Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Food Chem Toxicol ; 181: 114093, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37813178

ABSTRACT

A nationwide online survey assessed claimed usage of sunscreen products in 2283 self-identified regular sun protection factor (SPF) consumers (RSPFC) in the United States. Subjects applied sunscreen most frequently when spending more than 3 h in the sun. Sunscreen usage peaks during the summer, with sunny weather prompting 99% usage of beach/recreational SPF products but drops to approximately 50% and 30% on partly cloudy and cloudy days, respectively, regardless of SPF product category. About half of RSPFC augment sunscreen product usage by limiting time in the sun and wearing a hat. SPF products are not reapplied by approximately 20-60% of RSPFC, depending upon product category, and reapplication was less than 33% on cloudy and partly cloudy days. Primary reasons for reapplication were water exposure, number of hours in the sun, and being active/sweating, most notably for beach/recreational SPF products. Importantly, in children, 45% of parents reported "redness" as a signal for reapplying sunscreen product. Only 10% of respondents correctly identified sunscreen products as drugs. Based on these results, while sunscreens may share common ingredients and efficacy measures, their usage by consumers varies widely depending on product type, season, weather, gender, age, and geographical location.


Subject(s)
Sun Protection Factor , Sunscreening Agents , Child , Humans , United States , Sunlight , Erythema , Surveys and Questionnaires
2.
Mol Ther ; 31(3): 774-787, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36523164

ABSTRACT

Acute kidney injury occurs frequently in COVID-19 patients infected by the coronavirus SARS-CoV-2, and infection of kidney cells by this virus has been reported. However, little is known about the direct impact of the SARS-CoV-2 infection upon the renal tubular cells. We report that SARS-CoV-2 activated signal transducer and activator of transcription 3 (STAT3) signaling and caused cellular injury in the human renal tubular cell line. Mechanistically, the viral protein ORF3A of SARS-CoV-2 augmented both NF-κB and STAT3 signaling and increased the expression of kidney injury molecule 1. SARS-CoV-2 infection or expression of ORF3A alone elevated the protein level of tripartite motif-containing protein 59 (TRIM59), an E3 ubiquitin ligase, which interacts with both ORF3A and STAT3. The excessive TRIM59 in turn dissociated the phosphatase TCPTP from binding to STAT3 and hence inhibited the dephosphorylation of STAT3, leading to persistent STAT3 activation. Consistently, ORF3A induced renal injury in zebrafish and mice. In addition, expression of TRIM59 was elevated in the kidney autopsies of COVID-19 patients with acute kidney injury. Thus, the aberrant activation of STAT3 signaling by TRIM59 plays a significant role in the renal tubular cell injury caused by SARS-CoV-2, which suggests a potential targeted therapy for the renal complications of COVID-19.


Subject(s)
Acute Kidney Injury , COVID-19 , Humans , Animals , Mice , SARS-CoV-2 , COVID-19/metabolism , STAT3 Transcription Factor/metabolism , Zebrafish , Acute Kidney Injury/etiology , Viral Proteins/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
3.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34473647

ABSTRACT

Herein, we report that Shroom3 knockdown, via Fyn inhibition, induced albuminuria with foot process effacement (FPE) without focal segmental glomerulosclerosis (FSGS) or podocytopenia. Interestingly, knockdown mice had reduced podocyte volumes. Human minimal change disease (MCD), where podocyte Fyn inactivation was reported, also showed lower glomerular volumes than FSGS. We hypothesized that lower glomerular volume prevented the progression to podocytopenia. To test this hypothesis, we utilized unilateral and 5/6th nephrectomy models in Shroom3-KD mice. Knockdown mice exhibited less glomerular and podocyte hypertrophy after nephrectomy. FYN-knockdown podocytes had similar reductions in podocyte volume, implying that Fyn was downstream of Shroom3. Using SHROOM3 or FYN knockdown, we confirmed reduced podocyte protein content, along with significantly increased phosphorylated AMPK, a negative regulator of anabolism. AMPK activation resulted from increased cytoplasmic redistribution of LKB1 in podocytes. Inhibition of AMPK abolished the reduction in glomerular volume and induced podocytopenia in mice with FPE, suggesting a protective role for AMPK activation. In agreement with this, treatment of glomerular injury models with AMPK activators restricted glomerular volume, podocytopenia, and progression to FSGS. Glomerular transcriptomes from MCD biopsies also showed significant enrichment of Fyn inactivation and Ampk activation versus FSGS glomeruli. In summary, we demonstrated the important role of AMPK in glomerular volume regulation and podocyte survival. Our data suggest that AMPK activation adaptively regulates glomerular volume to prevent podocytopenia in the context of podocyte injury.


Subject(s)
Adenylate Kinase/metabolism , Kidney Glomerulus/metabolism , Microfilament Proteins/genetics , Nephrotic Syndrome/genetics , Podocytes/metabolism , Adenylate Kinase/antagonists & inhibitors , Adolescent , Adult , Aged , Albuminuria/genetics , Animals , Cell Size , Cell Survival/genetics , Child , Child, Preschool , Female , Gene Knockdown Techniques , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Hypertrophy , Infant , Kidney Glomerulus/pathology , Male , Mice , Middle Aged , Nephrectomy , Nephrosis, Lipoid/genetics , Nephrosis, Lipoid/pathology , Nephrotic Syndrome/pathology , Podocytes/pathology , Proportional Hazards Models , Proto-Oncogene Proteins c-fyn/genetics , Young Adult
4.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33998598

ABSTRACT

Genome-wide association studies (GWAS) for kidney function identified hundreds of risk regions; however, the causal variants, target genes, cell types, and disease mechanisms remain poorly understood. Here, we performed transcriptome-wide association studies (TWAS), summary Mendelian randomization, and MetaXcan to identify genes whose expression mediates the genotype effect on the phenotype. Our analyses identified Dachshund homolog 1 (DACH1), a cell-fate determination factor. GWAS risk variant was associated with lower DACH1 expression in human kidney tubules. Human and mouse kidney single-cell open chromatin data (snATAC-Seq) prioritized estimated glomerular filtration rate (eGFR) GWAS variants located on an intronic regulatory region in distal convoluted tubule cells. CRISPR-Cas9-mediated gene editing confirmed the role of risk variants in regulating DACH1 expression. Mice with tubule-specific Dach1 deletion developed more severe renal fibrosis both in folic acid and diabetic kidney injury models. Mice with tubule-specific Dach1 overexpression were protected from folic acid nephropathy. Single-cell RNA sequencing, chromatin immunoprecipitation, and functional analysis indicated that DACH1 controls the expression of cell cycle and myeloid chemotactic factors, contributing to macrophage infiltration and fibrosis development. In summary, integration of GWAS, TWAS, single-cell epigenome, expression analyses, gene editing, and functional validation in different mouse kidney disease models identified DACH1 as a kidney disease risk gene.


Subject(s)
Databases, Nucleic Acid , Eye Proteins , Kidney Diseases , Kidney Tubules/metabolism , Transcription Factors , Transcriptome , Animals , Disease Models, Animal , Eye Proteins/biosynthesis , Eye Proteins/genetics , Genome-Wide Association Study , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice , Mice, Transgenic , Risk Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics
5.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33998601

ABSTRACT

Dachshund homolog 1 (DACH1), a key cell-fate determinant, regulates transcription by DNA sequence-specific binding. We identified diminished Dach1 expression in a large-scale screen for mutations that convert injury-resistant podocytes into injury-susceptible podocytes. In diabetic kidney disease (DKD) patients, podocyte DACH1 expression levels are diminished, a condition that strongly correlates with poor clinical outcomes. Global Dach1 KO mice manifest renal hypoplasia and die perinatally. Podocyte-specific Dach1 KO mice, however, maintain normal glomerular architecture at baseline, but rapidly exhibit podocyte injury after diabetes onset. Furthermore, podocyte-specific augmentation of DACH1 expression in mice protects from DKD. Combined RNA sequencing and in silico promoter analysis reveal conversely overlapping glomerular transcriptomic signatures between podocyte-specific Dach1 and Pax transactivation-domain interacting protein (Ptip) KO mice, with upregulated genes possessing higher-than-expected numbers of promoter Dach1-binding sites. PTIP, an essential component of the activating histone H3 lysine 4 trimethylation (H3K4Me3) complex, interacts with DACH1 and is recruited by DACH1 to its promoter-binding sites. DACH1-PTIP recruitment represses transcription and reduces promoter H3K4Me3 levels. DACH1 knockdown in podocytes combined with hyperglycemia triggers target gene upregulation and increases promoter H3K4Me3. These findings reveal that in DKD, diminished DACH1 expression enhances podocyte injury vulnerability via epigenetic derepression of its target genes.


Subject(s)
DNA-Binding Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/prevention & control , Eye Proteins/biosynthesis , Histones/metabolism , Podocytes/metabolism , Animals , DNA-Binding Proteins/genetics , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Eye Proteins/genetics , Histones/genetics , Mice , Mice, Knockout , Podocytes/pathology
7.
medRxiv ; 2020 May 08.
Article in English | MEDLINE | ID: mdl-32511564

ABSTRACT

IMPORTANCE: Preliminary reports indicate that acute kidney injury (AKI) is common in coronavirus disease (COVID)-19 patients and is associated with worse outcomes. AKI in hospitalized COVID-19 patients in the United States is not well-described. OBJECTIVE: To provide information about frequency, outcomes and recovery associated with AKI and dialysis in hospitalized COVID-19 patients. DESIGN: Observational, retrospective study. SETTING: Admitted to hospital between February 27 and April 15, 2020. PARTICIPANTS: Patients aged ≥18 years with laboratory confirmed COVID-19 Exposures: AKI (peak serum creatinine increase of 0.3 mg/dL or 50% above baseline). Main Outcomes and Measures: Frequency of AKI and dialysis requirement, AKI recovery, and adjusted odds ratios (aOR) with mortality. We also trained and tested a machine learning model for predicting dialysis requirement with independent validation. RESULTS: A total of 3,235 hospitalized patients were diagnosed with COVID-19. AKI occurred in 1406 (46%) patients overall and 280 (20%) with AKI required renal replacement therapy. The incidence of AKI (admission plus new cases) in patients admitted to the intensive care unit was 68% (553 of 815). In the entire cohort, the proportion with stages 1, 2, and 3 AKI were 35%, 20%, 45%, respectively. In those needing intensive care, the respective proportions were 20%, 17%, 63%, and 34% received acute renal replacement therapy. Independent predictors of severe AKI were chronic kidney disease, systolic blood pressure, and potassium at baseline. In-hospital mortality in patients with AKI was 41% overall and 52% in intensive care. The aOR for mortality associated with AKI was 9.6 (95% CI 7.4-12.3) overall and 20.9 (95% CI 11.7-37.3) in patients receiving intensive care. 56% of patients with AKI who were discharged alive recovered kidney function back to baseline. The area under the curve (AUC) for the machine learned predictive model using baseline features for dialysis requirement was 0.79 in a validation test. CONCLUSIONS AND RELEVANCE: AKI is common in patients hospitalized with COVID-19, associated with worse mortality, and the majority of patients that survive do not recover kidney function. A machine-learned model using admission features had good performance for dialysis prediction and could be used for resource allocation.

8.
Kidney Int ; 96(3): 642-655, 2019 09.
Article in English | MEDLINE | ID: mdl-31171376

ABSTRACT

The essential role of membrane associated guanylate kinase 2 (MAGI2) in podocytes is indicated by the phenotypes of severe glomerulosclerosis of both MAGI2 knockout mice and in patients with congenital nephrotic syndrome (CNS) caused by mutations in MAGI2. Here, we show that MAGI2 forms a complex with the Rap1 guanine nucleotide exchange factor, RapGEF2, and that this complex is lost when expressing MAGI2 CNS variants. Co-expression of RapGEF2 with wild-type MAGI2, but not MAGI2 CNS variants, enhanced activation of the small GTPase Rap1, a central signaling node in podocytes. In mice, podocyte-specific RapGEF2 deletion resulted in spontaneous glomerulosclerosis, with qualitative glomerular features comparable to MAGI2 knockout mice. Knockdown of RapGEF2 or MAGI2 in human podocytes caused similar reductions in levels of Rap1 activation and Rap1-mediated downstream signaling. Furthermore, human podocytes expressing MAGI2 CNS variants show severe abnormalities of cellular morphology and dramatic loss of actin cytoskeletal organization, features completely rescued by pharmacological activation of Rap1 via a non-MAGI2 dependent upstream pathway. Finally, immunostaining of kidney sections from patients with congenital nephrotic syndrome and MAGI2 mutations showed reduced podocyte Rap1-mediated signaling. Thus, MAGI2-RapGEF2-Rap1 signaling is essential for normal podocyte function. Hence, disruption of this pathway is an important cause of the renal phenotype induced by MAGI2 CNS mutations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/genetics , Nephrotic Syndrome/genetics , Nerve Tissue Proteins/metabolism , Podocytes/pathology , Telomere-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Guanine Nucleotide Exchange Factors/genetics , Guanylate Kinases/metabolism , Humans , Mice , Mice, Knockout , Mutation , Nephrotic Syndrome/pathology , Shelterin Complex , Signal Transduction/drug effects , Signal Transduction/genetics , Telomere-Binding Proteins/agonists , rap1 GTP-Binding Proteins/metabolism
9.
J Am Soc Nephrol ; 29(11): 2641-2657, 2018 11.
Article in English | MEDLINE | ID: mdl-30341149

ABSTRACT

BACKGROUND: We previously showed that the presence of a CKD-associated locus in SHROOM3 in a donor kidney results in increased expression of SHROOM3 (an F-actin-binding protein important for epithelial morphogenesis, via rho-kinase [ROCK] binding); this facilitates TGF-b signaling and allograft fibrosis. However, other evidence suggests Shroom3 may have a protective role in glomerular development. METHODS: We used human data, Shroom3 knockdown podocytes, and inducible shRNA-mediated knockdown mice to study the role of Shroom3 in adult glomeruli. RESULTS: Expression data from the Nephroseq database showed glomerular and nonglomerular SHROOM3 had opposing associations with renal function in CKD biopsy samples. In human allografts, homozygosity at rs17319721, the SHROOM3 locus linked with lower GFR, was associated with reduced albuminuria by 2 years after transplant. Although our previous data showed reduced renal fibrosis with tubular Shroom3 knockdown, this study found that glomerular but not tubular Shroom3 knockdown induced albuminuria. Electron microscopy revealed diffuse foot process effacement, and glomerular RNA-sequencing showed enrichment of tyrosine kinase signaling and podocyte actin cytoskeleton pathways in knockdown mice. Screening SHROOM3-interacting proteins identified FYN (a src-kinase) as a candidate.We confirmed the interaction of endogenous SHROOM3 with FYN in human podocytes via a critical Src homology 3-binding domain, distinct from its ROCK-binding domain. Shroom3-Fyn interaction was required in vitro and in vivo for activation of Fyn kinase and downstream nephrin phosphorylation in podocytes. SHROOM3 knockdown altered podocyte morphology, cytoskeleton, adhesion, and migration. CONCLUSIONS: We demonstrate a novel mechanism that may explain SHROOM3's dichotomous associations in glomerular versus nonglomerular compartments in CKD.


Subject(s)
Albuminuria/metabolism , Kidney Transplantation , Kidney/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Actin Cytoskeleton/metabolism , Adolescent , Adult , Aged , Albuminuria/genetics , Albuminuria/pathology , Allografts , Animals , Child , Child, Preschool , Enhancer Elements, Genetic , Female , Gene Knockdown Techniques , Glomerular Filtration Rate/genetics , Homozygote , Humans , Kidney/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/chemistry , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Middle Aged , Phosphorylation , Podocytes/metabolism , Podocytes/pathology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-fyn/chemistry , RNA, Small Interfering/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/surgery , Signal Transduction , Young Adult , src Homology Domains
10.
J Biol Chem ; 292(51): 21137-21148, 2017 12 22.
Article in English | MEDLINE | ID: mdl-28982981

ABSTRACT

Kidney podocytes represent a key constituent of the glomerular filtration barrier. Identifying the molecular mechanisms of podocyte injury and survival is important for better understanding and management of kidney diseases. KIBRA (kidney brain protein), an upstream regulator of the Hippo signaling pathway encoded by the Wwc1 gene, shares the pro-injury properties of its putative binding partner dendrin and antagonizes the pro-survival signaling of the downstream Hippo pathway effector YAP (Yes-associated protein) in Drosophila and MCF10A cells. We recently identified YAP as an essential component of the glomerular filtration barrier that promotes podocyte survival by inhibiting dendrin pro-apoptotic function. Despite these recent advances, the signaling pathways that mediate podocyte injury remain poorly understood. Here we tested the hypothesis that, similar to its role in other model systems, KIBRA promotes podocyte injury. We found increased expression of KIBRA and phosphorylated YAP protein in glomeruli of patients with biopsy-proven focal segmental glomerulosclerosis (FSGS). KIBRA/WWc1 overexpression in murine podocytes promoted LATS kinase phosphorylation, leading to subsequent YAP Ser-127 phosphorylation, YAP cytoplasmic sequestration, and reduction in YAP target gene expression. Functionally, KIBRA overexpression induced significant morphological changes in podocytes, including disruption of the actin cytoskeletal architecture and reduction of focal adhesion size and number, all of which were rescued by subsequent YAP overexpression. Conversely, constitutive KIBRA knockout mice displayed reduced phosphorylated YAP and increased YAP expression at baseline. These mice were protected from acute podocyte foot process effacement following protamine sulfate perfusion. KIBRA knockdown podocytes were also protected against protamine-induced injury. These findings suggest an important role for KIBRA in the pathogenesis of podocyte injury and the progression of proteinuric kidney disease.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Biopsy , Female , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/pathology , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation , Podocytes/pathology , Podocytes/ultrastructure , Protein Processing, Post-Translational , RNA Interference , Serine/metabolism , Transcription Factors , YAP-Signaling Proteins
11.
J Biol Chem ; 291(47): 24406-24417, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27707879

ABSTRACT

MAGI-1 is a multidomain cytosolic scaffolding protein that in the kidney is specifically located at the podocyte slit diaphragm, a specialized junction that is universally injured in proteinuric diseases. There it interacts with several essential molecules, including nephrin and neph1, which are required for slit diaphragm formation and as an intracellular signaling hub. Here, we show that diminished MAGI-1 expression in cultured podocytes reduced nephrin and neph1 membrane localization and weakened tight junction integrity. Global magi1 knock-out mice, however, demonstrated normal glomerular histology and function into adulthood. We hypothesized that a second mild but complementary genetic insult might induce glomerular disease susceptibility in these mice. To identify such a gene, we utilized the developing fly eye to test for functional complementation between MAGI and its binding partners. In this way, we identified diminished expression of fly Hibris (nephrin) or Roughest (neph1) as dramatically exacerbating the effects of MAGI depletion. Indeed, when these combinations were studied in mice, the addition of nephrin, but not neph1, heterozygosity to homozygous deletion of MAGI-1 resulted in spontaneous glomerulosclerosis. In cultured podocytes, MAGI-1 depletion reduced intercellular contact-induced Rap1 activation, a pathway critical for proper podocyte function. Similarly, magi1 knock-out mice showed diminished glomerular Rap1 activation, an effect dramatically enhanced by concomitant nephrin haploinsufficiency. Finally, combined overexpression of MAGI-1 and nephrin increased Rap1 activation, but not when substituting a mutant MAGI-1 that cannot bind nephrin. We conclude that the interaction between nephrin and MAGI-1 regulates Rap1 activation in podocytes to maintain long term slit diaphragm structure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion Molecules , Enzyme Activation , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Guanylate Kinases , Membrane Proteins/genetics , Mice , Mice, Knockout , rap1 GTP-Binding Proteins/genetics
12.
J Clin Invest ; 124(4): 1757-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24642466

ABSTRACT

Injury to the specialized epithelial cells of the glomerulus (podocytes) underlies the pathogenesis of all forms of proteinuric kidney disease; however, the specific genetic changes that mediate podocyte dysfunction after injury are not fully understood. Here, we performed a large-scale insertional mutagenic screen of injury-resistant podocytes isolated from mice and found that increased expression of the gene Rap1gap, encoding a RAP1 activation inhibitor, ameliorated podocyte injury resistance. Furthermore, injured podocytes in murine models of disease and kidney biopsies from glomerulosclerosis patients exhibited increased RAP1GAP, resulting in diminished glomerular RAP1 activation. In mouse models, podocyte-specific inactivation of Rap1a and Rap1b induced massive glomerulosclerosis and premature death. Podocyte-specific Rap1a and Rap1b haploinsufficiency also resulted in severe podocyte damage, including features of podocyte detachment. Over-expression of RAP1GAP in cultured podocytes induced loss of activated ß1 integrin, which was similarly observed in kidney biopsies from patients. Furthermore, preventing elevation of RAP1GAP levels in injured podocytes maintained ß1 integrin-mediated adhesion and prevented cellular detachment. Taken together, our findings suggest that increased podocyte expression of RAP1GAP contributes directly to podocyte dysfunction by a mechanism that involves loss of RAP1-mediated activation of ß1 integrin.


Subject(s)
GTPase-Activating Proteins/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Podocytes/metabolism , Animals , GTPase-Activating Proteins/genetics , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Haploinsufficiency , Humans , Integrin beta1/metabolism , Kidney Glomerulus/injuries , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Mice, Transgenic , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , rap GTP-Binding Proteins/deficiency , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/deficiency , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 111(5): 2005-10, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449909

ABSTRACT

Many of the long-term effects of cocaine on the brain's reward circuitry have been shown to be mediated by alterations in gene expression. Several chromatin modifications, including histone acetylation and methylation, have been implicated in this regulation, but the effect of other histone modifications remains poorly understood. Poly(ADP-ribose) polymerase-1 (PARP-1), a ubiquitous and abundant nuclear protein, catalyzes the synthesis of a negatively charged polymer called poly(ADP-ribose) or PAR on histones and other substrate proteins and forms transcriptional regulatory complexes with several other chromatin proteins. Here, we identify an essential role for PARP-1 in cocaine-induced molecular, neural, and behavioral plasticity. Repeated cocaine administration, including self-administration, increased global levels of PARP-1 and its mark PAR in mouse nucleus accumbens (NAc), a key brain reward region. Using PARP-1 inhibitors and viral-mediated gene transfer, we established that PARP-1 induction in NAc mediates enhanced behavioral responses to cocaine, including increased self-administration of the drug. Using chromatin immunoprecipitation sequencing, we demonstrated a global, genome-wide enrichment of PARP-1 in NAc of cocaine-exposed mice and identified several PARP-1 target genes that could contribute to the lasting effects of cocaine. Specifically, we identified sidekick-1--important for synaptic connections during development--as a critical PARP-1 target gene involved in cocaine's behavioral effects as well as in its ability to induce dendritic spines on NAc neurons. These findings establish the involvement of PARP-1 and PARylation in the long-term actions of cocaine.


Subject(s)
Cocaine/pharmacology , Poly Adenosine Diphosphate Ribose/metabolism , Animals , Behavior, Animal/drug effects , Chromatin Immunoprecipitation , Cocaine/administration & dosage , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Genome/genetics , Immunoglobulin G/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Substrate Specificity/drug effects , Transcription, Genetic/drug effects
14.
Kidney Int ; 84(5): 950-61, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23842188

ABSTRACT

Activation of signal transducer and activator of transcription (STAT)3 correlates with proliferation of extracapillary glomerular epithelial cells and the extent of renal injury in glomerulonephritis. To delineate the role of STAT3 in glomerular epithelial cell proliferation, we examined the development of nephrotoxic serum-induced glomerulonephritis in mice with and without podocyte-restricted STAT3 deletion. Mice with STAT3 deletion in podocytes developed less crescents and loss of renal function compared with those without STAT3 deletion. Proliferation of glomerular cells, loss of podocyte markers, and recruitment of parietal epithelial cells were found in nephritic mice without STAT3 deletion, but mitigated in nephritic mice with podocyte STAT3 deletion. Glomerular expression of pro-inflammatory STAT3 target genes was significantly reduced in nephritic mice with, compared with those without, podocyte STAT3 deletion. However, the extent of glomerular immune complex deposition was not different. Podocytes with STAT3 deletion were resistant to interleukin-6-induced STAT3 phosphorylation and pro-inflammatory STAT3 target gene expression. Thus, podocyte STAT3 activation is critical for the development of crescentic glomerulonephritis.


Subject(s)
Glomerulonephritis/prevention & control , Podocytes/metabolism , STAT3 Transcription Factor/deficiency , Serum , Albuminuria/metabolism , Animals , Antigen-Antibody Complex/metabolism , Apoptosis , Cell Line , Cell Proliferation , Complement C3/metabolism , Disease Models, Animal , Epidermal Growth Factor/metabolism , Glomerulonephritis/chemically induced , Glomerulonephritis/genetics , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Immunoglobulins/metabolism , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Podocytes/pathology , Primary Cell Culture , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors
15.
AIDS ; 27(7): 1091-8, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23343908

ABSTRACT

OBJECTIVE: HIV-1 gene expression in kidney epithelial cells is thought to be responsible for the pathogenesis of HIV-1-associated nephropathy (HIVAN). Signal transducer and activator of transcription (STAT) 3 signaling is activated in podocytes of patients with HIVAN and drives the dedifferentiation and proliferation of podocytes in culture. We confirm here that deletion of podocyte STAT3 is sufficient to mitigate the glomerular as well as tubulointerstitial findings of HIVAN. METHODS: To demonstrate the functional role of podocyte STAT3 in the pathogenesis of HIVAN we compared the development of HIVAN in Tg26 HIV-transgenic mice with and without deletion of STAT3 in the podocyte. RESULTS: Tg26 mice with podocyte-specific STAT3 deletion developed significantly less weight loss, albuminuria, and renal function impairment compared to Tg26 mice without STAT3 deletion. Tg26 mice with podocyte STAT3 deletion also had significantly less glomerular collapse, sclerosis, epithelial cell hyperplasia, podocyte dedifferentiation, and proinflammatory STAT3 target gene expression; and tubulointerstitial changes of HIVAN, including tubular atrophy, degeneration, apoptosis, and lymphocyte infiltration, were also significantly reduced compared to Tg26 mice without STAT3 deletion. CONCLUSION: Development of glomerular as well as tubulointerstitial injuries in the Tg26 HIVAN model is dependent on podocyte STAT3 expression. Inhibition of STAT3 could be a potential adjunctive therapy for the treatment of HIVAN.


Subject(s)
AIDS-Associated Nephropathy/pathology , HIV Infections/pathology , Kidney Glomerulus/pathology , Podocytes/metabolism , STAT3 Transcription Factor/metabolism , AIDS-Associated Nephropathy/etiology , AIDS-Associated Nephropathy/genetics , Albuminuria , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Epithelial Cells/pathology , Gene Deletion , Gene Expression Profiling , HIV Infections/complications , HIV Infections/genetics , Humans , Kidney Glomerulus/metabolism , Mice , Mice, Transgenic , MicroRNAs , Podocytes/pathology , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Sclerosis , Signal Transduction/genetics , Weight Loss
16.
Kidney Int ; 83(4): 626-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23325078

ABSTRACT

With the widespread use of combination antiretroviral agents, the incidence of HIV-associated nephropathy has decreased. Currently, HIV-infected patients live much longer and often suffer from comorbidities such as diabetes mellitus. Recent epidemiological studies suggest that concurrent HIV infection and diabetes mellitus may have a synergistic effect on the incidence of chronic kidney disease. To address this, we determined whether HIV-1 transgene expression accelerates diabetic kidney injury using a diabetic HIV-1 transgenic (Tg26) murine model. Diabetes was initially induced with low-dose streptozotocin in both Tg26 and wild-type mice on a C57BL/6 background, which is resistant to classic HIV-associated nephropathy. Although diabetic nephropathy is minimally observed on the C57BL/6 background, diabetic Tg26 mice exhibited a significant increase in glomerular injury compared with nondiabetic Tg26 mice and diabetic wild-type mice. Validation of microarray gene expression analysis from isolated glomeruli showed a significant upregulation of proinflammatory pathways in diabetic Tg26 mice. Thus, our study found that expression of HIV-1 genes aggravates diabetic kidney disease.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , HIV Infections/complications , HIV-1/genetics , Kidney/virology , Albuminuria/etiology , Albuminuria/genetics , Albuminuria/virology , Animals , Biomarkers/urine , Collagen Type IV/metabolism , Creatinine/urine , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Diabetic Nephropathies/virology , Disease Progression , Fibrosis , Fusion Proteins, gag-pol/genetics , Gene Expression Profiling/methods , HIV Infections/blood , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , Inflammation Mediators/blood , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Phosphorylation , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Smad3 Protein/metabolism , Time Factors
17.
Toxicol In Vitro ; 25(3): 708-14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21172418

ABSTRACT

Personal care product manufacturers have used a broad spectrum of alternative ocular irritation assays during the past two decades because these tests do not require the use of live animals, they provide reliable predictive data, and they are relatively inexpensive to conduct. To complement these assays, the ex vivo Porcine Corneal Opacity Reversibility Assay (PorCORA) was recently developed using a corneal culture model to predict reversibility of ocular irritants. Three commercially available consumer products (a shampoo, a hair color glaze, and a hair colorant system containing 12% hydrogen peroxide) were each tested in two PorCORA study replicates in order to assess potential ocular damage reversibility for surfactant-, propylene carbonate-, and peroxide-based formulations, respectively. Under the exaggerated, in vitro study conditions, the surfactant-based shampoo may cause irreversible porcine corneal damage (histological changes in the epithelial squamous cell and/or basal cell layers), whereas the hair color glaze and 12% hydrogen peroxide product caused fully reversible ocular irritation (microscopic changes only in the superficial squamous cell layer). The hair color glaze and peroxide product results correlate with established in vivo data for similar compounds, but the shampoo results contradicted previous BCOP results (expected to be only a mild irritant). Therefore, although the PorCORA protocol shows promise in predicting the extent and reversibility of potential ocular damage caused by accidental consumer eye exposure to personal care products, the contradictory results for the surfactant-based shampoo indicate that more extensive validation testing of the PorCORA is necessary to definitively establish the protocol's reliability as a Draize test replacement.


Subject(s)
Consumer Product Safety , Corneal Diseases/chemically induced , Cosmetics/toxicity , Epithelium, Corneal/drug effects , Irritants/toxicity , Animal Testing Alternatives , Animals , Corneal Diseases/pathology , Cosmetics/classification , Endpoint Determination , Epithelium, Corneal/pathology , Irritants/classification , Necrosis , Organ Culture Techniques , Recovery of Function , Swine
18.
Kidney Int ; 79(6): 624-634, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21150871

ABSTRACT

All-trans retinoic acid protects against the development of HIV-associated nephropathy (HIVAN) in HIV-1 transgenic mice (Tg26). In vitro, all-trans retinoic acid inhibits HIV-induced podocyte proliferation and restores podocyte differentiation markers by activating its receptor-α (RARα). Here, we report that Am580, a water-soluble RARα-specific agonist, attenuated proteinuria, glomerosclerosis, and podocyte proliferation, and restored podocyte differentiation markers in kidneys of Tg26 mice. Furthermore, RARα-/- Tg26 mice developed more severe kidney and podocyte injury than did RARα+/- Tg26 mice. Am580 failed to ameliorate kidney injury in RARα-/- Tg26 mice, confirming our hypothesis that Am580 acts through RARα. Although the expression of RARα-target genes was suppressed in the kidneys of Tg26 mice and of patients with HIVAN, the expression of RARα in the kidney was not different between patients with HIVAN and minimal change disease. However, the tissue levels of retinoic acid were reduced in the kidney cortex and isolated glomeruli of Tg26 mice. Consistent with this, the expression of two key enzymes in the retinoic acid synthetic pathway, retinol dehydrogenase type 1 and 9, and the overall enzymatic activity for retinoic acid synthesis were significantly reduced in the glomeruli of Tg26 mice. Thus, a defect in the endogenous synthesis of retinoic acid contributes to loss of the protection by retinoic acid in HIVAN. Hence, RARα agonists may be potential agents for the treatment of HIVAN.


Subject(s)
AIDS-Associated Nephropathy/metabolism , HIV-1/genetics , Podocytes/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction , AIDS-Associated Nephropathy/genetics , AIDS-Associated Nephropathy/pathology , AIDS-Associated Nephropathy/prevention & control , AIDS-Associated Nephropathy/virology , Alcohol Oxidoreductases/metabolism , Animals , Benzoates/pharmacology , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Female , Glomerulonephritis/metabolism , Glomerulonephritis/prevention & control , Glomerulonephritis/virology , Humans , Hydroxysteroid Dehydrogenases/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Podocytes/drug effects , Podocytes/pathology , Podocytes/virology , Proteinuria/metabolism , Proteinuria/prevention & control , Proteinuria/virology , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Retinoids/metabolism , Severity of Illness Index , Signal Transduction/drug effects , Tetrahydronaphthalenes/pharmacology , Time Factors
19.
J Biol Chem ; 285(33): 25677-85, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20562105

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a leading cause of nephrotic syndrome and end-stage renal disease worldwide. Although the mechanisms underlying this important disease are poorly understood, the glomerular podocyte clearly plays a central role in disease pathogenesis. In the current work, we demonstrate that the homophilic adhesion molecule sidekick-1 (sdk-1) is up-regulated in podocytes in FSGS both in rodent models and in human kidney biopsy samples. Transgenic mice that have podocyte-specific overexpression of sdk-1 develop gradually progressive heavy proteinuria and severe FSGS. We also show that sdk-1 associates with the slit diaphragm linker protein MAGI-1, which is already known to interact with several critical podocyte proteins including synaptopodin, alpha-actinin-4, nephrin, JAM4, and beta-catenin. This interaction is mediated through a direct interaction between the carboxyl terminus of sdk-1 and specific PDZ domains of MAGI-1. In vitro expression of sdk-1 enables a dramatic recruitment of MAGI-1 to the cell membrane. Furthermore, a truncated version of sdk-1 that is unable to bind to MAGI-1 does not induce podocyte dysfunction when overexpressed. We conclude that the up-regulation of sdk-1 in podocytes is an important pathogenic factor in FSGS and that the mechanism involves disruption of the actin cytoskeleton possibly via alterations in MAGI-1 function.


Subject(s)
Cell Adhesion Molecules/metabolism , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Immunoglobulin G/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Glomerulosclerosis, Focal Segmental/genetics , Guanylate Kinases , Humans , Immunoglobulin G/genetics , Immunoprecipitation , In Vitro Techniques , Membrane Proteins/genetics , Mice , Mice, Transgenic , Polymerase Chain Reaction , Protein Binding
20.
Adv Chronic Kidney Dis ; 17(1): 36-43, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20005487

ABSTRACT

HIV-associated nephropathy (HIVAN) is one of the leading causes of ESRD in HIV-1-seropositive patients. Patients typically present with heavy proteinuria and chronic renal failure with pathologic findings of collapsing focal segmental glomerulosclerosis (FSGS). The disease is caused by direct infection of renal epithelial cells by HIV-1 in a genetically susceptible host. The genetic factors responsible for the susceptibility to HIVAN among blacks include a noncoding variant in the podocyte-expressed gene nonmuscle myosin, heavy chain 9 (MYH9) as well as other genes yet to be identified. Podocyte and tubular dysfunction results from the expression of viral genes, in particular nef and vpr, and the subsequent dysregulation of numerous host factors, including critical signaling pathways, inflammatory mediators, and others. The identification of these factors has the potential to provide novel therapeutic targets to prevent and treat this important disease.


Subject(s)
AIDS-Associated Nephropathy/etiology , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/virology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/virology , AIDS-Associated Nephropathy/pathology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Failure, Chronic/pathology , Podocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...