Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circulation ; 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38682338

RESUMO

BACKGROUND: Most organs are maintained lifelong by resident stem/progenitor cells. During development and regeneration, lineage-specific stem/progenitor cells can contribute to the growth or maintenance of different organs, whereas fully differentiated mature cells have less regenerative potential. However, it is unclear whether vascular endothelial cells (ECs) are also replenished by stem/progenitor cells with EC-repopulating potential residing in blood vessels. It has been reported recently that some EC populations possess higher clonal proliferative potential and vessel-forming capacity compared with mature ECs. Nevertheless, a marker to identify vascular clonal repopulating ECs (CRECs) in murine and human individuals is lacking, and, hence, the mechanism for the proliferative, self-renewal, and vessel-forming potential of CRECs is elusive. METHODS: We analyzed colony-forming, self-renewal, and vessel-forming potential of ABCG2 (ATP binding cassette subfamily G member 2)-expressing ECs in human umbilical vessels. To study the contribution of Abcg2-expressing ECs to vessel development and regeneration, we developed Abcg2CreErt2;ROSA TdTomato mice and performed lineage tracing during mouse development and during tissue regeneration after myocardial infarction injury. RNA sequencing and chromatin methylation chromatin immunoprecipitation followed by sequencing were conducted to study the gene regulation in Abcg2-expressing ECs. RESULTS: In human and mouse vessels, ECs with higher ABCG2 expression (ABCECs) possess higher clonal proliferative potential and in vivo vessel-forming potential compared with mature ECs. These cells could clonally contribute to vessel formation in primary and secondary recipients after transplantation. These features of ABCECs meet the criteria of CRECs. Results from lineage tracing experiments confirm that Abcg2-expressing CRECs (AbcCRECs) contribute to arteries, veins, and capillaries in cardiac tissue development and vascular tissue regeneration after myocardial infarction. Transcriptome and epigenetic analyses reveal that a gene expression signature involved in angiogenesis and vessel development is enriched in AbcCRECs. In addition, various angiogenic genes, such as Notch2 and Hey2, are bivalently modified by trimethylation at the 4th and 27th lysine residue of histone H3 (H3K4me3 and H3K27me3) in AbcCRECs. CONCLUSIONS: These results are the first to establish that a single prospective marker identifies CRECs in mice and human individuals, which holds promise to provide new cell therapies for repair of damaged vessels in patients with endothelial dysfunction.

2.
Am J Physiol Renal Physiol ; 326(1): F86-F94, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37881874

RESUMO

Renal reserve capacity may be compromised following recovery from acute kidney injury (AKI) and could be used to identify impaired renal function in the face of restored glomerular filtration rate (GFR) or plasma creatinine. To investigate the loss of hemodynamic renal reserve responses following recovery in a model of AKI, rats were subjected to left unilateral renal ischemia-reperfusion (I/R) injury and contralateral nephrectomy and allowed to recover for 5 wk. Some rats were treated 24 h post-I/R by hydrodynamic isotonic fluid delivery (AKI-HIFD) of saline through the renal vein, previously shown to improve recovery and inflammation relative to control rats that received saline through the vena cava (AKI-VC). At 5 wk after surgery, plasma creatinine and GFR recovered to levels observed in uninephrectomized sham controls. Baseline renal blood flow (RBF) was not different between AKI or sham groups, but infusion of l-arginine (7.5 mg/kg/min) significantly increased RBF in sham controls, whereas the RBF response to l-arginine was significantly reduced in AKI-VC rats relative to sham rats (22.6 ± 2.2% vs. 13.8 ± 1.8%, P < 0.05). RBF responses were partially protected in AKI-HIFD rats relative to AKI-VC rats (17.0 ± 2.2%) and were not significantly different from sham rats. Capillary rarefaction observed in AKI-VC rats was significantly protected in AKI-HIFD rats. There was also a significant increase in T helper 17 cell infiltration and interstitial fibrosis in AKI-VC rats versus sham rats, which was not present in AKI-HIFD rats. These data suggest that recovery from AKI results in impaired hemodynamic reserve and that associated CKD progression may be mitigated by HIFD in the early post-AKI period.NEW & NOTEWORTHY Despite the apparent recovery of renal filtration function following acute kidney injury (AKI) in rats, the renal hemodynamic reserve response is significantly attenuated, suggesting that clinical evaluation of this parameter may provide information on the potential development of chronic kidney disease. Treatments such as hydrodynamic isotonic fluid delivery, or other treatments in the early post-AKI period, could minimize chronic inflammation or loss of microvessels with the potential to promote a more favorable outcome on long-term function.


Assuntos
Injúria Renal Aguda , Insuficiência Renal Crônica , Traumatismo por Reperfusão , Ratos , Animais , Hidrodinâmica , Creatinina , Ratos Sprague-Dawley , Rim , Injúria Renal Aguda/terapia , Hemodinâmica/fisiologia , Inflamação , Arginina , Modelos Animais de Doenças
3.
Kidney Int ; 101(1): 15-18, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34991803

RESUMO

Cylocloxygenase-2 is an important mediator of arachidonic acid metabolism. Pan et al. recently identified a robust increase in the expression of cylocloxygenase-2 in proresolving macrophages (M2) during the repair phase of acute kidney injury. The investigators determined the prostaglandin E2 was produced in macrophages and demonstrated that signaling through the E-type prostanoid receptor 4 stimulated the expression of the anti-inflammatory transcription factor MafB. MafB was further shown to be essential for macrophage differentiation and mediation of the intrinsic repair response following experimental acute kidney injury.


Assuntos
Rim , Macrófagos , Anti-Inflamatórios , Ciclo-Oxigenase 2/metabolismo , Rim/metabolismo , Macrófagos/metabolismo , Transdução de Sinais
4.
Crit Care ; 26(1): 107, 2022 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-35422004

RESUMO

BACKGROUND: Interleukin-17 (IL-17) antagonism in rats reduces the severity and progression of AKI. IL-17-producing circulating T helper-17 (TH17) cells is increased in critically ill patients with AKI indicating that this pathway is also activated in humans. We aim to compare serum IL-17A levels in critically ill patients with versus without AKI and to examine their relationship with mortality and major adverse kidney events (MAKE). METHODS: Multicenter, prospective study of ICU patients with AKI stage 2 or 3 and without AKI. Samples were collected at 24-48 h after AKI diagnosis or ICU admission (in those without AKI) [timepoint 1, T1] and 5-7 days later [timepoint 2, T2]. MAKE was defined as the composite of death, dependence on kidney replacement therapy or a reduction in eGFR of ≥ 30% from baseline up to 90 days following hospital discharge. RESULTS: A total of 299 patients were evaluated. Patients in the highest IL-17A tertile (versus lower tertiles) at T1 had higher acuity of illness and comorbidity scores. Patients with AKI had higher levels of IL-17A than those without AKI: T1 1918.6 fg/ml (692.0-5860.9) versus 623.1 fg/ml (331.7-1503.4), p < 0.001; T2 2167.7 fg/ml (839.9-4618.9) versus 1193.5 fg/ml (523.8-2198.7), p = 0.006. Every onefold higher serum IL-17A at T1 was independently associated with increased risk of hospital mortality (aOR 1.35, 95% CI: 1.06-1.73) and MAKE (aOR 1.26, 95% CI: 1.02-1.55). The highest tertile of IL-17A (vs. the lowest tertile) was also independently associated with higher risk of MAKE (aOR 3.03, 95% CI: 1.34-6.87). There was no effect modification of these associations by AKI status. IL-17A levels remained significantly elevated at T2 in patients that died or developed MAKE. CONCLUSIONS: Serum IL-17A levels measured by the time of AKI diagnosis or ICU admission were differentially elevated in critically ill patients with AKI when compared to those without AKI and were independently associated with hospital mortality and MAKE.


Assuntos
Injúria Renal Aguda , Interleucina-17 , Injúria Renal Aguda/terapia , Animais , Estado Terminal/terapia , Feminino , Humanos , Unidades de Terapia Intensiva , Masculino , Estudos Prospectivos , Ratos
5.
Curr Opin Nephrol Hypertens ; 30(2): 151-158, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33394732

RESUMO

PURPOSE OF REVIEW: Hypertension has been demonstrated to be a chief contributor to morbidity and mortality throughout the world. Although the cause of hypertension is multifactorial, emerging evidence, obtained in experimental studies, as well as observational studies in humans, points to the role of inflammation and immunity. Many aspects of immune function have now been implicated in hypertension and end-organ injury; this review will focus upon the recently-described role of Th17 cells in this pathophysiological response. RECENT FINDINGS: Studies in animal models and human genetic studies point to a role in the adaptive immune system as playing a contributory role in hypertension and renal tissue damage. Th17 cells, which produce the cytokine IL17, are strongly pro-inflammatory cells, which may contribute to tissue damage if expressed in chronic disease conditions. The activity of these cells may be enhanced by physiological factors associated with hypertension such as dietary salt or Ang II. This activity may culminate in the increased sodium retaining activity and exacerbation of inflammation and renal fibrosis via multiple cellular mechanisms. SUMMARY: Th17 cells are a distinct component of the adaptive immune system that may strongly enhance pathways leading to increased sodium reabsorption, elevated vascular tone and end-organ damage. Moreover, this pathway may lend itself towards specific targeting for treatment of kidney disease and hypertension.


Assuntos
Hipertensão , Nefropatias , Animais , Humanos , Rim , Cloreto de Sódio na Dieta , Células Th17
6.
Am J Physiol Renal Physiol ; 319(5): F796-F808, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32924545

RESUMO

To investigate T helper type 17 (Th17) cells in the setting of acute kidney injury, the gene encoding the master regulator of Th17 cell differentiation, that is, RAR-related orphan receptor-γ (RORγT), was mutated in Lewis rats using CRISPR/Cas9 technology. In response to 40 min of bilateral renal ischemia-reperfusion (I/R), RAR-related orphan receptor C (Rorc)-/- rats were resistant to injury relative to wild-type Rorc+/+ rats. This protection was associated with inhibition of IL-17 expression and reduced infiltration of CD4+ cells, CD8+ cells, B cells, and macrophages. To evaluate the effect of Th17 cells on repair, ischemia was increased to 50 min in Rorc-/- rats. This maneuver equalized the initial level of injury in Rorc-/- and Rorc+/+ rats 1 to 2 days post-I/R based on serum creatinine values. However, Rorc-/- rats, but not Rorc+/+ rats, failed to successfully recover renal function and had high mortality by 4 days post-I/R. Histological assessment of kidney tubules showed evidence of repair by day 4 post-I/R in Rorc+/+ rats but persistent necrosis and elevated cell proliferation in Rorc-/- rats. Adoptive transfer of CD4+ cells from the spleen of Rorc+/+ rats or supplementation of exogenous rIL-17 by an osmotic minipump improved renal function and survival of Rorc-/- rats following 50 min of I/R. This was associated with a relative decrease in the number of M1-type macrophages and a relative increase in the percentage of T regulatory cells. Taken together, these data suggest that Th17 cells have both a deleterious and a beneficial role in kidney injury and recovery, contributing to early postischemic injury and inflammation but also possibly being critical in the resolution of inflammation during kidney repair.


Assuntos
Rim/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Traumatismo por Reperfusão/metabolismo , Linfócitos T Reguladores/metabolismo , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Inflamação/metabolismo , Isquemia/metabolismo , Mutação/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Ratos , Ratos Endogâmicos Lew , Recuperação de Função Fisiológica , Traumatismo por Reperfusão/patologia , Células Th17
8.
J Am Soc Nephrol ; 29(4): 1154-1164, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29371417

RESUMO

Ischemic preconditioning confers organ-wide protection against subsequent ischemic stress. A substantial body of evidence underscores the importance of mitochondria adaptation as a critical component of cell protection from ischemia. To identify changes in mitochondria protein expression in response to ischemic preconditioning, we isolated mitochondria from ischemic preconditioned kidneys and sham-treated kidneys as a basis for comparison. The proteomic screen identified highly upregulated proteins, including NADP+-dependent isocitrate dehydrogenase 2 (IDH2), and we confirmed the ability of this protein to confer cellular protection from injury in murine S3 proximal tubule cells subjected to hypoxia. To further evaluate the role of IDH2 in cell protection, we performed detailed analysis of the effects of Idh2 gene delivery on kidney susceptibility to ischemia-reperfusion injury. Gene delivery of IDH2 before injury attenuated the injury-induced rise in serum creatinine (P<0.05) observed in controls and increased the mitochondria membrane potential (P<0.05), maximal respiratory capacity (P<0.05), and intracellular ATP levels (P<0.05) above those in controls. This communication shows that gene delivery of Idh2 can confer organ-wide protection against subsequent ischemia-reperfusion injury and mimics ischemic preconditioning.


Assuntos
Precondicionamento Isquêmico , Isocitrato Desidrogenase/genética , Rim/irrigação sanguínea , Trifosfato de Adenosina/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Creatinina/sangue , Vetores Genéticos/administração & dosagem , Injeções Intravenosas , Isocitrato Desidrogenase/fisiologia , Túbulos Renais Proximais/citologia , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Consumo de Oxigênio , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Recidiva , Transfecção , Regulação para Cima
9.
Am J Physiol Regul Integr Comp Physiol ; 314(2): R265-R273, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29118018

RESUMO

Acute kidney injury (AKI) is associated with high mortality rates and predisposes development of chronic kidney disease (CKD). Distant organ damage, particularly in the lung, may contribute to mortality in AKI patients. Animal models of AKI demonstrate an increase in pulmonary infiltration of lymphocytes and reveal an acute compromise of lung function, but the chronic effects of AKI on pulmonary inflammation are unknown. We hypothesized that in response to renal ischemia/reperfusion (I/R), there is a persistent systemic increase in Th17 cells with potential effects on pulmonary structure and function. Renal I/R injury was performed on rats, and CKD progression was hastened by unilateral nephrectomy and exposure to 4.0% sodium diet between 35 and 63 days post-I/R. Th17 cells in peripheral blood showed a progressive increase up to 63 days after recovery from I/R injury. Infiltration of leukocytes including Th17 cells was also elevated in bronchiolar lavage (BAL) fluid 7 days after I/R and remained elevated for up to 63 days. Lung histology demonstrated an increase in alveolar cellularity and a significant increase in picrosirius red staining. Suppression of lymphocytes with mycophenolate mofetil (MMF) or an IL-17 antagonist significantly reduced Th17 cell infiltration and fibrosis in lung. In addition, tracheal smooth muscle contraction to acetylcholine was significantly enhanced 63-days after I/R relative to sham-operated controls. These data suggest that AKI is associated with a persistent increase in circulating and lung Th17 cells which may promote pulmonary fibrosis and the potential alteration in airway contractility.


Assuntos
Injúria Renal Aguda/imunologia , Pulmão/imunologia , Pneumonia/imunologia , Fibrose Pulmonar/imunologia , Insuficiência Renal Crônica/imunologia , Células Th17/imunologia , Injúria Renal Aguda/complicações , Injúria Renal Aguda/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Imunossupressores/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Contração Muscular , Músculo Liso/imunologia , Músculo Liso/patologia , Músculo Liso/fisiopatologia , Fenótipo , Pneumonia/etiologia , Pneumonia/patologia , Pneumonia/fisiopatologia , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Ratos Nus , Ratos Sprague-Dawley , Ratos Transgênicos , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/patologia , Fatores de Risco , Sódio na Dieta/toxicidade , Células Th17/efeitos dos fármacos , Fatores de Tempo , Traqueia/imunologia , Traqueia/patologia , Traqueia/fisiopatologia
11.
J Am Soc Nephrol ; 28(7): 2081-2092, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28122967

RESUMO

Highly aerobic organs like the kidney are innately susceptible to ischemia-reperfusion (I/R) injury, which can originate from sources including myocardial infarction, renal trauma, and transplant. Therapy is mainly supportive and depends on the cause(s) of damage. In the absence of hypervolemia, intravenous fluid delivery is frequently the first course of treatment but does not reverse established AKI. Evidence suggests that disrupting leukocyte adhesion may prevent the impairment of renal microvascular perfusion and the heightened inflammatory response that exacerbate ischemic renal injury. We investigated the therapeutic potential of hydrodynamic isotonic fluid delivery (HIFD) to the left renal vein 24 hours after inducing moderate-to-severe unilateral IRI in rats. HIFD significantly increased hydrostatic pressure within the renal vein. When conducted after established AKI, 24 hours after I/R injury, HIFD produced substantial and statistically significant decreases in serum creatinine levels compared with levels in animals given an equivalent volume of saline via peripheral infusion (P<0.05). Intravital confocal microscopy performed immediately after HIFD showed improved microvascular perfusion. Notably, HIFD also resulted in immediate enhancement of parenchymal labeling with the fluorescent dye Hoechst 33342. HIFD also associated with a significant reduction in the accumulation of renal leukocytes, including proinflammatory T cells. Additionally, HIFD significantly reduced peritubular capillary erythrocyte congestion and improved histologic scores of tubular injury 4 days after IRI. Taken together, these results indicate that HIFD performed after establishment of AKI rapidly restores microvascular perfusion and small molecule accessibility, with improvement in overall renal function.


Assuntos
Hidratação/métodos , Hidrodinâmica , Soluções Isotônicas/administração & dosagem , Rim/irrigação sanguínea , Traumatismo por Reperfusão/terapia , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença
12.
J Cell Mol Med ; 21(7): 1420-1430, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28455887

RESUMO

Damage to endothelial cells contributes to acute kidney injury (AKI) by causing impaired perfusion, while the permanent loss of the capillary network following AKI has been suggested to promote chronic kidney disease. Therefore, strategies to protect renal vasculature may impact both short-term recovery and long-term functional preservation post-AKI. Human adipose stromal cells (hASCs) possess pro-angiogenic and anti-inflammatory properties and therefore have been tested as a therapeutic agent to treat ischaemic conditions. This study evaluated hASC potential to facilitate recovery from AKI with specific attention to capillary preservation and inflammation. Male Sprague Dawley rats were subjected to bilateral ischaemia/reperfusion and allowed to recover for either two or seven days. At the time of reperfusion, hASCs or vehicle was injected into the suprarenal abdominal aorta. hASC-treated rats had significantly greater survival compared to vehicle-treated rats (88.7% versus 69.3%). hASC treatment showed hastened recovery as demonstrated by lower creatinine levels at 48 hrs, while tubular damage was significantly reduced at 48 hrs. hASC treatment resulted in a significant decrease in total T cell and Th17 cell infiltration into injured kidneys at 2 days post-AKI, but an increase in accumulation of regulatory T cells. By day 7, hASC-treated rats showed significantly attenuated capillary rarefaction in the cortex (15% versus 5%) and outer medulla (36% versus 18%) compared to vehicle-treated rats as well as reduced accumulation of interstitial alpha-smooth muscle actin-positive myofibroblasts. These results suggest for the first time that hASCs improve recovery from I/R-induced injury by mechanisms that contribute to decrease in inflammation and preservation of peritubular capillaries.


Assuntos
Injúria Renal Aguda/terapia , Inflamação/terapia , Traumatismo por Reperfusão/terapia , Células Estromais/transplante , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/fisiopatologia , Adipócitos/imunologia , Adipócitos/transplante , Tecido Adiposo/imunologia , Tecido Adiposo/transplante , Animais , Modelos Animais de Doenças , Humanos , Inflamação/fisiopatologia , Rim/imunologia , Rim/patologia , Rarefação Microvascular/imunologia , Rarefação Microvascular/fisiopatologia , Rarefação Microvascular/terapia , Ratos , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/fisiopatologia , Células Estromais/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
13.
Am J Physiol Renal Physiol ; 312(5): F897-F907, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28228404

RESUMO

Damage to endothelial cells contributes to acute kidney injury (AKI) by leading to impaired perfusion. Endothelial colony-forming cells (ECFC) are endothelial precursor cells with high proliferative capacity, pro-angiogenic activity, and in vivo vessel forming potential. We hypothesized that ECFC may ameliorate the degree of AKI and/or promote repair of the renal vasculature following ischemia-reperfusion (I/R). Rat pulmonary microvascular endothelial cells (PMVEC) with high proliferative potential were compared with pulmonary artery endothelial cells (PAEC) with low proliferative potential in rats subjected to renal I/R. PMVEC administration reduced renal injury and hastened recovery as indicated by serum creatinine and tubular injury scores, while PAEC did not. Vehicle-treated control animals showed consistent reductions in renal medullary blood flow (MBF) within 2 h of reperfusion, while PMVEC protected against loss in MBF as measured by laser Doppler. Interestingly, PMVEC mediated protection occurred in the absence of homing to the kidney. Conditioned medium (CM) from human cultured cord blood ECFC also conveyed beneficial effects against I/R injury and loss of MBF. Moreover, ECFC-CM significantly reduced the expression of ICAM-1 and decreased the number of differentiated lymphocytes typically recruited into the kidney following renal ischemia. Taken together, these data suggest that ECFC secrete factors that preserve renal function post ischemia, in part, by preserving microvascular function.


Assuntos
Injúria Renal Aguda/cirurgia , Proliferação de Células , Células Progenitoras Endoteliais/transplante , Endotélio Vascular/transplante , Rim/irrigação sanguínea , Neovascularização Fisiológica , Traumatismo por Reperfusão/cirurgia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Velocidade do Fluxo Sanguíneo , Comunicação Celular , Células Cultivadas , Quimiotaxia de Leucócito , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Sangue Fetal/citologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Fenótipo , Ratos Sprague-Dawley , Circulação Renal , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais , Fatores de Tempo
14.
Am J Physiol Renal Physiol ; 312(3): F385-F397, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27852609

RESUMO

T cells have been implicated in the pathogenesis of acute kidney injury (AKI) and its progression to chronic kidney disease (CKD). Previous studies suggest that Th17 cells participate during the AKI-to-CKD transition, and inhibition of T cell activity by mycophenolate mofetil (MMF) or losartan attenuates the development of fibrosis following AKI. We hypothesized that T cell-deficient rats may have reduced levels of IL-17 cytokine leading to decreased fibrosis following AKI. Renal ischemis-reperfusion (I/R) was performed on T cell-deficient athymic rats (Foxn1rnu-/rnu-) and control euthymic rats (Foxn1rnu-/+), and CKD progression was hastened by unilateral nephrectomy at day 33 and subsequent exposure to 4.0% sodium diet. Renal fibrosis developed in euthymic rats and was reduced by MMF treatment. Athymic rats exhibited a similar degree of fibrosis, but this was unaffected by MMF treatment. FACS analysis demonstrated that the number of IL-17+ cells was similar between postischemic athymic vs. euthymic rats. The source of IL-17 production in euthymic rats was predominately from conventional T cells (CD3+/CD161-). In the absence of conventional T cells in athymic rats, a compensatory pathway involving natural killer cells (CD3-/CD161+) was the primary source of IL-17. Blockade of IL-17 activity using IL-17Rc receptor significantly decreased fibrosis and neutrophil recruitment in both euthymic and athymic rats compared with vehicle-treated controls. Taken together, these data suggest that IL-17 secretion participates in the pathogenesis of AKI-induced fibrosis possibly via the recruitment of neutrophils and that the source of IL-17 may be from either conventional T cells or NK cells.


Assuntos
Injúria Renal Aguda/imunologia , Interleucina-17/imunologia , Rim/imunologia , Células Matadoras Naturais/imunologia , Infiltração de Neutrófilos , Neutrófilos/imunologia , Insuficiência Renal Crônica/imunologia , Traumatismo por Reperfusão/imunologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Fatores de Transcrição Forkhead/genética , Genótipo , Interleucina-17/metabolismo , Rim/metabolismo , Rim/patologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária , Masculino , Neutrófilos/metabolismo , Fenótipo , Ratos Nus , Ratos Transgênicos , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Fatores de Tempo
15.
J Am Soc Nephrol ; 27(3): 687-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26519085

RESUMO

Recent clinical studies indicate a strong link between AKI and progression of CKD. The increasing prevalence of AKI must compel the nephrology community to consider the long-term ramifications of this syndrome. Considerable gaps in knowledge exist regarding the connection between AKI and CKD. The 13th Acute Dialysis Quality Initiative meeting entitled "Therapeutic Targets of Human Acute Kidney Injury: Harmonizing Human and Experimental Animal Acute Kidney Injury" convened in April of 2014 and assigned a working group to focus on issues related to progression after AKI. This article provides a summary of the key conclusions and recommendations of the group, including an emphasis on terminology related to injury and repair processes for both clinical and preclinical studies, elucidation of pathophysiologic alterations of AKI, identification of potential treatment strategies, identification of patients predisposed to progression, and potential management strategies.


Assuntos
Injúria Renal Aguda/complicações , Progressão da Doença , Rim/patologia , Insuficiência Renal Crônica/etiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Animais , Biomarcadores/sangue , Biomarcadores/urina , Pontos de Checagem do Ciclo Celular , Reparo do DNA , Fibrose , Humanos , Hipóxia/complicações , Mitocôndrias/fisiologia , Estresse Oxidativo , Insuficiência Renal Crônica/terapia , Terminologia como Assunto
16.
Kidney Int ; 88(4): 776-84, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26200947

RESUMO

Exposure of rats to elevated dietary salt following recovery from acute kidney injury (AKI) accelerates the transition to chronic kidney disease (CKD), and is dependent on lymphocyte activity. Here we tested whether high salt diet triggers lymphocyte activation in postischemic kidneys to worsen renal inflammation and fibrosis. Male Sprague-Dawley rats on a 0.4% salt diet were subjected to left unilateral ischemia-reperfusion and allowed to recover for 5 weeks. This resulted in a mild elevation of CD4(+) T cells relative to sham animals. Contralateral unilateral nephrectomy and elevated dietary salt (4%) for 4 extra weeks hastened CKD and interstitial fibrosis. Activated T cells were increased in the kidney threefold after 4 weeks of elevated dietary salt exposure relative to post-AKI rats before salt feeding. The T cell subset was largely positive for IL-17, indicative of Th-17 cells. Because angiotensin II activity may influence lymphocyte activation, injured rats were given the AT1R antagonist, losartan, along with high salt diet. This significantly reduced the number of renal Th-17 cells to levels of sham rats, and significantly reduced the salt-induced increase in fibrosis to about half. In vitro studies in AKI-primed CD4(+) T cells indicated that angiotensin II and extracellular sodium enhanced, and losartan inhibited, IL-17 expression. Thus, dietary salt modulates immune cell activity in postischemic recovering kidneys because of the activity of local RAS, suggesting the participation of these cells in CKD progression post-AKI.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Rim/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Insuficiência Renal Crônica/prevenção & controle , Sistema Renina-Angiotensina/efeitos dos fármacos , Cloreto de Sódio na Dieta , Células Th17/efeitos dos fármacos , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Angiotensina II/farmacologia , Animais , Células Cultivadas , Citoproteção , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Rim/imunologia , Rim/metabolismo , Rim/patologia , Losartan/farmacologia , Masculino , Nefrectomia , Ratos Sprague-Dawley , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Células Th17/imunologia , Células Th17/metabolismo , Fatores de Tempo
17.
J Urol ; 193(4): 1409-16, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25245490

RESUMO

PURPOSE: We performed a pilot study to assess whether renal shock wave lithotripsy influences metabolic syndrome onset and severity. MATERIALS AND METHODS: Three-month-old juvenile female Ossabaw miniature pigs were treated with shock wave lithotripsy (2,000 shock waves at 24 kV with 120 shock waves per minute in 2) or sham shock wave lithotripsy (no shock waves in 2). Shock waves were targeted to the upper pole of the left kidney to model treatment that would also expose the pancreatic tail to shock waves. Pigs were then instrumented to directly measure arterial blood pressure via an implanted radiotelemetry device. They later received a hypercaloric atherogenic diet for about 7 months. Metabolic syndrome development was assessed by the intravenous glucose tolerance test. RESULTS: Metabolic syndrome progression and severity were similar in the sham treated and lithotripsy groups. The only exception arterial blood pressure, which remained relatively constant in sham treated pigs but began to increase at about 2 months towards hypertensive levels in lithotripsy treated pigs. Metabolic data on the 2 groups were pooled to provide a more complete assessment of metabolic syndrome development and progression in this juvenile pig model. The intravenous glucose tolerance test revealed substantial insulin resistance with impaired glucose tolerance within 2 months on the hypercaloric atherogenic diet with signs of further metabolic impairment at 7 months. CONCLUSIONS: These preliminary results suggest that renal shock wave lithotripsy is not a risk factor for worsening glucose tolerance or diabetes mellitus onset. However, it appears to be a risk factor for early onset hypertension in metabolic syndrome.


Assuntos
Litotripsia/efeitos adversos , Síndrome Metabólica/etiologia , Animais , Modelos Animais de Doenças , Feminino , Projetos Piloto , Risco , Suínos , Porco Miniatura
18.
J Cell Physiol ; 229(1): 10-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23794280

RESUMO

Progenitor cells for the endothelial lineage have been widely investigated for more than a decade, but continue to be controversial since no unique identifying marker has yet been identified. This review will begin with a discussion of the basic tenets originally proposed for proof that a cell displays properties of an endothelial progenitor cell. We then provide an overview of the methods for putative endothelial progenitor cell derivation, expansion, and enumeration. This discussion includes consideration of cells that are present in the circulation as well as cells resident in the vascular endothelial intima. Finally, we provide some suggested changes in nomenclature that would greatly clarify and demystify the cellular elements involved in vascular repair.


Assuntos
Diferenciação Celular , Células Endoteliais/citologia , Endotélio Vascular/citologia , Células-Tronco/citologia , Linhagem da Célula , Células Endoteliais/classificação , Células Endoteliais/metabolismo , Endotélio Vascular/crescimento & desenvolvimento , Humanos , Células-Tronco/classificação , Células-Tronco/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/patologia , Doenças Vasculares/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA