Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Am J Physiol Renal Physiol ; 326(2): F167-F177, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37969103

RESUMO

This study aimed to investigate the role of bone marrow stromal cell antigen-1 (Bst1; also known as CD157) in acute kidney injury (AKI). Bst1 is a cell surface molecule with various enzymatic activities and downstream intracellular signaling pathways that modulate the immune response. Previous research has linked Bst1 to diseases such as ovarian cancer, Parkinson's disease, and rheumatoid arthritis. We used bilateral ischemia-reperfusion injury (IRI) as an AKI model and created bone marrow chimeric mice to evaluate the role of Bst1 in bone marrow-derived cells. We also used flow cytometry to identify Bst1/CD157 expression in hematopoietic cells and evaluate immune cell dynamics in the kidney. The findings showed that Bst1-deficient (Bst1-/-) mice were protected against renal bilateral IRI. Bone marrow chimera experiments revealed that Bst1 expression on hematopoietic cells, but not parenchymal cells, induced renal IRI. Bst1 was mainly found in B cells and neutrophils by flow cytometry of the spleen and bone marrow. In vitro, migration of neutrophils from Bst1-/- mice was suppressed, and adoptive transfer of neutrophils from wild-type Bst1+/+ mice abolished the renal protective effect in Bst1 knockout mice. In conclusion, the study demonstrated that Bst1-/- mice are protected against renal IRI and that Bst1 expression in neutrophils plays a crucial role in inducing renal IRI. These findings suggest that targeting Bst1 in neutrophils could be a potential therapeutic strategy for AKI.NEW & NOTEWORTHY Acute kidney injury (AKI), a serious disease for which there is no effective Federal Drug Administration-approved treatment, is associated with high mortality rates. Bone marrow stromal cell antigen-1 (Bst1) is a cell surface molecule that can cause kidney fibrosis, but its role in AKI is largely unknown. Our study showed that Bst1-/- mice revealed a protective effect against renal bilateral ischemia-reperfusion injury (IRI). Adoptive transfer studies confirmed that Bst1 expression in hematopoietic cells, especially neutrophils, contributed to renal bilateral IRI.


Assuntos
Injúria Renal Aguda , Células-Tronco Mesenquimais , Traumatismo por Reperfusão , Camundongos , Animais , Injúria Renal Aguda/genética , Injúria Renal Aguda/prevenção & controle , Rim/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/prevenção & controle , Neutrófilos/metabolismo , Camundongos Knockout , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL
2.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33737395

RESUMO

Acute kidney injury is highly prevalent and associated with high morbidity and mortality, and there are no approved drugs for its prevention and treatment. Vagus nerve stimulation (VNS) alleviates inflammatory diseases including kidney disease; however, neural circuits involved in VNS-induced tissue protection remain poorly understood. The vagus nerve, a heterogeneous group of neural fibers, innervates numerous organs. VNS broadly stimulates these fibers without specificity. We used optogenetics to selectively stimulate vagus efferent or afferent fibers. Anterograde efferent fiber stimulation or anterograde (centripetal) sensory afferent fiber stimulation both conferred kidney protection from ischemia-reperfusion injury. We identified the C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis as the downstream pathway of vagus afferent fiber stimulation. Our study provides a map of the neural circuits important for kidney protection induced by VNS, which is critical for the safe and effective clinical application of VNS for protection from acute kidney injury.


Assuntos
Injúria Renal Aguda/etiologia , Suscetibilidade a Doenças , Neuroimunomodulação , Baço/imunologia , Baço/inervação , Estimulação do Nervo Vago , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Camundongos , Neurônios , Sistema Nervoso Simpático/fisiologia
3.
Kidney Int ; 100(3): 613-620, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34224760

RESUMO

Microcirculatory changes and oxidative stress have long been associated with acute kidney injury. Despite substantial progress made by two-photon microscopy of microvascular responses to acute kidney injury in rodent models, little is known about the underlying changes in blood oxygen delivery and tissue oxygen metabolism. To fill this gap, we developed a label-free kidney imaging technique based on photoacoustic microscopy, which enables simultaneous quantification of hemoglobin concentration, oxygen saturation of hemoglobin, and blood flow in peritubular capillaries in vivo. Based on these microvascular parameters, microregional oxygen metabolism was quantified. We demonstrated the utility of this technique by studying kidney hemodynamic and oxygen-metabolic responses to acute kidney injury in mice subject to lipopolysaccharide-induced sepsis. Dynamic photoacoustic microscopy of the peritubular capillary function and tissue oxygen metabolism revealed that sepsis induced an acute and significant reduction in peritubular capillary oxygen saturation of hemoglobin, concomitant with a marked reduction in kidney ATP levels and contrasted with nominal changes in peritubular capillary flow and plasma creatinine. Thus, our technique opens new opportunities to study microvascular and metabolic dysfunction in acute and chronic kidney diseases.


Assuntos
Capilares , Microscopia , Animais , Rim , Camundongos , Microcirculação , Oxigênio
4.
Am J Physiol Renal Physiol ; 317(3): F658-F669, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31364375

RESUMO

Progressive tubulointerstitial fibrosis may occur after acute kidney injury due to persistent inflammation. Purinergic signaling by 5'-ectonucleotidase, CD73, an enzyme that converts AMP to adenosine on the extracellular surface, can suppress inflammation. The role of CD73 in progressive kidney fibrosis has not been elucidated. We evaluated the effect of deletion of CD73 from kidney perivascular cells (including pericytes and/or fibroblasts of the Foxd1+ lineage) on fibrosis. Perivascular cell expression of CD73 was necessary to suppress inflammation and prevent kidney fibrosis in Foxd1CreCD73fl/fl mice evaluated 14 days after unilateral ischemia-reperfusion injury or folic acid treatment (250 mg/kg). Kidneys of Foxd1CreCD73fl/fl mice had greater collagen deposition, expression of proinflammatory markers (including various macrophage markers), and platelet-derived growth factor recepetor-ß immunoreactivity than CD73fl/fl mice. Kidney dysfunction and fibrosis were rescued by administration of soluble CD73 or by macrophage deletion. Isolated CD73-/- kidney pericytes displayed an activated phenotype (increased proliferation and α-smooth muscle actin mRNA expression) compared with wild-type controls. In conclusion, CD73 in perivascular cells may act to suppress myofibroblast transformation and influence macrophages to promote a wound healing response. These results suggest that the purinergic signaling pathway in the kidney interstitial microenvironment orchestrates perivascular cells and macrophages to suppress inflammation and prevent progressive fibrosis.


Assuntos
5'-Nucleotidase/metabolismo , Microambiente Celular , Fibroblastos/metabolismo , Rim/metabolismo , Macrófagos/metabolismo , Nefrite Intersticial/metabolismo , Pericitos/metabolismo , Traumatismo por Reperfusão/metabolismo , 5'-Nucleotidase/deficiência , 5'-Nucleotidase/genética , Actinas/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Fibroblastos/patologia , Fibrose , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Mediadores da Inflamação/metabolismo , Rim/imunologia , Rim/patologia , Macrófagos/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite Intersticial/genética , Nefrite Intersticial/imunologia , Nefrite Intersticial/patologia , Pericitos/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Cicatrização
5.
Kidney Int ; 95(3): 563-576, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30670317

RESUMO

The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic ß2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including ß2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.


Assuntos
Injúria Renal Aguda/imunologia , Macrófagos Peritoneais/imunologia , Traumatismo por Reperfusão/imunologia , Fatores de Transcrição HES-1/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/efeitos da radiação , Linfócitos T CD4-Positivos/transplante , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/transplante , Masculino , Camundongos , Neuroimunomodulação/efeitos da radiação , Células RAW 264.7 , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/imunologia , Terapia por Ultrassom , Regulação para Cima/efeitos da radiação , Estimulação do Nervo Vago , Receptor Nicotínico de Acetilcolina alfa7/imunologia
6.
J Am Soc Nephrol ; 29(1): 194-206, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29084809

RESUMO

The proximal tubule epithelium relies on mitochondrial function for energy, rendering the kidney highly susceptible to ischemic AKI. Dynamin-related protein 1 (DRP1), a mediator of mitochondrial fission, regulates mitochondrial function; however, the cell-specific and temporal role of DRP1 in AKI in vivo is unknown. Using genetic murine models, we found that proximal tubule-specific deletion of Drp1 prevented the renal ischemia-reperfusion-induced kidney injury, inflammation, and programmed cell death observed in wild-type mice and promoted epithelial recovery, which associated with activation of the renoprotective ß-hydroxybutyrate signaling pathway. Loss of DRP1 preserved mitochondrial structure and reduced oxidative stress in injured kidneys. Lastly, proximal tubule deletion of DRP1 after ischemia-reperfusion injury attenuated progressive kidney injury and fibrosis. These results implicate DRP1 and mitochondrial dynamics as an important mediator of AKI and progression to fibrosis and suggest that DRP1 may serve as a therapeutic target for AKI.


Assuntos
Ácido 3-Hidroxibutírico/metabolismo , Injúria Renal Aguda/genética , Dinaminas/genética , Túbulos Renais Proximais/patologia , Mitocôndrias/metabolismo , Insuficiência Renal Crônica/genética , Injúria Renal Aguda/etiologia , Animais , Apoptose/genética , Progressão da Doença , Dinaminas/antagonistas & inibidores , Fibrose , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/ultraestrutura , Dinâmica Mitocondrial/genética , Nefrite/etiologia , Nefrite/genética , Estresse Oxidativo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Traumatismo por Reperfusão/complicações , Transdução de Sinais
7.
J Am Soc Nephrol ; 29(7): 1887-1899, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29866797

RESUMO

Background Pannexin1 (Panx1), an ATP release channel, is present in most mammalian tissues, but the role of Panx1 in health and disease is not fully understood. Panx1 may serve to modulate AKI; ATP is a precursor to adenosine and may function to block inflammation, or ATP may act as a danger-associated molecular pattern and initiate inflammation.Methods We used pharmacologic and genetic approaches to evaluate the effect of Panx1 on kidney ischemia-reperfusion injury (IRI), a mouse model of AKI.Results Pharmacologic inhibition of gap junctions, including Panx1, by administration of carbenoxolone protected mice from IRI. Furthermore, global deletion of Panx1 preserved kidney function and morphology and diminished the expression of proinflammatory molecules after IRI. Analysis of bone marrow chimeric mice revealed that Panx1 expressed on parenchymal cells is necessary for ischemic injury, and both proximal tubule and vascular endothelial Panx1 tissue-specific knockout mice were protected from IRI. In vitro, Panx1-deficient proximal tubule cells released less and retained more ATP under hypoxic stress.Conclusions Panx1 is involved in regulating ATP release from hypoxic cells, and reducing this ATP release may protect kidneys from AKI.


Assuntos
Injúria Renal Aguda/metabolismo , Conexinas/antagonistas & inibidores , Conexinas/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Trifosfato de Adenosina/metabolismo , Animais , Antiulcerosos/farmacologia , Células da Medula Óssea/metabolismo , Carbenoxolona/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular , Células Epiteliais/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle
8.
J Am Soc Nephrol ; 28(9): 2681-2693, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28539382

RESUMO

CD4+Foxp3+ regulatory T cells (Tregs) protect the kidney during AKI. We previously found that IL-2, which is critical for Treg homeostasis, upregulates the IL-33 receptor (ST2) on CD4+ T cells, thus we hypothesized that IL-2 and IL-33 cooperate to enhance Treg function. We found that a major subset of Tregs in mice express ST2, and coinjection of IL-2 and IL-33 increased the number of Tregs in lymphoid organs and protected mice from ischemia-reperfusion injury (IRI) more efficiently than either cytokine alone. Accordingly, we generated a novel hybrid cytokine (IL233) bearing the activities of IL-2 and IL-33 for efficient targeting to Tregs. IL233 treatment increased the number of Tregs in blood and spleen and prevented IRI more efficiently than a mixture of IL-2 and IL-33. Injection of IL233 also increased the numbers of Tregs in renal compartments. Moreover, IL233-treated mice had fewer splenic Tregs and more Tregs in kidneys after IRI. In vitro, splenic Tregs from IL233-treated mice suppressed CD4+ T cell proliferation better than Tregs from saline-treated controls. IL233 treatment also improved the ability of isolated Tregs to inhibit IRI in adoptive transfer experiments and protected mice from cisplatin- and doxorubicin-induced nephrotoxic injury. Finally, treatment with IL233 increased the proportion of ST2-bearing innate lymphoid cells (ILC2) in blood and kidneys, and adoptive transfer of ILC2 also protected mice from IRI. Thus, the novel IL233 hybrid cytokine, which utilizes the cooperation of IL-2 and IL-33 to enhance Treg- and ILC2-mediated protection from AKI, bears strong therapeutic potential.


Assuntos
Injúria Renal Aguda/imunologia , Injúria Renal Aguda/prevenção & controle , Interleucina-2/farmacologia , Interleucina-33/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/prevenção & controle , Linfócitos T Reguladores/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Animais , Contagem de Linfócito CD4 , Proliferação de Células , Células Cultivadas , Cisplatino/efeitos adversos , Técnicas de Cocultura , Doxorrubicina/efeitos adversos , Proteína 1 Semelhante a Receptor de Interleucina-1/sangue , Interleucina-2/uso terapêutico , Interleucina-33/uso terapêutico , Rim/imunologia , Masculino , Camundongos , Proteínas Recombinantes de Fusão/uso terapêutico , Baço/imunologia
9.
J Am Soc Nephrol ; 28(3): 888-902, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27628903

RESUMO

CD73-derived adenosine plays an anti-inflammatory role in various organs. However, its role in renal ischemia-reperfusion injury (IRI) is controversial. We targeted CD73 mutant mice to determine the function of CD73 expressed by various renal cell types under mild IRI conditions. Mice with CD73 deletion in proximal tubules exhibited exacerbated IRI, comparable with that of CD73-/- mice compared with WT mice. Mice with CD73 deletions in other cell types, including cortical type 1 fibroblast-like cells, mesangial cells, macrophages, and dendritic cells, showed small or no increases in injury above control mice when subjected to threshold levels of ischemia. Results from adoptive transfer experiments between WT and CD73-/- mice and pharmacologic studies modulating enzymatic activity of CD73 and extracellular adenosine levels supported a critical role of adenosine generated by proximal tubule CD73 expression in abrogating IRI. Renal adenosine levels were lower before and after ischemia in CD73-deficient mice. However, reduction in total acid-extractable renal adenosine levels was inadequate to explain the marked difference in kidney injury in these CD73-deficient mice. Furthermore, CD73 inhibition and enzyme replacement studies showed no change in total kidney adenosine levels in treated mice compared with vehicle-treated controls. Protection from IRI in neutrophil-depleted WT recipients was sustained by repopulation with bone marrow neutrophils from WT mice but not by those lacking adenosine 2a receptors (from Adora2a-/- mice). These data support the thesis that local adenosine generated by cells at the injury site is critical for protection from IRI through bone marrow-derived adenosine 2a receptors.


Assuntos
5'-Nucleotidase/fisiologia , Rim/irrigação sanguínea , Traumatismo por Reperfusão/etiologia , Animais , Células Cultivadas , Túbulos Renais Proximais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
J Am Soc Nephrol ; 28(4): 1145-1161, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27799486

RESUMO

Maladaptive repair after AKI may lead to progressive fibrosis and decline in kidney function. Sphingosine 1-phosphate has an important role in kidney injury and pleiotropic effects in fibrosis. We investigated the involvement of sphingosine kinase 1 and 2 (SphK1 and SphK2), which phosphorylate sphingosine to produce sphingosine 1-phosphate, in kidney fibrosis induced by folic acid (FA) or unilateral ischemia-reperfusion injury. Analysis of Masson trichrome staining and fibrotic marker protein and mRNA expression 14 days after AKI revealed that wild-type (WT) and Sphk1-/- mice exhibited more kidney fibrosis than Sphk2-/- mice. Furthermore, kidneys of FA-treated WT and Sphk1-/- mice had greater immune cell infiltration and expression of fibrotic and inflammatory markers than kidneys of FA-treated Sphk2-/- mice. In contrast, kidneys of Sphk2-/- mice exhibited greater expression of Ifng and IFN-γ-responsive genes (Cxcl9 and Cxcl10) than kidneys of WT or Sphk1-/- mice did at this time point. Splenic T cells from untreated Sphk2-/- mice were hyperproliferative and produced more IFN-γ than did those of WT or Sphk1-/- mice. IFN-γ blocking antibody administered to Sphk2-/- mice or deletion of Ifng (Sphk2-/-Ifng-/- mice) blocked the protective effect of SphK2 deficiency in fibrosis. Moreover, adoptive transfer of Sphk2-/- (but not Sphk2-/-Ifng-/- ) CD4 T cells into WT mice blocked FA-induced fibrosis. Finally, a selective SphK2 inhibitor blocked FA-induced kidney fibrosis in WT mice. These studies demonstrate that SphK2 inhibition may serve as a novel therapeutic approach for attenuating kidney fibrosis.


Assuntos
Interferon gama/fisiologia , Nefropatias/enzimologia , Rim/enzimologia , Rim/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Animais , Fibrose/enzimologia , Fibrose/etiologia , Fibrose/prevenção & controle , Nefropatias/prevenção & controle , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores
11.
J Am Soc Nephrol ; 27(11): 3383-3393, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26961351

RESUMO

Epithelial and endothelial injury and a cascade of immune and interstitial cell activation in the kidney lead to AKI. After mild to moderate AKI, the epithelium can regenerate and restore kidney function, yet little is known about the endothelium during these repair processes. Sphingosine 1-phosphate receptor 1 (S1P1), a G protein-coupled receptor, is necessary for vascular homeostasis. Here, we used an inducible genetic approach in a mouse model of AKI, ischemia-reperfusion injury (IRI), to determine the temporal effects of endothelial S1P1 during AKI. Deletion of endothelial S1P1 before IRI exacerbated kidney injury and inflammation, and the delayed deletion of S1P1 after IRI prevented kidney recovery, resulting in chronic inflammation and progressive fibrosis. Specifically, S1P1 directly suppressed endothelial activation of leukocyte adhesion molecule expression and inflammation. Altogether, the data indicate activation of endothelial S1P1 is necessary to protect from IRI and permit recovery from AKI. Endothelial S1P1 may be a therapeutic target for the prevention of early injury as well as prevention of progressive kidney fibrosis after AKI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Receptores de Lisoesfingolipídeo/fisiologia , Receptores de Lisoesfingolipídeo/uso terapêutico , Animais , Endotélio , Fibrose/prevenção & controle , Rim/irrigação sanguínea , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Recuperação de Função Fisiológica , Traumatismo por Reperfusão/prevenção & controle
12.
J Am Soc Nephrol ; 27(4): 1076-90, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26286732

RESUMO

The plasticity of dendritic cells (DCs) permits phenotypic modulation ex vivo by gene expression or pharmacologic agents, and these modified DCs can exert therapeutic immunosuppressive effects in vivo through direct interactions with T cells, either inducing T regulatory cells (T(REG)s) or causing anergy. Sphingosine 1-phosphate (S1P) is a sphingolipid and the natural ligand for five G protein-coupled receptors (S1P1, S1P2, S1P3, S1P4, and S1P5), and S1PR agonists reduce kidney ischemia-reperfusion injury (IRI) in mice. S1pr3(-/-)mice are protected from kidney IRI, because DCs do not mature. We tested the therapeutic advantage of S1pr3(-/-) bone marrow-derived dendritic cell (BMDC) transfers in kidney IRI. IRI produced a rise in plasma creatinine (PCr) levels in mice receiving no cells (NCs) and mice pretreated with wild-type (WT) BMDCs. However, S1pr3(-/-) BMDC-pretreated mice were protected from kidney IRI. S1pr3(-/-) BMDC-pretreated mice had significantly higher numbers of splenic T(REG)s compared with NC and WT BMDC-pretreated mice. S1pr3(-/-) BMDCs did not attenuate IRI in splenectomized, Rag-1(-/-), or CD11c(+) DC-depleted mice. Additionally, S1pr3(-/-) BMDC-dependent protection required CD169(+)marginal zone macrophages and the macrophage-derived chemokine CCL22 to increase splenic CD4(+)Foxp3(+) T(REG)s. Pretreatment with S1pr3(-/-) BMDCs also induced T(REG)-dependent protection against IRI in an allogeneic mouse model. In summary, adoptively transferred S1pr3(-/-) BMDCs prevent kidney IRI through interactions within the spleen and expansion of splenic CD4(+)Foxp3(+) T(REG)s. We conclude that genetically induced deficiency of S1pr3 in allogenic BMDCs could serve as a therapeutic approach to prevent IRI-induced AKI.


Assuntos
Células Dendríticas , Rim/irrigação sanguínea , Receptores de Lisoesfingolipídeo/deficiência , Traumatismo por Reperfusão/fisiopatologia , Baço/fisiopatologia , Animais , Transplante de Medula Óssea , Células Dendríticas/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão/cirurgia
13.
Kidney Int ; 90(3): 462-5, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27521104

RESUMO

The cholinergic anti-inflammatory pathway has been shown to modulate inflammation in disease models such as rheumatoid arthritis and inflammatory bowel disease. A recent study demonstrated a protective effect of vagus nerve stimulation with activation of the cholinergic anti-inflammatory pathway in the ischemia reperfusion model of acute kidney injury.


Assuntos
Inflamação , Estimulação do Nervo Vago , Acetilcolina , Injúria Renal Aguda , Distrofias Hereditárias da Córnea , Humanos , Traumatismo por Reperfusão
14.
J Am Soc Nephrol ; 26(11): 2800-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25788528

RESUMO

Iron-mediated oxidative stress is implicated in the pathogenesis of renal ischemia-reperfusion injury. Hepcidin is an endogenous acute phase hepatic hormone that prevents iron export from cells by inducing degradation of the only known iron export protein, ferroportin. In this study, we used a mouse model to investigate the effect of renal ischemia-reperfusion injury on systemic iron homeostasis and determine if dynamic modulation of iron homeostasis with hepcidin has therapeutic benefit in the treatment of AKI. Renal ischemia-reperfusion injury induced hepatosplenic iron export through increased ferroportin expression, which resulted in hepatosplenic iron depletion and an increase in serum and kidney nonheme iron levels. Exogenous hepcidin treatment prevented renal ischemia-reperfusion-induced changes in iron homeostasis. Hepcidin also decreased kidney ferroportin expression and increased the expression of cytoprotective H-ferritin. Hepcidin-induced restoration of iron homeostasis was accompanied by a significant reduction in ischemia-reperfusion-induced tubular injury, apoptosis, renal oxidative stress, and inflammatory cell infiltration. Hepcidin -: deficient mice demonstrated increased susceptibility to ischemia-reperfusion injury compared with wild-type mice. Reconstituting hepcidin-deficient mice with exogenous hepcidin induced hepatic iron sequestration, attenuated the reduction in renal H-ferritin and reduced renal oxidative stress, apoptosis, inflammation, and tubular injury. Hepcidin-mediated protection was associated with reduced serum IL-6 levels. In summary, renal ischemia-reperfusion injury results in profound alterations in systemic iron homeostasis. Hepcidin treatment restores iron homeostasis and reduces inflammation to mediate protection in renal ischemia-reperfusion injury, suggesting that hepcidin-ferroportin pathway holds promise as a novel therapeutic target in the treatment of AKI.


Assuntos
Apoptose , Hepcidinas/química , Ferro/química , Rim/patologia , Traumatismo por Reperfusão/patologia , Injúria Renal Aguda/patologia , Animais , Regulação da Expressão Gênica , Hepcidinas/sangue , Homeostase , Marcação In Situ das Extremidades Cortadas , Inflamação , Interleucina-6/metabolismo , Ferro/sangue , Rim/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Estresse Oxidativo , Baço/citologia , Baço/patologia
15.
J Am Soc Nephrol ; 26(10): 2470-81, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25644106

RESUMO

We showed previously that prior exposure to a modified ultrasound regimen prevents kidney ischemia-reperfusion injury (IRI) likely via the splenic cholinergic anti-inflammatory pathway (CAP) and α7 nicotinic acetylcholine receptors (α7nAChR). However, it is unclear how ultrasound stimulates the splenic CAP. Further investigating the role of the spleen in ischemic injury, we found that prior splenectomy (-7d) or chemical sympathectomy of the spleen with 6-hydroxydopamine (6OHDA; -14d) exacerbated injury after subthreshold (24-minute ischemia) IRI. 6-OHDA-induced splenic denervation also prevented ultrasound-induced protection of kidneys from moderate (26-minute ischemia) IRI. Ultrasound-induced protection required hematopoietic but not parenchymal α7nAChRs, as shown by experiments in bone marrow chimeras generated with wild-type and α7nAChR(-/-) mice. Ultrasound protection was associated with reduced expression of circulating and kidney-derived cytokines. However, splenocytes isolated from mice 24 hours after ultrasound treatment released more IL-6 ex vivo in response to LPS than splenocytes from sham mice. Adoptive transfer of splenocytes from ultrasound-treated (but not sham) mice to naïve mice was sufficient to protect kidneys of recipient mice from IRI. Ultrasound treatment 24 hours before cecal ligation puncture-induced sepsis was effective in reducing plasma creatinine in this model of AKI. Thus, splenocytes of ultrasound-treated mice are capable of modulating IRI in vivo, supporting our ongoing hypothesis that a modified ultrasound regimen has therapeutic potential for AKI and other inflammatory conditions.


Assuntos
Injúria Renal Aguda/imunologia , Injúria Renal Aguda/prevenção & controle , Neuroimunomodulação/efeitos da radiação , Baço/imunologia , Baço/efeitos da radiação , Terapia por Ultrassom , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
J Am Soc Nephrol ; 26(4): 908-25, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25145931

RESUMO

Sphingosine 1-phosphate (S1P), the natural sphingolipid ligand for a family of five G protein- coupled receptors (S1P1-S1P5Rs), regulates cell survival and lymphocyte circulation. We have shown that the pan-S1PR agonist, FTY720, attenuates kidney ischemia-reperfusion injury by directly activating S1P1 on proximal tubule (PT) cells, independent of the canonical lymphopenic effects of S1P1 activation on B and T cells. FTY720 also reduces cisplatin-induced AKI. Therefore, in this study, we used conditional PT-S1P1-null (PepckCreS1pr1(fl/fl)) and control (PepckCreS1pr1(w/wt)) mice to determine whether the protective effect of FTY720 in AKI is mediated by PT-S1P1. Cisplatin induced more renal injury in PT-S1P1-null mice than in controls. Although FTY720 produced lymphopenia in both control and PT-S1P1-null mice, it reduced injury only in control mice. Furthermore, the increase in proinflammatory cytokine (CXCL1, MCP-1, TNF-α, and IL-6) expression and infiltration of neutrophils and macrophages induced by cisplatin treatment was attenuated by FTY720 in control mice but not in PT-S1P1-null mice. Similarly, S1P1 deletion rendered cultured PT cells more susceptible to cisplatin-induced injury, whereas S1P1 overexpression protected PT cells from injury and preserved mitochondrial function. We conclude that S1P1 may have an important role in stabilizing mitochondrial function and that FTY720 administration represents a novel strategy in the prevention of cisplatin-induced AKI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Túbulos Renais Proximais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Propilenoglicóis/uso terapêutico , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Injúria Renal Aguda/induzido quimicamente , Animais , Apoptose/efeitos dos fármacos , Respiração Celular , Cisplatino , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Cloridrato de Fingolimode , Masculino , Camundongos Endogâmicos C57BL , Dinâmica Mitocondrial , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/farmacologia , Esfingosina/uso terapêutico
17.
J Immunol ; 189(5): 2584-96, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22855711

RESUMO

Dendritic cells (DCs) are central to innate and adaptive immunity of early kidney ischemia-reperfusion injury (IRI), and strategies to alter DC function may provide new therapeutic opportunities. Sphingosine 1-phosphate (S1P) modulates immunity through binding to its receptors (S1P1-5), and protection from kidney IRI occurs in S1P3-deficient mice. Through a series of experiments we determined that this protective effect was owing in part to differences between S1P3-sufficient and -deficient DCs. Mice lacking S1P3 on bone marrow cells were protected from IRI, and S1P3-deficient DCs displayed an immature phenotype. Wild-type (WT) but not S1P3-deficient DCs injected into mice depleted of DCs prior to kidney IR reconstituted injury. Adoptive transfer (i.e., i.v. injection) of glycolipid (Ag)-loaded WT but not S1P3-deficient DCs into WT mice exacerbated IRI, suggesting that WT but not S1P3-deficient DCs activated NKT cells. Whereas WT DC transfers activated the Th1/IFN-γ pathway, S1P3-deficient DCs activated the Th2/IL-4 pathway, and an IL-4-blocking Ab reversed protection from IRI, supporting the concept that IL-4 mediates the protective effect of S1P3-deficient DCs. Administration of S1P3-deficient DCs 7 d prior to or 3 h after IRI protected mice from IRI and suggests their potential use in cell-based therapy. We conclude that absence of DC S1P3 prevents DC maturation and promotes a Th2/IL-4 response. These findings highlight the importance of DC S1P3 in modulating NKT cell function and IRI and support development of selective S1P3 antagonists for tolerizing DCs for cell-based therapy or for systemic administration for the prevention and treatment of IRI and autoimmune diseases.


Assuntos
Polaridade Celular/imunologia , Células Dendríticas/imunologia , Isquemia/patologia , Rim/patologia , Receptores de Lisoesfingolipídeo/fisiologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Isquemia/imunologia , Rim/irrigação sanguínea , Rim/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Células T Matadoras Naturais/patologia , Infiltração de Neutrófilos/imunologia , Receptores de Lisoesfingolipídeo/deficiência , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Receptores de Esfingosina-1-Fosfato , Células Th1/patologia , Células Th2/patologia
18.
J Am Soc Nephrol ; 24(9): 1451-60, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23907510

RESUMO

AKI affects both quality of life and health care costs and is an independent risk factor for mortality. At present, there are few effective treatment options for AKI. Here, we describe a nonpharmacologic, noninvasive, ultrasound-based method to prevent renal ischemia-reperfusion injury in mice, which is a model for human AKI. We exposed anesthetized mice to an ultrasound protocol 24 hours before renal ischemia. After 24 hours of reperfusion, ultrasound-treated mice exhibited preserved kidney morphology and function compared with sham-treated mice. Ultrasound exposure before renal ischemia reduced the accumulation of CD11b(+)Ly6G(high) neutrophils and CD11b(+)F4/80(high) myeloid cells in kidney tissue. Furthermore, splenectomy and adoptive transfer studies revealed that the spleen and CD4(+) T cells mediated the protective effects of ultrasound. Last, blockade or genetic deficiency of the α7 nicotinic acetylcholine receptor abrogated the protective effect of ultrasound, suggesting the involvement of the cholinergic anti-inflammatory pathway. Taken together, these results suggest that an ultrasound-based treatment could have therapeutic potential for the prevention of AKI, possibly by stimulating a splenic anti-inflammatory pathway.


Assuntos
Rim/irrigação sanguínea , Rim/patologia , Receptores Colinérgicos/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/fisiologia , Baço/fisiologia , Terapia por Ultrassom , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/fisiologia , Modelos Animais de Doenças , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , Neutrófilos/imunologia , Neutrófilos/patologia , Receptores Colinérgicos/deficiência , Receptores Colinérgicos/genética , Receptores Nicotínicos/deficiência , Receptores Nicotínicos/genética , Receptores Nicotínicos/fisiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Baço/cirurgia , Esplenectomia , Receptor Nicotínico de Acetilcolina alfa7
19.
Nephron Exp Nephrol ; 121(1-2): e1-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23095207

RESUMO

AIMS: The role of kidney infiltrating T cells in the pathology of lupus nephritis is unclear. This study was undertaken to investigate whether CD4+ T cell responses to a surrogate mesangial antigen can initiate glomerulonephritis. METHODS: Ovalbumin (OVA) was deposited in the glomerular mesangium of C57BL/6 (B6) mice using anti-α8-integrin immunoliposomes (α8ILs). This was followed by injection of activated OVA-reactive CD4+ transgenic OT2 T cells. Trafficking of antigen-specific OT2 T cells to kidneys and lymph nodes was studied by flow cytometry. Glomerular pathology and immune cell infiltration was characterized by immunostaining. Role of CCR2 deficiency on T cell-mediated glomerulonephritis was investigated using B6.ccr2(-/-) mice. RESULTS: α8ILs delivered OVA specifically to the renal glomeruli. Adoptively transferred OT2 T cells preferentially accumulated in renal lymph nodes and in the renal cortex. Kidneys showed glomerular inflammation with recruitment of endogenous T cells, dendritic cells and macrophages. T cell-mediated inflammation induced mesangial cell activation and an increase in glomerular MCP1 and fibronectin. The formation of inflammatory foci was driven by Ly6C monocytes and was CCR2 dependent. CONCLUSIONS: The findings from this study show that T cells reactive with antigens in the mesangium are sufficient to initiate glomerular pathology. Antigen-specific CD4 T cells act by inducing glomerular MCP1 production which mediates recruitment of inflammatory monocytes resulting in glomerulonephritis. Thus, down-modulation of T cell responses within the kidneys of lupus patients will be a beneficial therapeutic approach.


Assuntos
Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Glomerulonefrite/imunologia , Rim/imunologia , Ativação Linfocitária/imunologia , Células Mesangiais/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Feminino , Glomerulonefrite/patologia , Rim/patologia , Células Mesangiais/patologia , Camundongos , Camundongos Endogâmicos C57BL
20.
J Am Soc Nephrol ; 22(3): 416-25, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21335516

RESUMO

The response to exogenous pathogens leads to activation of innate immunity through the release of pathogen-associated molecular patterns (PAMPs) and their binding to pattern recognition receptors. A classic example is septic shock where Toll receptor 4 recognizes PAMPs. Although well accepted, this concept does not explain the activation of innate immunity and inflammation occurs with transplantation, autoimmunity, or trauma. Increasingly recognized is that endogenous molecules released by dying cells (damage-associated molecular patterns; DAMPs) activate cellular receptors leading to downstream inflammation. Thus endogenous danger signals and exogenous PAMPs elicit similar responses through seemingly similar mechanisms. Also emerging is our understanding that normal repair processes benefit from dampening the immune response to these endogenous danger molecules. Here we focus on the role of DAMPs and their putative receptors in the pathogenesis of acute and chronic kidney diseases.


Assuntos
Imunidade Adaptativa/fisiologia , Imunidade Inata/fisiologia , Nefropatias/imunologia , Doença Aguda , Morte Celular/fisiologia , Doença Crônica , Humanos , Nefropatias/fisiopatologia , Receptores de Reconhecimento de Padrão/fisiologia , Receptores Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa