation in infiltrating myeloid cells during renal fibrogenesis.


Assuntos
Aciltransferases/metabolismo , Proteínas de Membrana/metabolismo , Nefroesclerose/metabolismo , Via de Sinalização Wnt , Animais , Quimiocinas/metabolismo , Feminino , Fibrose , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo , Nefroesclerose/etiologia , Obstrução Ureteral
13.
J Am Soc Nephrol ; 30(10): 1925-1938, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31337692

RESUMO

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.


Assuntos
Nefropatias/etiologia , Rim/patologia , Fatores de Transcrição Kruppel-Like/fisiologia , Macrófagos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Fibrose/etiologia , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Fator de Necrose Tumoral alfa/antagonistas & inibidores
14.
J Am Soc Nephrol ; 30(9): 1674-1685, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31315922

RESUMO

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.


Assuntos
Matriz Extracelular/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Rim/patologia , Macrófagos/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Actinas/metabolismo , Animais , Benzofuranos/farmacologia , Receptor 1 de Quimiocina CX3C/metabolismo , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Fibrose , Expressão Gênica , Hidroxiprolina/metabolismo , Nefropatias/etiologia , Nefropatias/patologia , Macrófagos Peritoneais/metabolismo , Masculino , Metaloproteinase 13 da Matriz/genética , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos , Morfolinas/farmacologia , Células Mieloides/enzimologia , Proteína 1 Relacionada a Twist/genética , Obstrução Ureteral/complicações
15.
Am J Physiol Renal Physiol ; 317(2): F221-F228, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31141402

RESUMO

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.


Assuntos
Rim/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Rim/patologia , Camundongos Knockout , Necroptose , Necrose , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Receptores Tipo I de Interleucina-1/deficiência , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
JCI Insight ; 4(7)2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30944256

RESUMO

The antidiuretic hormone vasopressin (AVP), acting through its type 2 receptor (V2R) in the collecting duct (CD), critically controls urine concentrating capability. Here, we report that site-1 protease-derived (S1P-derived) soluble (pro)renin receptor (sPRR) participates in regulation of fluid homeostasis via targeting V2R. In cultured inner medullary collecting duct (IMCD) cells, AVP-induced V2R expression was blunted by a PRR antagonist, PRO20; a PRR-neutralizing antibody; or a S1P inhibitor, PF-429242. In parallel, sPRR release was increased by AVP and reduced by PF-429242. Administration of histidine-tagged sPRR, sPRR-His, stimulated V2R expression and also reversed the inhibitory effect of PF-429242 on the expression induced by AVP. PF-429242 treatment in C57/BL6 mice impaired urine concentrating capability, which was rescued by sPRR-His. This observation was recapitulated in mice with renal tubule-specific deletion of S1P. During the pharmacological or genetic manipulation of S1P alone or in combination with sPRR-His, the changes in urine concentration were paralleled with renal expression of V2R and aquaporin-2 (AQP2). Together, these results support that S1P-derived sPRR exerts a key role in determining renal V2R expression and, thus, urine concentrating capability.


Assuntos
Capacidade de Concentração Renal/fisiologia , Túbulos Renais Coletores/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Vasopressinas/metabolismo , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Aquaporina 2/genética , Células Cultivadas , Células Epiteliais , Capacidade de Concentração Renal/efeitos dos fármacos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Fragmentos de Peptídeos/farmacologia , Cultura Primária de Células , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Pirrolidinas/farmacologia , Ratos , Receptores de Vasopressinas/genética , Renina/metabolismo , Renina/farmacologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Urotélio/citologia , ATPases Vacuolares Próton-Translocadoras
17.
Curr Hypertens Rep ; 20(12): 103, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30377822

RESUMO

PURPOSE OF REVIEW: Low-grade inflammation drives elevations in blood pressure (BP) and consequent target organ damage in diverse experimental models of hypertension. Here, we discuss recent advances elucidating immune-mediated mechanisms of BP elevation and associated target organ damage. RECENT FINDINGS: Inflammatory mediators produced by immune cells or target organs act on the kidney, vasculature, skin, and nervous system to modulate hypertension. For example, cells of the innate immune system, including monocytes, neutrophils, and dendritic cells (DCs), can all promote BP elevation via actions in the vasculature and kidney. Macrophages expressing VEGF-C impact non-osmotic sodium storage in the skin that in turn regulates salt sensitivity. Within the adaptive immune system, 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 BP and renal damage in pre-clinical models. Inversely, deficiency of IL-17a in mice blunts the hypertensive response and attenuates renal sodium retention via a serum- and glucocorticoid-regulated kinase 1 (SGK1)-dependent pathway. Linking innate and adaptive immune responses, dendritic cells activated by augmented extracellular sodium concentrations stimulate T lymphocytes to produce pro-hypertensive cytokines. By contrast, regulatory T cells (Tregs) can protect against hypertension and associated kidney injury. Rodent studies reveal diverse mechanisms via which cells of the innate and adaptive immune systems drive blood pressure elevation by altering the inflammatory milieu in the kidney, vasculature, and brain.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão , Inflamação/imunologia , Rim , Cloreto de Sódio/metabolismo , Animais , Humanos , Hipertensão/imunologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Rim/metabolismo , Rim/fisiopatologia , Camundongos
18.
Am J Physiol Renal Physiol ; 315(3): F682-F691, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29790392

RESUMO

Acute kidney injury (AKI) is a leading cause of morbidity and mortality. Drug-induced/toxic AKI can be caused by a number of therapeutic agents. Cisplatin is an effective chemotherapeutic agent whose administration is limited by significant nephrotoxicity. Therapies to prevent cisplatin-induced AKI are lacking. Although tumor necrosis factor-α (TNF) plays a key role in the pathogenesis of cisplatin nephrotoxicity, the innate immune signaling pathways that trigger TNF generation in this context require elucidation. In this regard, sterile injury triggers the release and activation of both isoforms of interleukin(IL)-1, IL-1α and IL-1ß. In turn, stimulation of the interleukin-1 receptor (IL-1R1) by these ligands engages a proinflammatory signaling cascade that induces TNF induction. We therefore hypothesized that IL-1R1 activation exacerbates cisplatin-induced AKI by inducing TNF production, thereby augmenting inflammatory signals between kidney parenchymal cells and infiltrating myeloid cells. IL-1R1+/+ (WT) and IL-1R1-/- (KO) mice were subjected to cisplatin-induced AKI. Compared with WT mice, IL-1R1 KO mice had attenuated AKI as measured by serum creatinine and BUN, renal NGAL mRNA levels, and blinded histological analysis of kidney pathology. In the cisplatin-injured kidney, IL-1R1 KO mice had diminished levels of whole kidney TNF, and fewer Ly6G-expressing neutrophils. In addition, an unbiased machine learning analysis of intrarenal immune cells revealed a diminished number of CD11bint/CD11cint myeloid cells in IL-1R1 KO injured kidneys compared with IL-1R1 WT kidneys. Following cisplatin, IL-1R1 KO kidneys, compared with WTs, had fewer TNF-producing: macrophages, CD11bint/CD11cint cells, and neutrophils, consistent with an effect of IL-1R1 to polarize intrarenal myeloid cells toward a proinflammatory phenotype. Interruption of IL-1-dependent signaling pathways warrants further evaluation to decrease nephrotoxicity during cisplatin therapy.


Assuntos
Injúria Renal Aguda/metabolismo , Cisplatino , Rim/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Animais , Comunicação Celular , Separação Celular/métodos , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Mediadores da Inflamação/metabolismo , Rim/patologia , Aprendizado de Máquina , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores Tipo I de Interleucina-1/deficiência , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais , Processos Estocásticos , Fator de Necrose Tumoral alfa/metabolismo
19.
Hypertension ; 69(2): 339-348, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27993957

RESUMO

A high-fructose diet is shown to induce salt-sensitive hypertension, but the underlying mechanism largely remains unknown. The major goal of the present study was to test the role of renal (pro)renin receptor (PRR) in this model. In Sprague-Dawley rats, high-fructose intake increased renal expression of full-length PRR, which were attenuated by allopurinol. High-fructose intake also upregulated renal mRNA and protein expression of sodium/hydrogen exchanger 3 and Na/K/2Cl cotransporter, as well as in vivo Na/K/2Cl cotransporter activity, all of which were nearly completely blocked by a PRR decoy inhibitor PRO20 or allopurinol treatment. Parallel changes were observed for indices of intrarenal renin-angiotensin-system including renal and urinary renin and angiotensin II levels. Radiotelemetry demonstrated that high-fructose or a high-salt diet alone did not affect mean arterial pressure, but the combination of the 2 maneuvers induced a ≈10-mm Hg increase of mean arterial pressure, which was blunted by PRO20 or allopurinol treatment. In cultured human kidney 2 cells, both fructose and uric acid increased protein expression of soluble PRR in a time- and dose-dependent manner; fructose-induced PRR upregulation was inhibited by allopurinol. Taken together, our data suggest that fructose via uric acid stimulates renal expression of PRR/soluble PRR that stimulate sodium/hydrogen exchanger 3 and Na/K/2Cl cotransporter expression and intrarenal renin-angiotensin system to induce salt-sensitive hypertension.


Assuntos
Alopurinol/farmacologia , Hipertensão/metabolismo , Receptores de Superfície Celular/biossíntese , Sistema Renina-Angiotensina/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Frutose/efeitos adversos , Frutose/toxicidade , Humanos , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Rim/metabolismo , Rim/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/efeitos dos fármacos , Receptor de Pró-Renina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA