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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 326(5): F839-F854, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38450434

RESUMEN

Resident memory T cells (TRMs), which are memory T cells that are retained locally within tissues, have recently been described as antigen-specific frontline defenders against pathogens in barrier and nonbarrier epithelial tissues. They have also been noted for perpetuating chronic inflammation. The conditions responsible for TRM differentiation are still poorly understood, and their contributions, if any, to sterile models of chronic kidney disease (CKD) remain a mystery. In this study, we subjected male C57BL/6J mice and OT-1 transgenic mice to five consecutive days of 2 mg/kg aristolochic acid (AA) injections intraperitoneally to induce CKD or saline injections as a control. We evaluated their kidney immune profiles at 2 wk, 6 wk, and 6 mo after treatment. We identified a substantial population of TRMs in the kidneys of mice with AA-induced CKD. Flow cytometry of injured kidneys showed T cells bearing TRM surface markers and single-cell (sc) RNA sequencing revealed these cells as expressing well-known TRM transcription factors and receptors responsible for TRM differentiation and maintenance. Although kidney TRMs expressed Cd44, a marker of antigen experience and T cell activation, their derivation was independent of cognate antigen-T cell receptor interactions, as the kidneys of transgenic OT-1 mice still harbored considerable proportions of TRMs after injury. Our results suggest a nonantigen-specific or antigen-independent mechanism capable of generating TRMs in the kidney and highlight the need to better understand TRMs and their involvement in CKD.NEW & NOTEWORTHY Resident memory T cells (TRMs) differentiate and are retained within the kidneys of mice with aristolochic acid (AA)-induced chronic kidney disease (CKD). Here, we characterized this kidney TRM population and demonstrated TRM derivation in the kidneys of OT-1 transgenic mice with AA-induced CKD. A better understanding of TRMs and the processes by which they can differentiate independent of antigen may help our understanding of the interactions between the immune system and kidneys.


Asunto(s)
Ácidos Aristolóquicos , Diferenciación Celular , Riñón , Células T de Memoria , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica , Animales , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Masculino , Ácidos Aristolóquicos/toxicidad , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Ratones Transgénicos , Memoria Inmunológica , Modelos Animales de Enfermedad , Ratones
2.
J Pharmacol Exp Ther ; 388(2): 605-612, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37699712

RESUMEN

Arsenicals are deadly chemical warfare agents that primarily cause death through systemic capillary fluid leakage and hypovolemic shock. Arsenical exposure is also known to cause acute kidney injury, a condition that contributes to arsenical-associated death due to the necessity of the kidney in maintaining whole-body fluid homeostasis. Because of the global health risk that arsenicals pose, a nuanced understanding of how arsenical exposure can lead to kidney injury is needed. We used a nontargeted transcriptional approach to evaluate the effects of cutaneous exposure to phenylarsine oxide, a common arsenical, in a murine model. Here we identified an upregulation of metabolic pathways such as fatty acid oxidation, fatty acid biosynthesis, and peroxisome proliferator-activated receptor (PPAR)-α signaling in proximal tubule epithelial cell and endothelial cell clusters. We also revealed highly upregulated genes such as Zbtb16, Cyp4a14, and Pdk4, which are involved in metabolism and metabolic switching and may serve as future therapeutic targets. The ability of arsenicals to inhibit enzymes such as pyruvate dehydrogenase has been previously described in vitro. This, along with our own data, led us to conclude that arsenical-induced acute kidney injury may be due to a metabolic impairment in proximal tubule and endothelial cells and that ameliorating these metabolic effects may lead to the development of life-saving therapies. SIGNIFICANCE STATEMENT: In this study, we demonstrate that cutaneous arsenical exposure leads to a transcriptional shift enhancing fatty acid metabolism in kidney cells, indicating that metabolic alterations might mechanistically link topical arsenical exposure to acute kidney injury. Targeting metabolic pathways may generate promising novel therapeutic approaches in combating arsenical-induced acute kidney injury.


Asunto(s)
Lesión Renal Aguda , Arsenicales , Ratones , Humanos , Animales , Células Endoteliales/metabolismo , Riñón/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Arsenicales/efectos adversos , Arsenicales/metabolismo
3.
Mol Med ; 29(1): 67, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37217845

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is one of the most prevalent monogenic human diseases. It is mostly caused by pathogenic variants in PKD1 or PKD2 genes that encode interacting transmembrane proteins polycystin-1 (PC1) and polycystin-2 (PC2). Among many pathogenic processes described in ADPKD, those associated with cAMP signaling, inflammation, and metabolic reprogramming appear to regulate the disease manifestations. Tolvaptan, a vasopressin receptor-2 antagonist that regulates cAMP pathway, is the only FDA-approved ADPKD therapeutic. Tolvaptan reduces renal cyst growth and kidney function loss, but it is not tolerated by many patients and is associated with idiosyncratic liver toxicity. Therefore, additional therapeutic options for ADPKD treatment are needed. METHODS: As drug repurposing of FDA-approved drug candidates can significantly decrease the time and cost associated with traditional drug discovery, we used the computational approach signature reversion to detect inversely related drug response gene expression signatures from the Library of Integrated Network-Based Cellular Signatures (LINCS) database and identified compounds predicted to reverse disease-associated transcriptomic signatures in three publicly available Pkd2 kidney transcriptomic data sets of mouse ADPKD models. We focused on a pre-cystic model for signature reversion, as it was less impacted by confounding secondary disease mechanisms in ADPKD, and then compared the resulting candidates' target differential expression in the two cystic mouse models. We further prioritized these drug candidates based on their known mechanism of action, FDA status, targets, and by functional enrichment analysis. RESULTS: With this in-silico approach, we prioritized 29 unique drug targets differentially expressed in Pkd2 ADPKD cystic models and 16 prioritized drug repurposing candidates that target them, including bromocriptine and mirtazapine, which can be further tested in-vitro and in-vivo. CONCLUSION: Collectively, these results indicate drug targets and repurposing candidates that may effectively treat pre-cystic as well as cystic ADPKD.


Asunto(s)
Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Humanos , Ratones , Reposicionamiento de Medicamentos , Expresión Génica , Riñón/metabolismo , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/complicaciones , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Tolvaptán/farmacología , Tolvaptán/uso terapéutico , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
4.
J Am Soc Nephrol ; 33(4): 747-768, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35110364

RESUMEN

BACKGROUND: Inducible disruption of cilia-related genes in adult mice results in slowly progressive cystic disease, which can be greatly accelerated by renal injury. METHODS: To identify in an unbiased manner modifier cells that may be influencing the differential rate of cyst growth in injured versus non-injured cilia mutant kidneys at a time of similar cyst severity, we generated a single-cell atlas of cystic kidney disease. We conducted RNA-seq on 79,355 cells from control mice and adult-induced conditional Ift88 mice (hereafter referred to as cilia mutant mice) that were harvested approximately 7 months post-induction or 8 weeks post 30-minute unilateral ischemia reperfusion injury. RESULTS: Analyses of single-cell RNA-seq data of CD45+ immune cells revealed that adaptive immune cells differed more in cluster composition, cell proportion, and gene expression than cells of myeloid origin when comparing cystic models with one another and with non-cystic controls. Surprisingly, genetic deletion of adaptive immune cells significantly reduced injury-accelerated cystic disease but had no effect on cyst growth in non-injured cilia mutant mice, independent of the rate of cyst growth or underlying genetic mutation. Using NicheNet, we identified a list of candidate cell types and ligands that were enriched in injured cilia mutant mice compared with aged cilia mutant mice and non-cystic controls that may be responsible for the observed dependence on adaptive immune cells during injury-accelerated cystic disease. CONCLUSIONS: Collectively, these data highlight the diversity of immune cell involvement in cystic kidney disease.


Asunto(s)
Quistes , Enfermedades Renales Poliquísticas , Animales , Cilios/metabolismo , Quistes/genética , Riñón/metabolismo , Ratones , Mutación , Enfermedades Renales Poliquísticas/metabolismo
5.
Am J Transplant ; 22(4): 1037-1053, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35049121

RESUMEN

A radical solution is needed for the organ supply crisis, and the domestic pig is a promising organ source. In preparation for a clinical trial of xenotransplantation, we developed an in vivo pre-clinical human model to test safety and feasibility tenets established in animal models. After performance of a novel, prospective compatible crossmatch, we performed bilateral native nephrectomies in a human brain-dead decedent and subsequently transplanted two kidneys from a pig genetically engineered for human xenotransplantation. The decedent was hemodynamically stable through reperfusion, and vascular integrity was maintained despite the exposure of the xenografts to human blood pressure. No hyperacute rejection was observed, and the kidneys remained viable until termination 74 h later. No chimerism or transmission of porcine retroviruses was detected. Longitudinal biopsies revealed thrombotic microangiopathy that did not progress in severity, without evidence of cellular rejection or deposition of antibody or complement proteins. Although the xenografts produced variable amounts of urine, creatinine clearance did not recover. Whether renal recovery was impacted by the milieu of brain death and/or microvascular injury remains unknown. In summary, our study suggests that major barriers to human xenotransplantation have been surmounted and identifies where new knowledge is needed to optimize xenotransplantation outcomes in humans.


Asunto(s)
Rechazo de Injerto , Riñón , Animales , Animales Modificados Genéticamente , Rechazo de Injerto/patología , Xenoinjertos , Humanos , Estudios Prospectivos , Porcinos , Trasplante Heterólogo
6.
Am J Physiol Renal Physiol ; 321(2): F162-F169, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34180717

RESUMEN

Kidney resident macrophages (KRMs) are involved in maintaining renal homeostasis and in controlling the pathological outcome of acute kidney injury and cystic kidney disease in mice. In adult mice, KRMs maintain their population through self-renewal with little or no input from the peripheral blood. Despite recent data suggesting that a transcriptionally similar population of KRM-like cells is present across species, the idea that they are self-renewing and minimally dependent on peripheral blood input in other species has yet to be proven due to the lack of an appropriate model and cross-species expression markers. In this study, we used our recently identified cross-species KRM cell surface markers and parabiosis surgery in inbred Lewis rats to determine if rat KRMs are maintained independent of peripheral blood input, similar to their mouse counterparts. Flow cytometry analysis indicated that parabiosis surgery in the rat results in the establishment of chimerism of T/B cells, neutrophils, and monocyte-derived infiltrating macrophages in the blood, spleen, and kidney 3 wk after parabiosis surgery. Analysis of KRMs using the cell surface markers CD81 and C1q indicated that these cells have minimal chimerism and, therefore, receive little input from the peripheral blood. These data indicate that KRM properties are conserved in at least two different species.NEW & NOTEWORTHY In this report, we performed parabiosis surgery on inbred Lewis rats and showed that rat kidney resident macrophages (KRMs), identified using our novel cross-species markers, are minimally dependent on peripheral blood input. Thus, for the first time, to our knowledge, we confirm that a hallmark of mouse KRMs is also present in KRMs isolated from another species.


Asunto(s)
Riñón/citología , Macrófagos/citología , Monocitos/citología , Animales , Femenino , Masculino , Parabiosis , Ratas , Ratas Endogámicas Lew , Bazo/citología
7.
Am J Physiol Renal Physiol ; 320(5): F706-F718, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33719570

RESUMEN

Cellular metabolic rates in the kidney are critical for maintaining normal renal function. In a hypoxic milieu, cells rely on glycolysis to meet energy needs, resulting in the generation of pyruvate and NADH. In the absence of oxidative phosphorylation, the continuation of glycolysis is dependent on the regeneration of NAD+ from NADH accompanied by the fermentation of pyruvate to lactate. This reaction is catalyzed by lactate dehydrogenase (LDH) isoform A (LDHA), whereas LDH isoform B (LDHB) catalyzes the opposite reaction. LDH is widely used as a potential injury marker as it is released from damaged cells into the urine and serum; however, the precise isoform-specific cellular localization of the enzyme along the nephron has not been characterized. By combining immunohistochemistry results and single-cell RNA-sequencing data on healthy mouse kidneys, we identified that LDHA is primarily expressed in proximal segments, whereas LDHB is expressed in the distal parts of the nephron. In vitro experiments in mouse and human renal proximal tubule cells showed an increase in LDHA following hypoxia with no change in LDHB. Using immunofluorescence, we observed that the overall expression of both LDHA and LDHB proteins decreased following renal ischemia-reperfusion injury as well as in the adenine-diet-induced model of chronic kidney disease. Single-nucleus RNA-sequencing analyses of kidneys following ischemia-reperfusion injury revealed a significant decline in the number of cells expressing detectable levels of Ldha and Ldhb; however, cells that were positive showed increased average expression postinjury, which subsided during the recovery phase. These data provide information on the cell-specific expression of LDHA and LDHB in the normal kidney as well as following acute and chronic kidney disease.NEW & NOTEWORTHY Cellular release of lactate dehydrogenase (LDH) is being used as an injury marker; however, the exact localization of LDH within the nephron remains unclear. We show that LDH isoform A is expressed proximally, whereas isoform B is expressed distally. Both subunit expressions were significantly altered in models of acute kidney injury and chronic kidney disease. Our study provides new insights into basal and postinjury renal lactate metabolism.


Asunto(s)
Lesión Renal Aguda/enzimología , Riñón/enzimología , L-Lactato Deshidrogenasa/metabolismo , Insuficiencia Renal Crónica/enzimología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Biomarcadores/metabolismo , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Humanos , Isoenzimas , Riñón/patología , L-Lactato Deshidrogenasa/genética , Masculino , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Factores de Tiempo
8.
J Am Soc Nephrol ; 30(5): 767-781, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30948627

RESUMEN

BACKGROUND: Resident macrophages regulate homeostatic and disease processes in multiple tissues, including the kidney. Despite having well defined markers to identify these cells in mice, technical limitations have prevented identification of a similar cell type across species. The inability to identify resident macrophage populations across species hinders the translation of data obtained from animal model to human patients. METHODS: As an entry point to determine novel markers that could identify resident macrophages across species, we performed single-cell RNA sequencing (scRNAseq) analysis of all T and B cell-negative CD45+ innate immune cells in mouse, rat, pig, and human kidney tissue. RESULTS: We identified genes with enriched expression in mouse renal resident macrophages that were also present in candidate resident macrophage populations across species. Using the scRNAseq data, we defined a novel set of possible cell surface markers (Cd74 and Cd81) for these candidate kidney resident macrophages. We confirmed, using parabiosis and flow cytometry, that these proteins are indeed enriched in mouse resident macrophages. Flow cytometry data also indicated the existence of a defined population of innate immune cells in rat and human kidney tissue that coexpress CD74 and CD81, suggesting the presence of renal resident macrophages in multiple species. CONCLUSIONS: Based on transcriptional signatures, our data indicate that there is a conserved population of innate immune cells across multiple species that have been defined as resident macrophages in the mouse. Further, we identified potential cell surface markers to allow for future identification and characterization of this candidate resident macrophage population in mouse, rat, and pig translational studies.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunidad Innata/genética , Molécula 1 de Adhesión Intercelular/inmunología , Macrófagos/metabolismo , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Células Cultivadas , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Parabiosis , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia de ARN , Especificidad de la Especie
9.
J Am Soc Nephrol ; 30(10): 1841-1856, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31337691

RESUMEN

BACKGROUND: Mutations affecting cilia proteins have an established role in renal cyst formation. In mice, the rate of cystogenesis is influenced by the age at which cilia dysfunction occurs and whether the kidney has been injured. Disruption of cilia function before postnatal day 12-14 results in rapid cyst formation; however, cyst formation is slower when cilia dysfunction is induced after postnatal day 14. Rapid cyst formation can also be induced in conditional adult cilia mutant mice by introducing renal injury. Previous studies indicate that macrophages are involved in cyst formation, however the specific role and type of macrophages responsible has not been clarified. METHODS: We analyzed resident macrophage number and subtypes during postnatal renal maturation and after renal injury in control and conditional Ift88 cilia mutant mice. We also used a pharmacological inhibitor of resident macrophage proliferation and accumulation to determine the importance of these cells during rapid cyst formation. RESULTS: Our data show that renal resident macrophages undergo a phenotypic switch from R2b (CD11clo) to R2a (CD11chi) during postnatal renal maturation. The timing of this switch correlates with the period in which cyst formation transitions from rapid to slow following induction of cilia dysfunction. Renal injury induces the reaccumulation of juvenile-like R2b resident macrophages in cilia mutant mice and restores rapid cystogenesis. Loss of primary cilia in injured conditional Ift88 mice results in enhanced epithelial production of membrane-bound CSF1, a cytokine that promotes resident macrophage proliferation. Inhibiting CSF1/CSF1-receptor signaling with a CSF1R kinase inhibitor reduces resident macrophage proliferation, R2b resident macrophage accumulation, and renal cyst formation in two mouse models of cystic disease. CONCLUSIONS: These data uncover an important pathogenic role for resident macrophages during rapid cyst progression.


Asunto(s)
Enfermedades Renales Quísticas/etiología , Macrófagos/fisiología , Animales , Cilios/genética , Femenino , Riñón/crecimiento & desarrollo , Macrófagos/clasificación , Masculino , Ratones , Mutación
10.
Am J Physiol Renal Physiol ; 314(5): F702-F714, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515173

RESUMEN

Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.


Asunto(s)
Lesión Renal Aguda/enzimología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/enzimología , Proteínas de la Membrana/metabolismo , Acetilcisteína/farmacología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Antioxidantes/farmacología , Carbolinas/toxicidad , Muerte Celular , Línea Celular , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Relación Dosis-Respuesta a Droga , Compuestos Férricos/toxicidad , Glutatión/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Quelantes del Hierro/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Noqueados , Fenilendiaminas/farmacología , Piperazinas/toxicidad , Compuestos de Amonio Cuaternario/toxicidad , Transducción de Señal , Factores de Tiempo
11.
Lab Invest ; 98(3): 391-402, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29251733

RESUMEN

The immune cellular compartment of the kidney is involved in organ development and homeostasis, as well as in many pathological conditions. Little is known about the mechanisms that drive intrarenal immune responses in the presence of renal tubular and interstitial cell death. However, it is known that tissue-resident leukocytes have the potential to have distinct roles compared with circulating cells. We used a parabiosis model in C57BL/6 CD45 congenic and green fluorescent protein transgenic mice to better understand the dynamics of immune cells in the kidney. We found F4/80Hi intrarenal macrophages exhibit minimal exchange with the peripheral circulation in two models of parabiosis, whether mice were attached for 4 or 16 weeks. Other intrarenal inflammatory cells demonstrate near total exchange with the circulating immune cell pool in healthy kidneys, indicating that innate and adaptive immune cells extensively traffic through the kidney interstitium during normal physiology. Neutrophils, dendritic cells, F4/80Low macrophages, T cells, B cells, and NK cells are renewed from the circulating immune cell pool. However, a fraction of double-negative T (CD4- CD8-) and NKT cells are long-lived or tissue resident. This study provides direct evidence of leukocyte sub-populations that are resident in the renal tissue, cells which demonstrate minimal to no exchange with the peripheral blood. In addition, the data demonstrate continual exchange of other sub-populations through uninflamed tissue.


Asunto(s)
Riñón/inmunología , Linfocitos/fisiología , Parabiosis , Animales , Quimerismo , Ratones Endogámicos C57BL , Bazo/inmunología
12.
Am J Physiol Renal Physiol ; 312(4): F640-F646, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28100500

RESUMEN

Mononuclear phagocytes are the most common cells in the kidney associated with immunity and inflammation. Although the presence of these cells in the kidney has been known for decades, the study of mononuclear phagocytes in the context of kidney function and dysfunction is still at an early stage. The purpose of this review is to summarize the present knowledge regarding classification of these cells in the mouse kidney and to identify relevant questions that would further advance the field and potentially lead to new opportunities for treatment of acute kidney injury and other kidney diseases.


Asunto(s)
Lesión Renal Aguda/inmunología , Plasticidad de la Célula , Riñón/inmunología , Nefritis/inmunología , Fagocitos/inmunología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Biomarcadores/metabolismo , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Ratones , Nefritis/metabolismo , Nefritis/patología , Fagocitos/clasificación , Fagocitos/metabolismo , Fagocitos/patología , Fenotipo , Receptores Inmunológicos/metabolismo , Transducción de Señal
13.
Hum Mol Genet ; 24(14): 4078-93, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25904107

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common known genetic cause of Parkinson's disease, and LRRK2 is also linked to Crohn's and Hansen's disease. LRRK2 is expressed in many organs in mammals but is particularly abundant in the kidney. We find that LRRK2 protein is predominantly localized to collecting duct cells in the rat kidney, with much lower expression in other kidney cells. While genetic knockout (KO) of LRRK2 expression is well-tolerated in mice and rats, a unique age-dependent pathology develops in the kidney. The cortex and medulla of LRRK2 KO rat kidneys become darkly pigmented in early adulthood, yet aged animals display no overt signs of kidney failure. Accompanying the dark pigment we find substantial macrophage infiltration in LRRK2 KO kidneys, suggesting the presence of chronic inflammation that may predispose to kidney disease. Unexpectedly, the dark kidneys of the LRRK2 KO rats are highly resistant to rhabdomyolysis-induced acute kidney injury compared with wild-type rats. Biochemical profiling of the LRRK2 KO kidneys using immunohistochemistry, proteomic and lipidomic analyses show a massive accumulation of hemoglobin and lipofuscin in renal tubules that account for the pigmentation. The proximal tubules demonstrate a corresponding up-regulation of the cytoprotective protein heme oxygenase-1 (HO-1) which is capable of mitigating acute kidney injury. The unusual kidney pathology of LRRK2 KO rats highlights several novel physiological roles for LRRK2 and provides indirect evidence for HO-1 expression as a protective mechanism in acute kidney injury in LRRK2 deficiency.


Asunto(s)
Enfermedades Renales/genética , Proteínas Serina-Treonina Quinasas/genética , Rabdomiólisis/genética , Animales , Citoprotección , Células Epiteliales/metabolismo , Predisposición Genética a la Enfermedad , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Enfermedades Renales/etiología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica , Ratas , Rabdomiólisis/complicaciones , Regulación hacia Arriba
14.
J Am Soc Nephrol ; 26(9): 2139-51, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25677389

RESUMEN

Renal ischemia-reperfusion injury is mediated by a complex cascade of events, including the immune response, that occur secondary to injury to renal epithelial cells. We tested the hypothesis that heme oxygenase-1 (HO-1) expression, which is protective in ischemia-reperfusion injury, regulates trafficking of myeloid-derived immune cells in the kidney. Age-matched male wild-type (HO-1(+/+)), HO-1-knockout (HO-1(-/-)), and humanized HO-1-overexpressing (HBAC) mice underwent bilateral renal ischemia for 10 minutes. Ischemia-reperfusion injury resulted in significantly worse renal structure and function and increased mortality in HO-1(-/-) mice. In addition, there were more macrophages (CD45(+) CD11b(hi)F4/80(lo)) and neutrophils (CD45(+) CD11b(hi) MHCII(-) Gr-1(hi)) in HO-1(-/-) kidneys than in sham and HO-1(+/+) control kidneys subjected to ischemia-reperfusion. However, ischemic injury resulted in a significant decrease in the intrarenal resident dendritic cell (DC; CD45(+)MHCII(+)CD11b(lo)F4/80(hi)) population in HO-1(-/-) kidneys compared with controls. Syngeneic transplant experiments utilizing green fluorescent protein-positive HO-1(+/+) or HO-1(-/-) donor kidneys and green fluorescent protein-negative HO-1(+/+) recipients confirmed increased migration of the resident DC population from HO-1(-/-) donor kidneys, compared to HO-1(+/+) donor kidneys, to the peripheral lymphoid organs. This effect on renal DC migration was corroborated in myeloid-specific HO-1(-/-) mice subjected to bilateral ischemia. These mice also displayed impaired renal recovery and increased fibrosis at day 7 after injury. These results highlight an important role for HO-1 in orchestrating the trafficking of myeloid cells in AKI, which may represent a key pathway for therapeutic intervention.


Asunto(s)
Lesión Renal Aguda/patología , Movimiento Celular/fisiología , Hemo-Oxigenasa 1/fisiología , Células Mieloides , Lesión Renal Aguda/etiología , Lesión Renal Aguda/fisiopatología , Animales , Movimiento Celular/genética , Células Dendríticas , Fibrosis , Hemo-Oxigenasa 1/genética , Inmunidad Innata , Interleucina-6/metabolismo , Isquemia/etiología , Riñón/irrigación sanguínea , Riñón/patología , Ganglios Linfáticos/patología , Macrófagos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Neutrófilos , Daño por Reperfusión/complicaciones , Bazo/patología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Lab Invest ; 95(2): 132-41, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25437645

RESUMEN

Atherosclerosis and valvular heart disease often require treatment with corrective surgery to prevent future myocardial infarction, ischemic heart disease, and heart failure. Mechanisms underlying the development of the associated complications of surgery are multifactorial and have been linked to inflammation and oxidative stress, classically as measured in the blood or plasma of patients. Postoperative pericardial fluid (PO-PCF) has not been investigated in depth with respect to the potential to induce oxidative stress. This is important because cardiac surgery disrupts the integrity of the pericardial membrane surrounding the heart and causes significant alterations in the composition of the pericardial fluid (PCF). This includes contamination with hemolyzed blood and high concentrations of oxidized hemoglobin, which suggests that cardiac surgery results in oxidative stress within the pericardial space. Accordingly, we tested the hypothesis that PO-PCF is highly pro-oxidant and that the potential interaction between inflammatory cell-derived hydrogen peroxide with hemoglobin is associated with oxidative stress. Blood and PCF were collected from 31 patients at the time of surgery and postoperatively from 4 to 48 h after coronary artery bypass grafting, valve replacement, or valve repair (mitral or aortic). PO-PCF contained high concentrations of neutrophils and monocytes, which are capable of generating elevated amounts of superoxide and hydrogen peroxide through the oxidative burst. In addition, PO-PCF primed naive neutrophils resulting in an enhanced oxidative burst upon stimulation. The PO-PCF also contained increased concentrations of cell-free oxidized hemoglobin that was associated with elevated levels of F2α isoprostanes and prostaglandins, consistent with both oxidative stress and activation of cyclooxygenase. Lastly, protein analysis of the PO-PCF revealed evidence of protein thiol oxidation and protein carbonylation. We conclude that PO-PCF is highly pro-oxidant and speculate that it may contribute to the risk of postoperative complications.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/efectos adversos , Líquido Extracelular/metabolismo , Hemoglobinas/metabolismo , Estrés Oxidativo/fisiología , Pericardio/fisiopatología , Complicaciones Posoperatorias/fisiopatología , Análisis de Varianza , Recuento de Células Sanguíneas , Electroforesis en Gel de Poliacrilamida , F2-Isoprostanos/metabolismo , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/metabolismo , Peroxidación de Lípido/fisiología , Espectrometría de Masas , Neutrófilos/metabolismo , Oxidación-Reducción , Pericardio/metabolismo , Carbonilación Proteica , Colorantes de Rosanilina , Compuestos de Sulfhidrilo/metabolismo
16.
Kidney Int ; 88(1): 95-108, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25874599

RESUMEN

Inflammation culminating in fibrosis contributes to progressive kidney disease. Cross-talk between the tubular epithelium and interstitial cells regulates inflammation by a coordinated release of cytokines and chemokines. Here we studied the role of heme oxygenase-1 (HO-1) and the heavy subunit of ferritin (FtH) in macrophage polarization and renal inflammation. Deficiency in HO-1 was associated with increased FtH expression, accumulation of macrophages with a dysregulated polarization profile, and increased fibrosis following unilateral ureteral obstruction in mice: a model of renal inflammation and fibrosis. Macrophage polarization in vitro was predominantly dependent on FtH expression in isolated bone marrow-derived mouse monocytes. Using transgenic mice with conditional deletion of FtH in the proximal tubules (FtH(PT-/-)) or myeloid cells (FtH(LysM-/-)), we found that myeloid FtH deficiency did not affect polarization or accumulation of macrophages in the injured kidney compared with wild-type (FtH(+/+)) controls. However, tubular FtH deletion led to a marked increase in proinflammatory macrophages. Furthermore, injured kidneys from FtH(PT-/-) mice expressed significantly higher levels of inflammatory chemokines and fibrosis compared with kidneys from FtH(+/+) and FtH(LysM-/-) mice. Thus, there are differential effects of FtH in macrophages and epithelial cells, which underscore the critical role of FtH in tubular-macrophage cross-talk during kidney injury.


Asunto(s)
Apoferritinas/genética , Células Epiteliales/metabolismo , Hemo-Oxigenasa 1/deficiencia , Riñón/patología , Macrófagos/fisiología , Células Mieloides/metabolismo , Nefritis/metabolismo , Animales , Apoferritinas/metabolismo , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica , Hemo-Oxigenasa 1/genética , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nefritis/etiología , ARN Mensajero/metabolismo , Obstrucción Ureteral/complicaciones
17.
Endoscopy ; 46(11): 986-91, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25290096

RESUMEN

Obscure overt gastrointestinal bleeding (OGIB) is a challenge in patients with left ventricular assist devices (LVADs). We evaluated the utility and safety of double-balloon enteroscopy (DBE) in patients with LVADs in an observational consecutive-patient cohort from a single tertiary referral center. Ten patients with LVADs underwent thirteen DBEs for obscure OGIB. The first OGIB event necessitating DBE occurred after a mean of 512 ± 363 days of LVAD support. All patients underwent DBE, eleven anterograde and two retrograde, with a mean insertion depth 176 ± 85 cm. Diagnostic yield was 69 % with the primary bleeding lesion most frequently found in the mid-bowel. The most common lesions were arteriovenous malformations. Therapeutic yield with argon plasma coagulation (APC), epinephrine injection, and/or hemoclip placement was 89 %. There were no procedure-related complications. DBE in patients with LVADs has good diagnostic yield and high therapeutic yield for obscure OGIB and is safe and well tolerated.


Asunto(s)
Enteroscopía de Doble Balón , Corazón Auxiliar/efectos adversos , Hemostasis Endoscópica , Enfermedades Intestinales/terapia , Melena/terapia , Anciano , Enteroscopía de Doble Balón/efectos adversos , Femenino , Humanos , Enfermedades Intestinales/diagnóstico , Enfermedades Intestinales/etiología , Masculino , Melena/diagnóstico , Melena/etiología , Persona de Mediana Edad
18.
Clin Transplant ; 28(9): 946-52, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24930563

RESUMEN

Data from Cardiac Transplant Research Database (CTRD) were analyzed from 1999 to 2006 to examine the effects of different induction strategies at the time of cardiac transplantation. A total of 2090 primary heart transplants were categorized by induction with interleukin-2 receptor blocker (IL-2RB), antithymocyte globulin (ATG), or no induction (NI). Probabilities for rejection and infection were estimated with parametric time-related models. Using these models, hazard was calculated for two theoretical patient profiles, one at lower risk for rejection and higher risk of infection (Profile 1) and higher risk for rejection and lower risk of infection (Profile 2). Of the 2090 transplants, 49.8% (1095) did not receive induction, 27.3% (599) received IL-2RB, and 18.0% (396) received ATG. Profile 1 patients had lower hazard for rejection with IL-2RB compared to ATG and NI (p < 0.01), but at the cost of increased risk of infection (5.0 vs. 1.8 vs. 1.6, respectively, at four wk, p < 0.01). Profile 2 patients experienced a fivefold decreased hazard for rejection when treated with IL-2RB compared with ATG and NI (p < 0.01). In patients at high risk of infection, IL-2RB reduced risk of rejection but at the expense of increased hazard for infection.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Enfermedad de la Arteria Coronaria/cirugía , Rechazo de Injerto/epidemiología , Trasplante de Corazón , Inmunosupresores/uso terapéutico , Infecciones/epidemiología , Receptores de Interleucina-2/antagonistas & inhibidores , Adulto , Estudios de Seguimiento , Supervivencia de Injerto/efectos de los fármacos , Humanos , Persona de Mediana Edad , Pronóstico , Inducción de Remisión , Factores de Riesgo
19.
Surgery ; 175(4): 1244-1246, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38123371

RESUMEN

Building a competitive research program within a department of surgery requires a significant commitment by the department and the institution to provide the necessary resources for faculty recruitment, retention of current faculty, and physical space/infrastructure to support research activities. We expanded the academic footprint of our department as demonstrated by the expansion of the department of surgery research funding by 13-fold over a period of 7 years, resulting in an increase in national ranking from 55th place to 10th place in the National Institutes of Health extramural funding. This required attention to multiple factors that affect the ability of faculty to establish and maintain competitive research programs. We executed a plan that established a leadership structure that coordinates resources and provides mentorship to faculty. The department invested heavily in the recruitment of new faculty, especially junior faculty, but also some mid-career and senior investigators to develop a critical mass in specific areas for competitive large grant and program project applications. The pipeline of new trainees interested in research was augmented by successful training grant applications that created a mechanism by which residents and fellows can pursue research for periods ranging from a few weeks to 2 years. Administrative infrastructure was created to assist faculty in grant submissions as well as post-award management. Finally, in partnership with institutional leadership, the department acquired the physical space necessary to support both dry-lab and wet-lab research activities. To achieve true excellence, an academic surgery department must maintain excellence in both the clinical and research areas, which, in the context of an academic medical center, are not separate goals.


Asunto(s)
Investigación Biomédica , Mentores , Estados Unidos , Humanos , Docentes , National Institutes of Health (U.S.) , Centros Médicos Académicos , Liderazgo
20.
bioRxiv ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38559147

RESUMEN

Uterine natural killer cells (uNKs) are a tissue resident lymphocyte population that are critical for pregnancy success. Although mouse models have demonstrated that NK deficiency results in abnormal placentation and poor pregnancy outcomes, the generalizability of this knowledge to humans remains unclear. Here we identify uterus transplant (UTx) recipients as a human population with reduced endometrial NK cells and altered pregnancy phenotypes. We further show that the NK reduction in UTx is due to impaired transcriptional programming of NK tissue residency due to blockade of the transcription factor nuclear factor of activated T cells (NFAT). NFAT-dependent genes played a role in multiple molecular circuits governing tissue residency in uNKs, including early residency programs involving AP-1 transcription factors as well as TGFß-mediated upregulation of surface integrins. Collectively, our data identify a previously undescribed role for NFAT in uterine NK tissue residency and provide novel mechanistic insights into the biologic basis of pregnancy complications due to alteration of tissue resident NK subsets in humans. One Sentence Summary: Role of NFAT in uterine NK cell tissue residency.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA