Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Pharm ; 20(2): 987-996, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36626167

ABSTRACT

Despite the understanding that renal clearance is pivotal for driving the pharmacokinetics of numerous therapeutic proteins and peptides, the specific processes that occur following glomerular filtration remain poorly defined. For instance, sites of catabolism within the proximal tubule can occur at the brush border, within lysosomes following endocytosis, or even within the tubule lumen itself. The objective of the current study was to address these limitations and develop methodology to study the kidney disposition of a model therapeutic protein. Exenatide is a peptide used to treat type 2 diabetes mellitus. Glomerular filtration and ensuing renal catabolism have been shown to be its principal clearance pathway. Here, we designed and validated a Förster resonance energy transfer-quenched exenatide derivative to provide critical information on the renal handling of exenatide. A combination of in vitro techniques was used to confirm substantial fluorescence quenching of intact peptide that was released upon proteolytic cleavage. This evaluation was then followed by an assessment of the in vivo disposition of quenched exenatide directly within kidneys of living rats via intravital two-photon microscopy. Live imaging demonstrated rapid glomerular filtration and identified exenatide metabolism occurred within the subapical regions of the proximal tubule epithelia, with subsequent intracellular trafficking of cleaved fragments. These results provide a novel examination into the real-time, intravital disposition of a protein therapeutic within the kidney and offer a platform to build upon for future work.


Subject(s)
Diabetes Mellitus, Type 2 , Exenatide , Kidney , Animals , Rats , Diabetes Mellitus, Type 2/metabolism , Exenatide/metabolism , Exenatide/pharmacokinetics , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Peptides/metabolism
2.
J Am Soc Nephrol ; 32(11): 2795-2813, 2021 11.
Article in English | MEDLINE | ID: mdl-34479966

ABSTRACT

BACKGROUND: Podocyte depletion precedes progressive glomerular damage in several kidney diseases. However, the current standard of visual detection and quantification of podocyte nuclei from brightfield microscopy images is laborious and imprecise. METHODS: We have developed PodoSighter, an online cloud-based tool, to automatically identify and quantify podocyte nuclei from giga-pixel brightfield whole-slide images (WSIs) using deep learning. Ground-truth to train the tool used immunohistochemically or immunofluorescence-labeled images from a multi-institutional cohort of 122 histologic sections from mouse, rat, and human kidneys. To demonstrate the generalizability of our tool in investigating podocyte loss in clinically relevant samples, we tested it in rodent models of glomerular diseases, including diabetic kidney disease, crescentic GN, and dose-dependent direct podocyte toxicity and depletion, and in human biopsies from steroid-resistant nephrotic syndrome and from human autopsy tissues. RESULTS: The optimal model yielded high sensitivity/specificity of 0.80/0.80, 0.81/0.86, and 0.80/0.91, in mouse, rat, and human images, respectively, from periodic acid-Schiff-stained WSIs. Furthermore, the podocyte nuclear morphometrics extracted using PodoSighter were informative in identifying diseased glomeruli. We have made PodoSighter freely available to the general public as turnkey plugins in a cloud-based web application for end users. CONCLUSIONS: Our study demonstrates an automated computational approach to detect and quantify podocyte nuclei in standard histologically stained WSIs, facilitating podocyte research, and enabling possible future clinical applications.


Subject(s)
Cloud Computing , Image Processing, Computer-Assisted/methods , Kidney Diseases/pathology , Kidney Glomerulus/cytology , Podocytes/ultrastructure , Animals , Automation , Cell Count , Cell Nucleus/ultrastructure , Datasets as Topic , Deep Learning , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microscopy , Periodic Acid-Schiff Reaction , Rats , Species Specificity
3.
Biochem Biophys Res Commun ; 529(3): 740-746, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32736701

ABSTRACT

Endocytosis by podocytes is gaining increased attention as a biologic means of removing large proteins such as serum albumin from the glomerular barrier. Some of this function has been attributed to the megalin/cubilin (Lrp2/Cubn) receptor complex and the albumin recycling protein FcRn (Fcgrt). However, whether other glomerular cells possess the potential to perform this same phenomenon or express these proteins remains uncharacterized. Mesangial cells are uniquely positioned in glomeruli and represent a cell type capable of performing several diverse functions. Here, the expression of megalin and FcRn in murine mesangial cells along with the megalin adaptor protein Dab-2 (Dab2) was shown for the first time. Cubilin mRNA expression was detected, but the absence of the cubilin partner amnionless (Amn) suggested that cubilin is minimally functional, if at all, in these cells. Mesangial cell endocytosis of albumin was characterized and shown to involve a receptor-mediated process. Albumin endocytosis was significantly impaired (p < 0.01) under inducible megalin knockdown conditions in stably transduced mesangial cells. The current work provides both the novel identification of megalin and FcRn in mesangial cells and the functional demonstration of megalin-mediated albumin endocytosis.


Subject(s)
Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Mesangial Cells/cytology , Serum Albumin, Bovine/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cattle , Cell Line , Histocompatibility Antigens Class I/metabolism , Mesangial Cells/metabolism , Mice , Receptors, Fc/metabolism
4.
J Am Soc Nephrol ; 30(10): 1953-1967, 2019 10.
Article in English | MEDLINE | ID: mdl-31488606

ABSTRACT

BACKGROUND: Pathologists use visual classification of glomerular lesions to assess samples from patients with diabetic nephropathy (DN). The results may vary among pathologists. Digital algorithms may reduce this variability and provide more consistent image structure interpretation. METHODS: We developed a digital pipeline to classify renal biopsies from patients with DN. We combined traditional image analysis with modern machine learning to efficiently capture important structures, minimize manual effort and supervision, and enforce biologic prior information onto our model. To computationally quantify glomerular structure despite its complexity, we simplified it to three components consisting of nuclei, capillary lumina and Bowman spaces; and Periodic Acid-Schiff positive structures. We detected glomerular boundaries and nuclei from whole slide images using convolutional neural networks, and the remaining glomerular structures using an unsupervised technique developed expressly for this purpose. We defined a set of digital features which quantify the structural progression of DN, and a recurrent network architecture which processes these features into a classification. RESULTS: Our digital classification agreed with a senior pathologist whose classifications were used as ground truth with moderate Cohen's kappa κ = 0.55 and 95% confidence interval [0.50, 0.60]. Two other renal pathologists agreed with the digital classification with κ1 = 0.68, 95% interval [0.50, 0.86] and κ2 = 0.48, 95% interval [0.32, 0.64]. Our results suggest computational approaches are comparable to human visual classification methods, and can offer improved precision in clinical decision workflows. We detected glomerular boundaries from whole slide images with 0.93±0.04 balanced accuracy, glomerular nuclei with 0.94 sensitivity and 0.93 specificity, and glomerular structural components with 0.95 sensitivity and 0.99 specificity. CONCLUSIONS: Computationally derived, histologic image features hold significant diagnostic information that may augment clinical diagnostics.


Subject(s)
Diabetic Nephropathies/classification , Diabetic Nephropathies/pathology , Diagnosis, Computer-Assisted , Kidney Glomerulus/pathology , Humans
5.
Am J Physiol Renal Physiol ; 315(5): F1191-F1207, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29949391

ABSTRACT

The megalin/cubilin complex is responsible for the majority of serum protein reclamation in the proximal tubules. The current study examined if decreases in their renal expression, along with the albumin recycling protein neonatal Fc receptor (FcRn) could account for proteinuria/albuminuria in the Zucker diabetic fatty rat model of type 2 diabetes. Immunoblots of renal cortex samples obtained at worsening disease stages demonstrated no loss in megalin, cubilin, or FcRn, even when proteinuria was measured. Additionally, early diabetic rats exhibited significantly increased renal megalin expression when compared with controls (adjusted P < 0.01). Based on these results, the ability of insulin to increase megalin was examined in a clonal subpopulation of the opossum kidney proximal tubule cell line. Insulin treatments (24 h, 100 nM) under high glucose conditions significantly increased megalin protein ( P < 0.0001), mRNA ( P < 0.0001), and albumin endocytosis. The effect on megalin expression was prevented with inhibitors against key effectors of insulin intracellular signaling, phosphatidylinositide 3-kinase and Akt. Studies using rapamycin to inhibit the mechanistic target of rapamycin complex 1 (mTORC1) resulted in a loss of insulin-induced megalin expression. However, subsequent evaluation demonstrated these effects were independent of initial mTORC1 suppression. The presented results provide insight into the expression of megalin, cubilin, and FcRn in type 2 diabetes, which may be impacted by elevated insulin and glucose. Furthermore, proximal tubule endocytic activity in early diabetics may be enhanced, a process that could have a significant role in proteinuria-induced renal damage.


Subject(s)
Albuminuria/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/metabolism , Insulin/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Albuminuria/etiology , Albuminuria/genetics , Albuminuria/physiopathology , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Disease Progression , Endocytosis/drug effects , Glycogen Synthase Kinase 3/metabolism , Histocompatibility Antigens Class I/metabolism , Kidney Tubules, Proximal/physiopathology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Opossums , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Zucker , Receptors, Cell Surface/metabolism , Receptors, Fc/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
6.
Am J Physiol Renal Physiol ; 315(3): F487-F502, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29693447

ABSTRACT

Several lines of evidence suggest that gut bacterial microbiota is altered in patients with chronic kidney disease (CKD), though the mechanism of which this dysbiosis takes place is not well understood. Recent studies delineated changes in gut microbiota in both CKD patients and experimental animal models using microarray chips. We present 16S ribosomal RNA gene sequencing of both stool pellets and small bowel contents of C57BL/6J mice that underwent a remnant kidney model and establish that changes in microbiota take place in the early gastrointestinal tract. Increased intestinal urea concentration has been hypothesized as a leading contributor to dysbiotic changes in CKD. We show that urea transporters (UT)-A and UT-B mRNA are both expressed throughout the whole gastrointestinal tract. The noted increase in intestinal urea concentration appears to be independent of UTs' expression. Urea supplementation in drinking water resulted in alteration in bacterial gut microbiota that is quite different than that seen in CKD. This indicates that increased intestinal urea concentration might not fully explain the CKD- associated dysbiosis.


Subject(s)
Bacteria/metabolism , Dysbiosis , Gastrointestinal Microbiome , Intestine, Small/microbiology , Renal Insufficiency, Chronic/microbiology , Urea/metabolism , Uremia/microbiology , Administration, Oral , Animals , Bacteria/classification , Bacteria/genetics , Disease Models, Animal , Feces/microbiology , Host-Pathogen Interactions , Hydrolysis , Intestine, Small/metabolism , Male , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Renal Insufficiency, Chronic/metabolism , Ribotyping , Urea/administration & dosage , Urease/metabolism , Uremia/metabolism
7.
Kidney Int ; 93(2): 482-490, 2018 02.
Article in English | MEDLINE | ID: mdl-28965746

ABSTRACT

HLA matching and mismatching, while inversely related, are not exact opposites. Here we determined the independent effects of HLA matching and mismatching on outcomes in deceased donor kidney transplant recipients. The United Network for Organ Sharing database (1995-2012) was utilized and analyzed for delayed graft function, one-year acute rejection, and death-censored graft survival using combined multivariable models including HLA matching and mismatching. Sensitivity analyses were performed using the subgroup of deceased donor kidney transplant patients after 2003 with more uniform HLA nomenclature and resampling analyses using bootstrapping on complete data available from 96,236 recipients. Individually, both HLA matching and mismatching showed significant associations with graft survival. Adjusting the model to take into account both matching and mismatching simultaneously, the degree of HLA mismatching lost significance while matching continued to have a significant prediction for delayed graft function, the one-year acute rejection rate, and graft survival. Sensitivity analyses and bootstrapping showed similar results for all studied outcomes. Thus, analysis of this large cohort demonstrates the apparent greater association of HLA matching over HLA mismatching on both early allograft events as well as graft survival. Future analyses should preferentially utilize HLA matching as a covariate over mismatching for accurately reflecting impact on graft outcomes.


Subject(s)
Delayed Graft Function/immunology , Graft Rejection/immunology , Graft Survival , HLA Antigens/immunology , Histocompatibility Testing , Histocompatibility , Kidney Transplantation , Tissue and Organ Procurement , Delayed Graft Function/mortality , Delayed Graft Function/prevention & control , Graft Rejection/mortality , Graft Rejection/prevention & control , Humans , Kidney Transplantation/adverse effects , Kidney Transplantation/mortality , Predictive Value of Tests , Protective Factors , Registries , Retrospective Studies , Risk Factors , Time Factors , Treatment Outcome , United States/epidemiology
8.
J Am Soc Nephrol ; 28(1): 166-184, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27288011

ABSTRACT

Podocyte injury is the inciting event in primary glomerulopathies, such as minimal change disease and primary FSGS, and glucocorticoids remain the initial and often, the primary treatment of choice for these glomerulopathies. Because inflammation is not readily apparent in these diseases, understanding the direct effects of glucocorticoids on the podocyte, independent of the immunomodulatory effects, may lead to the identification of targets downstream of glucocorticoids that minimize toxicity without compromising efficacy. Several studies showed that treatment with glucocorticoids restores podocyte differentiation markers and normal ultrastructure and improves cell survival in murine podocytes. We previously determined that Krüppel-like factor 15 (KLF15), a kidney-enriched zinc finger transcription factor, is required for restoring podocyte differentiation markers in mice and human podocytes under cell stress. Here, we show that in vitro treatment with dexamethasone induced a rapid increase of KLF15 expression in human and murine podocytes and enhanced the affinity of glucocorticoid receptor binding to the promoter region of KLF15 In three independent proteinuric murine models, podocyte-specific loss of Klf15 abrogated dexamethasone-induced podocyte recovery. Furthermore, knockdown of KLF15 reduced cell survival and destabilized the actin cytoskeleton in differentiated human podocytes. Conversely, overexpression of KLF15 stabilized the actin cytoskeleton under cell stress in human podocytes. Finally, the level of KLF15 expression in the podocytes and glomeruli from human biopsy specimens correlated with glucocorticoid responsiveness in 35 patients with minimal change disease or primary FSGS. Thus, these studies identify the critical role of KLF15 in mediating the salutary effects of glucocorticoids in the podocyte.


Subject(s)
Cell Differentiation/drug effects , DNA-Binding Proteins/physiology , Glucocorticoids/pharmacology , Podocytes/cytology , Podocytes/drug effects , Transcription Factors/physiology , Adolescent , Adult , Animals , Antigens, Differentiation/drug effects , Child , Dexamethasone/pharmacology , Female , Glomerulosclerosis, Focal Segmental/immunology , Humans , Kruppel-Like Transcription Factors , Male , Mice , Middle Aged , Nephrosis, Lipoid/immunology , Young Adult
9.
Am J Physiol Renal Physiol ; 313(3): F621-F628, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28615249

ABSTRACT

Both the incidence and prevalence of chronic kidney disease are increasing in the elderly population. Although aging is known to induce kidney injury, the underlying molecular mechanisms remain unclear. Sirtuin 1 (Sirt1), a longevity gene, is known to protect kidney cell injury from various cellular stresses. In previous studies, we showed that the podocyte-specific loss of Sirt1 aggravates diabetic kidney injury. However, the role of Sirt1 in aging-induced podocyte injury is not known. Therefore, in this study we sought to determine the effects of podocyte-specific reduction of Sirt1 in age-induced kidney injury. We employed the inducible podocyte-specific Sirt1 knockdown mice that express shRNA against Sirt1 (Pod-Sirt1RNAi) and control mice that express shRNA for luciferase (Pod-LuciRNAi). We found that reduction of podocyte Sirt1 led to aggravated aging-induced glomerulosclerosis and albuminuria. In addition, urinary level of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a marker of oxidative stress, was markedly increased in aged Pod-Sirt1RNAi mice compared with aged Pod-LuciRNAi mice. Although podocyte-specific markers decreased in aged mice compared with the young controls, the decrease was further exacerbated in aged Pod-Sirt1RNAi compared with Pod-LuciRNAi mice. Interestingly, expression of cellular senescence markers was significantly higher in the glomeruli of Pod-Sirt1RNAi mice than Pod-LuciRNAi mice, suggesting that cellular senescence may contribute to podocyte loss in aging kidneys. Finally, we confirmed that Pod-Sirt1RNAi glomeruli were associated with reduced activation of the transcription factors peroxisome proliferator-activated receptor (PPAR)-α coactivador-1 (PGC1α)/PPARγ, forkhead box O (FOXO)3, FOXO4, and p65 NF-κB, through SIRT1-mediated deacetylation. Together, our data suggest that SIRT1 may be a potential therapeutic target to treat patients with aging-related kidney disease.


Subject(s)
Aging/metabolism , Albuminuria/enzymology , Podocytes/enzymology , Renal Insufficiency, Chronic/enzymology , Sirtuin 1/deficiency , 8-Hydroxy-2'-Deoxyguanosine , Acetylation , Age Factors , Aging/genetics , Aging/pathology , Albuminuria/genetics , Albuminuria/pathology , Animals , Cell Cycle Proteins , Cellular Senescence , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/urine , Forkhead Box Protein O3/metabolism , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Genotype , Glomerulonephritis/enzymology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Mice , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phenotype , Podocytes/pathology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Signal Transduction , Sirtuin 1/genetics , Transcription Factor RelA/metabolism
10.
Kidney Int ; 91(3): 521-523, 2017 03.
Article in English | MEDLINE | ID: mdl-28202164

ABSTRACT

The uremic solute indoxyl sulfate has been associated with increased mortality and other adverse outcomes in patients with chronic kidney disease. In a recent study published in Cell Host & Microbe, Devlin et al. describe a novel approach to alter the production of indoxyl sulfate through manipulation of the gut microbiota. Although this approach is far from clinical application, it may allow investigators to determine the contribution of uremic solutes to disease pathogenesis.


Subject(s)
Bacteroides/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Indican/metabolism , Uremia/metabolism , Animals , Bacteroides/genetics , Genotype , Humans , Indican/toxicity , Mutation , Phenotype , Uremia/microbiology , Uremia/mortality
11.
Nutr J ; 15: 10, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818246

ABSTRACT

BACKGROUND: Data from experimental animals suggest that probiotic supplements may retard CKD progression. However, the relationship between probiotic use, frequent yogurt consumption (as a natural probiotic source), and kidney parameters have not been evaluated in humans. FINDINGS: We utilized NHANES data, and analyzed the association of probiotic alone (1999-2012) and yogurt/probiotic (2003-2006) use with albuminuria and eGFR after adjustment for demographic and clinical parameters. Frequent yogurt consumption was defined as thrice or more weekly over the year prior to the interview. Frequent yogurt/probiotic consumers had lower adjusted odds of developing combined outcome (albuminuria and/or eGFR < 60 ml/min/1.73 m(2)) compared to infrequent consumers (OR = 0.76; 95 % CI = 0.61-0.94). When evaluated separately, frequent consumers had lower odds of albuminuria and nonsignificant trend towards decreased odds of low eGFR compared to infrequent consumers. In the probiotic cohort, probiotic consumers were found to have a lower adjusted odds of albuminuria compared to nonusers (OR = 0.59; 95 % CI = 0.37-0.94). CONCLUSION: Frequent yogurt and/or probiotics use is associated with decreased odds of proteinuric kidney disease. These hypothesis-generating results warrant further translational studies to further delineate the relationship between yogurt/probiotics with kidney dysfunction, as well as microbiome and dysbiosis as potential mediators.


Subject(s)
Kidney Diseases/epidemiology , Nutrition Surveys , Probiotics/administration & dosage , Yogurt , Adult , Cross-Sectional Studies , Female , Glomerular Filtration Rate , Humans , Logistic Models , Male , Multivariate Analysis , Sample Size , Socioeconomic Factors
12.
J Am Soc Nephrol ; 26(10): 2361-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25644109

ABSTRACT

Nephrin is required during kidney development for the maturation of podocytes and formation of the slit diaphragm junctional complex. Because nephrin expression is downregulated in acquired glomerular diseases, nephrin deficiency is considered a pathologic feature of glomerular injury. However, whether nephrin deficiency exacerbates glomerular injury in glomerular diseases has not been experimentally confirmed. Here, we generated mice with inducible RNA interference-mediated nephrin knockdown. Short-term nephrin knockdown (6 weeks), starting after the completion of kidney development at 5 weeks of age, did not affect glomerular structure or function. In contrast, mice with long-term nephrin knockdown (20 weeks) developed mild proteinuria, foot process effacement, filtration slit narrowing, mesangial hypercellularity and sclerosis, glomerular basement membrane thickening, subendothelial zone widening, and podocyte apoptosis. When subjected to an acquired glomerular insult induced by unilateral nephrectomy or doxorubicin, mice with short-term nephrin knockdown developed more severe glomerular injury compared with mice without nephrin knockdown. Additionally, nephrin-knockdown mice developed more exaggerated glomerular enlargement when subjected to unilateral nephrectomy and more podocyte apoptosis and depletion after doxorubicin challenge. AKT phosphorylation, which is a slit diaphragm-mediated and nephrin-dependent pathway in the podocyte, was markedly reduced in mice with long-term or short-term nephrin knockdown challenged with uninephrectomy or doxorubicin. Taken together, our data establish that under the basal condition and in acquired glomerular diseases, nephrin is required to maintain slit diaphragm integrity and slit diaphragm-mediated signaling to preserve glomerular function and podocyte viability in adult mice.


Subject(s)
Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/physiology , Membrane Proteins/physiology , Podocytes/cytology , Podocytes/physiology , Age Factors , Animals , Cell Survival , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic
13.
Am J Pathol ; 184(7): 1940-56, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24952428

ABSTRACT

The silent mating type information regulation 2 homolog 1 gene (Sirt1) encodes an NAD-dependent deacetylase that modifies the activity of well-known transcriptional regulators affected in kidney diseases. Sirt1 is expressed in the kidney podocyte, but its function in the podocyte is not clear. Genetically engineered mice with inducible and reversible Sirt1 knockdown in widespread, podocyte-specific, or tubular-specific patterns were generated. We found that mice with 80% knockdown of renal Sirt1 expression have normal glomerular function under the basal condition. When challenged with doxorubicin (Adriamycin), these mice develop marked albuminuria, glomerulosclerosis, mitochondrial injury, and impaired autophagy of damaged mitochondria. Reversal of Sirt1 knockdown during the early phase of Adriamycin-induced nephropathy prevented the progression of glomerular injury and reduced the accumulation of dysmorphic mitochondria in podocytes but did not reverse the progression of albuminuria and glomerulosclerosis. Sirt1 knockdown mice with diabetes mellitus, which is known to cause mitochondrial dysfunction in the kidney, developed more albuminuria and mitochondrial dysfunction compared with diabetic mice without Sirt1 knockdown. In conclusion, these results demonstrate that our RNA interference-mediated Sirt1 knockdown models are valid and versatile tools for characterizing the function of Sirt1 in the kidney; Sirt1 plays a role in homeostatic maintenance of podocytes under the condition of mitochondrial stress/injury.


Subject(s)
Disease Models, Animal , Podocytes/cytology , RNA Interference , Sirtuin 1/metabolism , Albuminuria , Animals , Autophagy , Cell Line , Diabetes Mellitus, Experimental/chemically induced , Doxorubicin , Gene Knockdown Techniques , Kidney/cytology , Kidney/metabolism , Kidney Diseases/chemically induced , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Mice, Knockout , Mitochondria/pathology , Podocytes/metabolism , Sirtuin 1/genetics
14.
Curr Diab Rep ; 15(7): 42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25971619

ABSTRACT

Diabetic kidney disease (DKD) is a common, complex condition that has become a significant public health problem. The beneficial effects of intensive glycemic control in type 1 diabetes mellitus on development of DKD are proven; however, the evidence for nephroprotection in patients with type 2 diabetes is conflicting. Moreover, a strategy of intensive glycemic control increases the risk for adverse effects (hypoglycemic episodes) with no obvious impact on macrovascular events or mortality in recent large randomized controlled trials. The risk for hypoglycemia with intensive therapy is heightened in patients with significant renal dysfunction, due to decreased renal clearance of insulin. Establishing an ideal level of glycemic control in patients requires an individualized approach taking into account duration of diabetes and presence of coexisting comorbidities and pre-existing DKD. In this article, we review the available evidence from both observational studies and randomized controlled trials and provide suggestions about evaluating the potential benefits and harm from intensive glycemic control in patients. We also discuss how in the future, a personalized approach using biomarkers might help identify patients most likely to respond as well as those most susceptible to harm. We believe that using the optimal level of glycemic control in diabetic patients using a multi-pronged strategy will improve individual patient outcomes and decrease the overall burden of morbidity and mortality.


Subject(s)
Diabetic Nephropathies/complications , Diabetic Nephropathies/drug therapy , Glucose/metabolism , Hyperglycemia/complications , Hyperglycemia/drug therapy , Biomarkers/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Disease Progression , Humans , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Kidney Function Tests , Randomized Controlled Trials as Topic
15.
J Interv Cardiol ; 28(3): 288-95, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25989717

ABSTRACT

BACKGROUND: Hemodialysis (HD) access failure is a common cause of increased morbidity and healthcare cost in patients with end stage renal disease (ESRD). Percutaneous balloon angioplasty has been used to treat hemodialysis access stenosis but is complicated by a high rate of restenosis. Percutaneous cutting balloon (PCB) angioplasty is an alternative approach that has shown to reduce restenosis. OBJECTIVES: The aim of the study is to assess the safety and efficacy of PCB angioplasty in comparison with conventional and high-pressure balloon angioplasty in the treatment of hemodialysis access site stenosis. METHODS: We searched PubMed, EMBASE and the Cochrane Central register of controlled trials (CENTRAL) databases through August 2014 and selected studies using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist. We included all randomized clinical trials with a head-to-head comparison between PCB and conventional or high-pressure balloon angioplasty RESULTS: Three studies with 1034 participants (age 60.7 (±12.9) years and 50.1% males) with 525 in PCB and 509 in control arm were included in the analysis. The immediate procedural success rate was not significantly different in the PCB angioplasty and control arm respectively, (87.2% vs. 83.7% RD -0.02; 95%CI -0.06 to 0.01; P = 0.38). The six-month target lesion patency was significantly higher in the PCB angioplasty arm (67.2% vs. 55.6% RD 0.12; 95%CI 0.05-0.19; P < 0.05) with number needed to treat (NNT) of 9. The device related complications were not statistically significant between groups (RD 0.03; 95%CI -0.02 to 0.07; P = 0.26). CONCLUSIONS: PCB angioplasty is effective in treatment of hemodialysis access stenosis, with significantly higher six-month patency compared to balloon angioplasty.


Subject(s)
Angioplasty, Balloon/methods , Arteriovenous Shunt, Surgical , Constriction, Pathologic/therapy , Humans , Randomized Controlled Trials as Topic , Renal Dialysis , Vascular Patency
16.
Am J Kidney Dis ; 62(6): 1077-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23791246

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is a common complication after cardiovascular surgery. The use of renin angiotensin system (RAS) blockers preoperatively is controversial due to conflicting results of their effect on the incidence of postoperative AKI and mortality. STUDY DESIGN: Meta-analysis of prospective or retrospective observational studies (1950 to January 2013) using MEDLINE, EMBASE, the Cochrane Library, conferences, and ClinicalTrials.gov, without language restriction. SETTING & POPULATION: Patients undergoing cardiovascular surgery. SELECTION CRITERIA FOR STUDIES: Retrospective or prospective studies evaluating the effect of preoperative use of RAS blockers in the development of postoperative AKI and/or mortality in adult patients. INTERVENTION: Preoperative use of RAS blockers. RAS-blocker use was defined as long-term use of either angiotensin-converting enzyme inhibitors or angiotensin receptor blockers until the day of surgery. OUTCOMES: The primary outcome was the development of postoperative AKI; the secondary outcome was mortality. AKI was defined by different authors using different criteria. Death was ascertained in the hospital, at 30 days, or at 90 days in different studies. RESULTS: 29 studies were included (4 prospective and 25 retrospective); 23 of these involving 69,027 patients examined AKI, and 18 involving 54,418 patients studied mortality. Heterogeneity was found across studies regarding AKI (I2 = 82.5%), whereas studies were homogeneous regarding mortality (I2 = 20.5%). Preoperative RAS-blocker use was associated with increased odds for both postoperative AKI (OR, 1.17; 95% CI, 1.01-1.36; P = 0.04) and mortality (OR, 1.20; 95% CI, 1.06-1.35; P = 0.005). LIMITATIONS: Lack of randomized controlled trials, different definitions of AKI, different durations of follow-up used to analyze death outcome, and inability to exclude outcome reporting bias. CONCLUSIONS: In retrospective studies, preoperative use of RAS blockers was associated with increased odds of postoperative AKI and mortality in patients undergoing cardiovascular surgery. A large, multicenter, randomized, controlled trial should be performed to confirm these findings.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/mortality , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Cardiac Surgical Procedures , Renin-Angiotensin System/drug effects , Thoracic Surgical Procedures , Adult , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronary Artery Bypass , Humans , Postoperative Complications/chemically induced , Postoperative Complications/mortality , Preoperative Care
17.
Article in English | MEDLINE | ID: mdl-37817877

ABSTRACT

Podocyte injury plays a crucial role in the progression of diabetic kidney disease (DKD). Injured podocytes demonstrate variations in nuclear shape and chromatin distribution. These morphometric changes have not yet been quantified in podocytes. Furthermore, the molecular mechanisms underlying these variations are poorly understood. Recent advances in omics have shed new lights into the biological mechanisms behind podocyte injury. However, there currently exists no study analyzing the biological mechanisms underlying podocyte morphometric variations during DKD. First, to study the importance of nuclear morphometrics, we performed morphometric quantification of podocyte nuclei from whole slide images of renal tissue sections obtained from murine models of DKD. Our results indicated that podocyte nuclear textural features demonstrate statistically significant difference in diabetic podocytes when compared to control. Additionally, the morphometric features demonstrated the existence of multiple subpopulations of podocytes suggesting a potential cause for their varying response to injury. Second, to study the underlying pathophysiology, we employed single cell RNA sequencing data from the murine models. Our results again indicated five subpopulations of podocytes in control and diabetic mouse models, validating the morphometrics-based results. Additionally, gene set enrichment analysis revealed epithelial to mesenchymal transition and apoptotic pathways in a subgroup of podocytes exclusive to diabetic mice, suggesting the molecular mechanism behind injury. Lastly, our results highlighted two distinct lineages of podocytes in control and diabetic cases suggesting a phenotypical change in podocytes during DKD. These results suggest that textural variations in podocyte nuclei may be key to understanding the pathophysiology behind podocyte injury.

18.
Article in English | MEDLINE | ID: mdl-34366543

ABSTRACT

In diabetic kidney disease (DKD), podocyte depletion, and the subsequent migration of parietal epithelial cells (PECs) to the tuft, is a precursor to progressive glomerular damage, but the limitations of brightfield microscopy currently preclude direct pathological quantitation of these cells. Here we present an automated approach to podocyte and PEC detection developed using kidney sections from mouse model emulating DKD, stained first for Wilms' Tumor 1 (WT1) (podocyte and PEC marker) by immunofluorescence, then post-stained with periodic acid-Schiff (PAS). A generative adversarial network (GAN)-based pipeline was used to translate these PAS-stained sections into WT1-labeled IF images, enabling in silico label-free podocyte and PEC identification in brightfield images. Our method detected WT1-positive cells with high sensitivity/specificity (0.87/0.92). Additionally, our algorithm performed with a higher Cohen's kappa (0.85) than the average manual identification by three renal pathologists (0.78). We propose that this pipeline will enable accurate detection of WT1-positive cells in research applications.

19.
JCI Insight ; 6(11)2021 06 08.
Article in English | MEDLINE | ID: mdl-33914709

ABSTRACT

Patients with chronic kidney disease (CKD) and end-stage renal disease suffer from increased cardiovascular events and cardiac mortality. Prior studies have demonstrated that a portion of this enhanced risk can be attributed to the accumulation of microbiota-derived toxic metabolites, with most studies focusing on the sulfonated form of p-cresol (PCS). However, unconjugated p-cresol (uPC) itself was never assessed due to rapid and extensive first-pass metabolism that results in negligible serum concentrations of uPC. These reports thus failed to consider the host exposure to uPC prior to hepatic metabolism. In the current study, not only did we measure the effect of altering the intestinal microbiota on lipid accumulation in coronary arteries, but we also examined macrophage lipid uptake and handling pathways in response to uPC. We found that atherosclerosis-prone mice fed a high-fat diet exhibited significantly higher coronary artery lipid deposits upon receiving fecal material from CKD mice. Furthermore, treatment with uPC increased total cholesterol, triglycerides, and hepatic and aortic fatty deposits in non-CKD mice. Studies employing an in vitro macrophage model demonstrated that uPC exposure increased apoptosis whereas PCS did not. Additionally, uPC exhibited higher potency than PCS to stimulate LDL uptake and only uPC induced endocytosis- and pinocytosis-related genes. Pharmacological inhibition of varying cholesterol influx and efflux systems indicated that uPC increased macrophage LDL uptake by activating macropinocytosis. Overall, these findings indicate that uPC itself had a distinct effect on macrophage biology that might have contributed to increased cardiovascular risk in patients with CKD.


Subject(s)
Aorta/metabolism , Cholesterol, LDL/metabolism , Coronary Artery Disease/metabolism , Cresols/metabolism , Gastrointestinal Microbiome , Liver/metabolism , Macrophages/metabolism , Pinocytosis/physiology , Renal Insufficiency, Chronic/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Cholesterol/metabolism , Cholesterol, LDL/drug effects , Coronary Artery Disease/pathology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cresols/pharmacology , Diet, High-Fat , Fecal Microbiota Transplantation , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/microbiology , Liver/drug effects , Liver/pathology , Macrophages/drug effects , Mice , Pinocytosis/drug effects , Renal Insufficiency, Chronic/microbiology , Triglycerides/metabolism
20.
AAPS J ; 23(2): 40, 2021 03 07.
Article in English | MEDLINE | ID: mdl-33677748

ABSTRACT

Therapeutic immunoglobulin G (IgG) antibodies comprise the largest class of protein therapeutics. Several factors that influence their overall disposition have been well-characterized, including target-mediated mechanics and convective flow. What remains poorly defined is the potential for non-targeted entry into various tissues or cell types by means of uptake via cell surface receptors at those sites. Megalin and cubilin are large endocytic receptors whose cooperative function plays important physiological roles at the tissues in which they are expressed. One such example is the kidney, where loss of either results in significant declines in proximal tubule protein reabsorption. Due to their diverse ligand profile and broad tissue expression, megalin and cubilin represent potential candidates for receptor-mediated uptake of IgG into various epithelia. Therefore, the objective of the current work was to determine if IgG was a novel ligand of megalin and/or cubilin. Direct binding was measured for human IgG with both megalin and the cubilin/amnionless complex. Additional work focusing on the megalin-IgG interaction was then conducted to build upon these findings. Cell uptake studies using megalin ligands for competitive inhibition or proximal tubule cells stably transduced with megalin-targeted shRNA constructs supported a role for megalin in the endocytosis of human IgG. Furthermore, a pharmacokinetic study using transgenic mice with a kidney-specific mosaic knockout of megalin demonstrated increased urinary excretion of human IgG in megalin knockout mice when compared to wild-type controls. These findings indicate that megalin is capable of binding and internalizing IgG via a high affinity interaction.


Subject(s)
Immunoglobulin G/pharmacology , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Animals , Cell Line , Endocytosis , Female , Gene Knockdown Techniques , Humans , Immunoglobulin G/isolation & purification , Immunoglobulin G/therapeutic use , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Knockout , Opossums , Rats , Renal Elimination
SELECTION OF CITATIONS
SEARCH DETAIL