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
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.
Front Med (Lausanne) ; 9: 931293, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35966871

RESUMO

Adenosine triphosphate (ATP) released from injured or dying cells is a potent pro-inflammatory "danger" signal. Alkaline phosphatase (AP), an endogenous enzyme that de-phosphorylates extracellular ATP, likely plays an anti-inflammatory role in immune responses. We hypothesized that ilofotase alfa, a human recombinant AP, protects kidneys from ischemia-reperfusion injury (IRI), a model of acute kidney injury (AKI), by metabolizing extracellular ATP to adenosine, which is known to activate adenosine receptors. Ilofotase alfa (iv) with or without ZM241,385 (sc), a selective adenosine A2A receptor (A2AR) antagonist, was administered 1 h before bilateral IRI in WT, A2AR KO (Adora2a-/- ) or CD73-/- mice. In additional studies recombinant alkaline phosphatase was given after IRI. In an AKI-on-chronic kidney disease (CKD) ischemic rat model, ilofotase alfa was given after the three instances of IRI and rats were followed for 56 days. Ilofotase alfa in a dose dependent manner decreased IRI in WT mice, an effect prevented by ZM241,385 and partially prevented in Adora2a-/- mice. Enzymatically inactive ilofotase alfa was not protective. Ilofotase alfa rescued CD73-/- mice, which lack a 5'-ectonucleotidase that dephosphorylates AMP to adenosine; ZM241,385 inhibited that protection. In both rats and mice ilofotase alfa ameliorated IRI when administered after injury, thus providing relevance for therapeutic dosing of ilofotase alfa following established AKI. In an AKI-on-CKD ischemic rat model, ilofotase alfa given after the third instance of IRI reduced injury. These results suggest that ilofotase alfa promotes production of adenosine from liberated ATP in injured kidney tissue, thereby amplifying endogenous mechanisms that can reverse tissue injury, in part through A2AR-and non-A2AR-dependent signaling pathways.

3.
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
4.
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
5.
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
6.
J Clin Invest ; 126(5): 1939-52, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27088805

RESUMO

The nervous and immune systems interact in complex ways to maintain homeostasis and respond to stress or injury, and rapid nerve conduction can provide instantaneous input for modulating inflammation. The inflammatory reflex referred to as the cholinergic antiinflammatory pathway regulates innate and adaptive immunity, and modulation of this reflex by vagus nerve stimulation (VNS) is effective in various inflammatory disease models, such as rheumatoid arthritis and inflammatory bowel disease. Effectiveness of VNS in these models necessitates the integration of neural signals and α7 nicotinic acetylcholine receptors (α7nAChRs) on splenic macrophages. Here, we sought to determine whether electrical stimulation of the vagus nerve attenuates kidney ischemia-reperfusion injury (IRI), which promotes the release of proinflammatory molecules. Stimulation of vagal afferents or efferents in mice 24 hours before IRI markedly attenuated acute kidney injury (AKI) and decreased plasma TNF. Furthermore, this protection was abolished in animals in which splenectomy was performed 7 days before VNS and IRI. In mice lacking α7nAChR, prior VNS did not prevent IRI. Conversely, adoptive transfer of VNS-conditioned α7nAChR splenocytes conferred protection to recipient mice subjected to IRI. Together, these results demonstrate that VNS-mediated attenuation of AKI and systemic inflammation depends on α7nAChR-positive splenocytes.


Assuntos
Injúria Renal Aguda/prevenção & controle , Rim/imunologia , Macrófagos/imunologia , Traumatismo por Reperfusão/terapia , Baço/imunologia , Estimulação do Nervo Vago , Receptor Nicotínico de Acetilcolina alfa7/imunologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Rim/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Baço/patologia , Receptor Nicotínico de Acetilcolina alfa7/genética
7.
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
8.
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
9.
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
10.
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
11.
J Clin Invest ; 122(11): 3931-42, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23093781

RESUMO

DC-mediated NKT cell activation is critical in initiating the immune response following kidney ischemia/reperfusion injury (IRI), which mimics human acute kidney injury (AKI). Adenosine is an important antiinflammatory molecule in tissue inflammation, and adenosine 2A receptor (A2AR) agonists protect kidneys from IRI through their actions on leukocytes. In this study, we showed that mice with A2AR-deficient DCs are more susceptible to kidney IRI and are not protected from injury by A2AR agonists. In addition, administration of DCs treated ex vivo with an A2AR agonist protected the kidneys of WT mice from IRI by suppressing NKT production of IFN-γ and by regulating DC costimulatory molecules that are important for NKT cell activation. A2AR agonists had no effect on DC antigen presentation or on Tregs. We conclude that ex vivo A2AR-induced tolerized DCs suppress NKT cell activation in vivo and provide a unique and potent cell-based strategy to attenuate organ IRI.


Assuntos
Injúria Renal Aguda/prevenção & controle , Agonistas do Receptor A2 de Adenosina/farmacologia , Células Dendríticas/imunologia , Tolerância Imunológica/efeitos dos fármacos , Rim/imunologia , Receptor A2A de Adenosina/imunologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Células Dendríticas/patologia , Células Dendríticas/transplante , Humanos , Tolerância Imunológica/genética , Interferon gama/genética , Interferon gama/imunologia , Rim/patologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/patologia , Receptor A2A de Adenosina/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle
12.
J Am Soc Nephrol ; 21(6): 955-65, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20338995

RESUMO

Agonists of the sphingosine-1-phosphate receptor (S1PR) attenuate kidney ischemia-reperfusion injury (IRI). Previous studies suggested that S1P1R-induced lymphopenia mediates this protective effect, but lymphocyte-independent mechanisms could also contribute. Here, we investigated the effects of S1PR agonists on kidney IRI in mice that lack T and B lymphocytes (Rag-1 knockout mice). Administration of the nonselective S1PR agonist FTY720 or the selective S1P1R agonist SEW2871 reduced injury in both Rag-1 knockout and wild-type mice. In vitro, SEW2871 significantly attenuated LPS- or hypoxia/reoxygenation-induced apoptosis in cultured mouse proximal tubule epithelial cells, supporting a direct protective effect of S1P1R agonists via mitogen-activated protein kinase and/or Akt pathways. S1P1Rs in the proximal tubule mediated IRI in vivo as well: Mice deficient in proximal tubule S1P1Rs experienced a greater decline in renal function after IRI than control mice and their kidneys were no longer protected by SEW2871 administration. In summary, S1PRs in the proximal tubule are necessary for stress-induced cell survival, and S1P1R agonists are renoprotective via direct effects on the tubule cells. Selective agonists of S1P1Rs may hold therapeutic potential for the prevention and treatment of acute kidney injury.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/patologia , Cloridrato de Fingolimode , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Túbulos Renais Proximais/patologia , Leucócitos/efeitos dos fármacos , Leucócitos/patologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oxidiazóis/farmacologia , Propilenoglicóis/farmacologia , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tiofenos/farmacologia
13.
ScientificWorldJournal ; 9: 1321-44, 2009 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19936569

RESUMO

Basal ganglia processing results from a balanced activation of direct and indirect striatal efferent pathways, which are controlled by dopamine D1 and D2 receptors, respectively. Adenosine A2A receptors are considered novel antiparkinsonian targets, based on their selective postsynaptic localization in the indirect pathway, where they modulate D2 receptor function. The present study provides evidence for the existence of an additional, functionally significant, segregation of A2A receptors at the presynaptic level. Using integrated anatomical, electrophysiological, and biochemical approaches, we demonstrate that presynaptic A2A receptors are preferentially localized in cortical glutamatergic terminals that contact striatal neurons of the direct pathway, where they exert a selective modulation of corticostriatal neurotransmission. Presynaptic striatal A2A receptors could provide a new target for the treatment of neuropsychiatric disorders.


Assuntos
Gânglios da Base/fisiologia , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Receptor A2A de Adenosina/fisiologia , Transmissão Sináptica/fisiologia , Antagonistas do Receptor A2 de Adenosina , Animais , Benzazepinas/farmacologia , Corpo Estriado/ultraestrutura , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores , Ácido Glutâmico/metabolismo , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Terminações Pré-Sinápticas , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor A2A de Adenosina/imunologia , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D2/fisiologia , Sinaptossomos/fisiologia , Xantinas/farmacologia
14.
Kidney Int ; 74(12): 1526-37, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18843253

RESUMO

Chemokines and their receptors such as CCR2 and CX3CR1 mediate leukocyte adhesion and migration into injured tissue. To further define mechanisms of monocyte trafficking during kidney injury we identified two groups of F4/80-positive cells (F4/80(low) and F4/80(high)) in the normal mouse kidney that phenotypically correspond to macrophages and dendritic cells, respectively. Following ischemia and 3 h of reperfusion, there was a large influx of F4/80(low) inflamed monocytes, but not dendritic cells, into the kidney. These monocytes produced TNF-alpha, IL-6, IL-1alpha and IL-12. Ischemic injury induced in CCR2(-/-) mice or in CCR2(+/+) mice, made chimeric with CCR2(-/-) bone marrow, resulted in lower plasma creatinine levels and their kidneys had fewer infiltrated F4/80(low) macrophages compared to control mice. CX3CR1 expression contributed to monocyte recruitment into inflamed kidneys, as ischemic injury in CX3CR1(-/-) mice was reduced, with fewer F4/80(low) macrophages than controls. Monocytes transferred from CCR2(+/+) or CX3CR1(+/-) mice migrated into reperfused kidneys better than monocytes from either CCR2(-/-) or CX3CR1(-/-) mice. Adoptive transfer of monocytes from CCR2(+/+) mice, but not CCR2(-/-) mice, reversed the protective effect in CCR2(-/-) mice following ischemia-reperfusion. Egress of CD11b(+)Ly6C(high) monocytes from blood into inflamed kidneys was CCR2- and CX3CR1-dependent. Our study shows that inflamed monocyte migration, through CCR2- and CX3CR1-dependent mechanisms, plays a critical role in kidney injury following ischemia reperfusion.


Assuntos
Quimiotaxia/imunologia , Nefropatias/imunologia , Receptores CCR2/fisiologia , Receptores de Quimiocinas/fisiologia , Traumatismo por Reperfusão/imunologia , Transferência Adotiva , Animais , Receptor 1 de Quimiocina CX3C , Isquemia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Monócitos/fisiologia
15.
J Am Soc Nephrol ; 19(1): 59-68, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18045850

RESUMO

Adenosine 2A receptor (A(2A)R) activation was recently shown to be renoprotective in diabetic nephropathy. A(2A)R are found in glomeruli and have been shown to associate with the podocyte cytoskeletal protein alpha-actinin-4, but the effect of their activation on podocyte structure and function is unknown. Podocyte injury was induced in C57BL/6 mice with puromycin aminonucleoside, and the selective A(2A)R agonist ATL313 was found to attenuate the resulting albuminuria and foot process fusion. The selective A(2A)R antagonist ZM241385 reversed the effects of ATL313. In vitro, A(2A)R mRNA and protein were expressed in a conditionally immortalized podocyte cell line, and A(2A)R-like immunoreactivity co-localized with the actin cytoskeleton. Treatment with ATL313 also blocked the increased podocyte permeability to albumin and disruption of the actin cytoskeleton that accompanied puromycin aminonucleoside-induced injury in vitro. ATL313 was ineffective, however, in the presence of the A(2A)R antagonist and in A(2A)R-deficient podocytes. It was concluded that A(2A)R activation reduces glomerular proteinuria, at least in part, by preserving the normal structure of podocyte foot processes, slit diaphragms, and actin cytoskeleton.


Assuntos
Albuminúria/prevenção & controle , Podócitos/fisiologia , Receptores A2 de Adenosina/fisiologia , Agonistas do Receptor A2 de Adenosina , Animais , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/uso terapêutico , Podócitos/efeitos dos fármacos , Proteinúria/induzido quimicamente , Proteinúria/prevenção & controle , Nucleosídeos de Purina/efeitos adversos , Receptores A2 de Adenosina/efeitos dos fármacos , Triazinas/farmacologia , Triazinas/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico
16.
Synapse ; 60(7): 496-509, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16952160

RESUMO

Adenosine A1 and A2A receptors mediate distinct cardiovascular components of defense reactions that are ascribed, in part, to opposing actions within the nucleus tractus solitarius. To assess the cellular sites of relevance to these actions, we examined the light and electron microscopic immunolabeling of adenosine A1 and A2A receptors in the rat dorsomedial nucleus of the solitary tract at the level of the area postrema (dmNTS-AP), a region crucial for cardiovascular regulation involving vagal baroreceptor afferents. Immunoreactivity for each receptor was independently localized to distinct segments of plasma membranes and endomembranes in somatodendritic, axonal, and glial profiles. The dendritic labeling for each receptor also was detected within and near asymmetric, excitatory-type synapses. Of all peroxidase labeled profiles exclusive of somata, approximately 58% were A1- and 39% were A2A-labeled dendrites. Dendrites and astrocytic glia were the profiles that most often expressed both subtypes of adenosine receptors. The axonal labeling for A2A receptors was seen mainly in unmyelinated axons, whereas the A1 receptors were prominently localized within axon terminals. These terminals often formed single or multisynaptic excitatory-type junctions or single symmetric synapses on dendrites, a few of which expressed A1 and A2A receptors. These results provide the first ultrastructural evidence that A1 and A2A receptors have distributions conductive to their dual involvement in modulating the output of single neurons and glial function in the dmNTS-AP, where the predominate presynaptic effects of adenosine are mediated through A1 receptors.


Assuntos
Adenosina/metabolismo , Neurônios/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Núcleo Solitário/metabolismo , Animais , Área Postrema/anatomia & histologia , Astrócitos/metabolismo , Astrócitos/ultraestrutura , Fenômenos Fisiológicos Cardiovasculares , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Dendritos/metabolismo , Dendritos/ultraestrutura , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/genética , Sistemas do Segundo Mensageiro/fisiologia , Núcleo Solitário/ultraestrutura , Membranas Sinápticas/metabolismo , Membranas Sinápticas/ultraestrutura , Transmissão Sináptica/fisiologia , Fibras Aferentes Viscerais/metabolismo , Fibras Aferentes Viscerais/ultraestrutura
17.
Neurology ; 61(11 Suppl 6): S12-8, 2003 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-14663003

RESUMO

A2A adenosine receptors (A(2A)Rs) are expressed with the greatest abundance in the striatum and other nuclei of the basal ganglia. The segregated expression of A(2A)Rs on the GABAergic striatopallidal medium spiny neurons, where A(2A)R and D2 dopamine receptor mRNAs are colocalized, and the opposing functional interaction between adenosine and dopamine suggest that A(2A)Rs may be an important therapeutic target. To further explore the role of A(2A)Rs in the synaptic organization of the basal ganglia, the authors developed an antibody directed against the purified A(2A)R. Immunohistochemical studies in rat brain showed dense labeling of the neuropil in the striatum, nucleus accumbens, and olfactory tubercles with lighter labeling of terminals in the globus pallidus (GP), where A(2A)R transcript is not detected. Stimulation of A(2A)Rs on GP terminals may facilitate GABAergic signaling and contribute to the overactivation observed in Parkinson's disease (PD). Analysis at the ultrastructural level allowed a more detailed characterization of the mechanism(s) of A2A-mediated control of striatal output. In the striatum, terminals expressing A(2A)Rs accounted for 25% of the labeled elements. These presynaptic receptors may facilitate excitatory glutamatergic, inhibitory GABAergic, and possibly cholinergic striatal transmission. However, the majority of striatal A(2A)R immunoreactivity was found on postsynaptic elements, including dendrites of striatopallidal neurons, in which A(2A)R and GABA immunoreactivity is colocalized. Activation of these receptors may promote GABAergic signaling in striatopallidal output neurons and their local axon collaterals in the striatum. Many of the A2A-labeled dendrites were contacted by terminals forming asymmetric (excitatory) possibly glutamatergic synapses. Using the vesicular glutamate transporters (VGLUTs) as markers of glutamatergic terminals, the authors have found that VGLUT1-immunoreactive (ir) terminals make asymmetric contacts on A2A-ir spines and spine heads in the striatum, suggesting that regulation of striatal output by A(2A)R stimulation may involve facilitation of the cortical glutamatergic excitatory input to striatopallidal neurons. These ultrastructural findings suggest several pathways through which A2A receptor blockade may act to dampen the elevated striatopallidal GABAergic signaling that occurs in PD.


Assuntos
Encéfalo/anatomia & histologia , Receptor A2A de Adenosina/biossíntese , Animais , Gânglios da Base/anatomia & histologia , Gânglios da Base/metabolismo , Encéfalo/metabolismo , Corpo Estriado/anatomia & histologia , Corpo Estriado/metabolismo , Dendritos/metabolismo , Dendritos/ultraestrutura , Humanos , Neurônios/metabolismo , Neurônios/ultraestrutura
18.
J Clin Invest ; 112(6): 883-91, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12975473

RESUMO

Activation of A2A adenosine receptors (A2ARs) protects kidneys from ischemia-reperfusion injury (IRI). A2ARs are expressed on bone marrow-derived (BM-derived) cells and renal smooth muscle, epithelial, and endothelial cells. To measure the contribution of A2ARs on BM-derived cells in suppressing renal IRI, we examined the effects of a selective agonist of A2ARs, ATL146e, in chimeric mice in which BM was ablated by lethal radiation and reconstituted with donor BM cells derived from GFP, A2AR-KO, or WT mice to produce GFP-->WT, A2A-KO-->WT, or WT-->WT mouse chimera. We found little or no repopulation of renal vascular endothelial cells by donor BM with or without renal IRI. ATL146e had no effect on IRI in A2A-KO mice or A2A-KO-->WT chimera, but reduced the rise in plasma creatinine from IRI by 75% in WT mice and by 60% in WT-->WT chimera. ATL146e reduced the induction of IL-6, IL-1beta, IL-1ra, and TGF-alpha mRNA in WT-->WT mice but not in A2A-KO-->WT mice. Plasma creatinine was significantly greater in A2A-KO than in WT mice after IRI, suggesting some renal protection by endogenous adenosine. We conclude that protection from renal IRI by A2AR agonists or endogenous adenosine requires activation of receptors expressed on BM-derived cells.


Assuntos
Células da Medula Óssea/metabolismo , Rim/metabolismo , Receptores Purinérgicos P1/metabolismo , Traumatismo por Reperfusão/metabolismo , Adenosina/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Transplante de Células , Quimiocinas/genética , Quimiocinas/metabolismo , Ácidos Cicloexanocarboxílicos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Genótipo , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Agonistas do Receptor Purinérgico P1 , Purinas/metabolismo , Receptores Purinérgicos P1/genética , Quimeras de Transplante
19.
J Comp Neurol ; 444(3): 207-20, 2002 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-11840475

RESUMO

The main source of excitatory drive to the sympathetic preganglionic neurons that control blood pressure is from neurons located in the rostral ventrolateral medulla (RVLM). This monosynaptic input includes adrenergic (C1), peptidergic, and noncatecholaminergic neurons. Some of the cells in this pathway are suspected to be glutamatergic, but conclusive evidence is lacking. In the present study we sought to determine whether these presympathetic neurons express the vesicular glutamate transporter BNPI/VGLUT1 or the closely related gene DNPI, the rat homolog of the mouse vesicular glutamate transporter VGLUT2. Both BNPI/VGLUT1 and DNPI/VGLUT2 mRNAs were detected in the medulla oblongata by in situ hybridization, but only DNPI/VGLUT2 mRNA was present in the RVLM. Moreover, BNPI immunoreactivity was absent from the thoracic spinal cord lateral horn. DNPI/VGLUT2 mRNA was present in many medullary cells retrogradely labeled with Fluoro-Gold from the spinal cord (T2; four rats). Within the RVLM, 79% of the bulbospinal C1 cells contained DNPI/VGLUT2 mRNA. Bulbospinal noradrenergic A5 neurons did not contain DNPI/VGLUT2 mRNA. The RVLM of six unanesthetized rats subjected to 2 hours of hydralazine-induced hypotension contained tenfold more c-Fos-ir DNPI/VGLUT2 neurons than that of six saline-treated controls. c-Fos-ir DNPI/VGLUT2 neurons included C1 and non-C1 neurons (3:2 ratio). In seven barbiturate-anesthetized rats, 16 vasomotor presympathetic neurons were filled with biotinamide and analyzed for the presence of tyrosine hydroxylase immunoreactivity and/or DNPI/VGLUT2 mRNA. Biotinamide-labeled neurons included C1 and non-C1 cells. Most non-C1 (9/10) and C1 presympathetic cells (5/6) contained DNPI/VGLUT2 mRNA. In conclusion, DNPI/VGLUT2 is expressed by most blood pressure-regulating presympathetic cells of the RVLM. The data suggest that these neurons may be glutamatergic and that the C1 adrenergic phenotype is one of several secondary phenotypes that are differentially expressed by subgroups of these cells.


Assuntos
Proteínas de Transporte/metabolismo , Bulbo/fisiologia , Proteínas de Membrana Transportadoras , Neurônios/metabolismo , Ratos/metabolismo , Sistema Nervoso Simpático/metabolismo , Sistema Vasomotor/metabolismo , Proteínas de Transporte Vesicular , Aminas/metabolismo , Animais , Proteínas de Transporte/genética , Eletrofisiologia , Inibidores do Crescimento , Hidralazina , Hipotensão/induzido quimicamente , Hipotensão/metabolismo , Masculino , Bulbo/citologia , Bulbo/metabolismo , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Medula Espinal/citologia , Medula Espinal/metabolismo , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia , Sistema Vasomotor/citologia , Sistema Vasomotor/fisiologia , Proteína Vesicular 1 de Transporte de Glutamato , Proteína Vesicular 2 de Transporte de Glutamato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA