Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell Proteomics ; 21(2): 100190, 2022 02.
Article in English | MEDLINE | ID: mdl-34958949

ABSTRACT

Hypoxia-induced intrauterine growth restriction increases the risk for cardiovascular, renal, and other chronic diseases in adults, representing thus a major public health problem. Still, not much is known about the fetal mechanisms that predispose these individuals to disease. Using a previously validated mouse model of fetal hypoxia and bottom-up proteomics, we characterize the response of the fetal kidney to chronic hypoxic stress. Fetal kidneys exhibit a dichotomous response to chronic hypoxia, comprising on the one hand cellular adaptations that promote survival (glycolysis, autophagy, and reduced DNA and protein synthesis), but on the other processes that induce a senescence-like phenotype (infiltration of inflammatory cells, DNA damage, and reduced proliferation). Importantly, chronic hypoxia also reduces the expression of the antiaging proteins klotho and Sirt6, a mechanism that is evolutionary conserved between mice and humans. Taken together, we uncover that predetermined aging during fetal development is a key event in chronic hypoxia, establishing a solid foundation for Barker's hypothesis of fetal programming of adult diseases. This phenotype is associated with a characteristic biomarker profile in tissue and serum samples, exploitable for detecting and targeting accelerated aging in chronic hypoxic human diseases.


Subject(s)
Fetal Hypoxia , Sirtuins , Aging , Animals , Fetal Development , Hypoxia , Mice , Phenotype
2.
Kidney Int ; 102(4): 708-719, 2022 10.
Article in English | MEDLINE | ID: mdl-35964799

ABSTRACT

The 13th International Podocyte Conference was held in Manchester, UK, and online from July 28 to 30, 2021. Originally planned for 2020, this biannual meeting was postponed by a year because of the coronavirus disease 2019 (COVID-19) pandemic and proceeded as an innovative hybrid meeting. In addition to in-person attendance, online registration was offered, and this attracted 490 conference registrations in total. As a Podocyte Conference first, a day for early-career researchers was introduced. This premeeting included talks from graduate students and postdoctoral researchers. It gave early career researchers the opportunity to ask a panel, comprising academic leaders and journal editors, about career pathways and the future for podocyte research. The main meeting over 3 days included a keynote talk and 4 focused sessions each day incorporating invited talks, followed by selected abstract presentations, and an open panel discussion. The conference concluded with a Patient Day, which brought together patients, clinicians, researchers, and industry representatives. The Patient Day was an interactive and diverse day. As well as updates on improving diagnosis and potential new therapies, the Patient Day included a PodoArt competition, exercise and cooking classes with practical nutrition advice, and inspirational stories from patients and family members. This review summarizes the exciting science presented during the 13th International Podocyte Conference and demonstrates the resilience of researchers during a global pandemic.


Subject(s)
COVID-19 , Podocytes , COVID-19/epidemiology , Humans , Translational Research, Biomedical
3.
Cell Tissue Res ; 385(2): 445-456, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34131806

ABSTRACT

Current therapeutic options for renal diseases are limited, and the search for disease-specific treatments is ongoing. Nanobodies, single-domain antibodies with many advantages over conventional antibodies, provide flexible, easy-to-format biologicals with many possible applications. Here, we discuss the potential use of nanobodies for renal diseases.


Subject(s)
Kidney Diseases/therapy , Single-Domain Antibodies/therapeutic use , Animals , Humans
4.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923831

ABSTRACT

A growing body of evidence suggests that low nephron numbers at birth can increase the risk of chronic kidney disease or hypertension later in life. Environmental stressors, such as maternal malnutrition, medication and smoking, can influence renal size at birth. Using metanephric organ cultures to model single-variable environmental conditions, models of maternal disease were evaluated for patterns of developmental impairment. While hyperthermia had limited effects on renal development, fetal iron deficiency was associated with severe impairment of renal growth and nephrogenesis with an all-proximal phenotype. Culturing kidney explants under high glucose conditions led to cellular and transcriptomic changes resembling human diabetic nephropathy. Short-term high glucose culture conditions were sufficient for long-term alterations in DNA methylation-associated epigenetic memory. Finally, the role of epigenetic modifiers in renal development was tested using a small compound library. Among the selected epigenetic inhibitors, various compounds elicited an effect on renal growth, such as HDAC (entinostat, TH39), histone demethylase (deferasirox, deferoxamine) and histone methyltransferase (cyproheptadine) inhibitors. Thus, metanephric organ cultures provide a valuable system for studying metabolic conditions and a tool for screening for epigenetic modifiers in renal development.


Subject(s)
Diabetic Nephropathies/genetics , Environment , Epigenesis, Genetic , Glucose/toxicity , Kidney/metabolism , Prenatal Exposure Delayed Effects/genetics , Animals , DNA Methylation , Female , Humans , Iron Deficiencies , Kidney/drug effects , Mice , Organ Culture Techniques/methods , Pregnancy , Transcriptome
5.
Kidney Int ; 98(6): 1434-1448, 2020 12.
Article in English | MEDLINE | ID: mdl-32603735

ABSTRACT

Podocyte maintenance and stress resistance are exquisitely based on high basal rates of autophagy making these cells a unique model to unravel mechanisms of autophagy regulation. Polyamines have key cellular functions such as proliferation, nucleic acid biosynthesis and autophagy. Here we test whether endogenous spermidine signaling is a driver of basal and dynamic autophagy in podocytes by using genetic and pharmacologic approaches to interfere with different steps of polyamine metabolism. Translational studies revealed altered spermidine signaling in focal segmental glomerulosclerosis in vivo and in vitro. Exogenous spermidine supplementation emerged as new treatment strategy by successfully activating autophagy in vivo via inhibition of EP300, a protein with an essential role in controlling cell growth, cell division and prompting cells to differentiate to take on specialized functions. Surprisingly, gas chromatography-mass spectroscopy based untargeted metabolomics of wild type and autophagy deficient primary podocytes revealed a positive feedback mechanism whereby autophagy itself maintains polyamine metabolism and spermidine synthesis. The transcription factor MAFB acted as an upstream regulator of polyamine metabolism. Thus, our data highlight a novel positive feedback loop of autophagy and spermidine signaling allowing maintenance of high basal levels of autophagy as a key mechanism to sustain the filtration barrier. Hence, spermidine supplementation may emerge as a new therapeutic to restore autophagy in glomerular disease.


Subject(s)
Glomerulosclerosis, Focal Segmental , Podocytes , Spermidine , Autophagy , Cell Proliferation , Humans , Spermidine/metabolism
6.
J Am Soc Nephrol ; 30(1): 63-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30518531

ABSTRACT

BACKGROUND: Nephron number is a major determinant of long-term renal function and cardiovascular risk. Observational studies suggest that maternal nutritional and metabolic factors during gestation contribute to the high variability of nephron endowment. However, the underlying molecular mechanisms have been unclear. METHODS: We used mouse models, including DNA methyltransferase (Dnmt1, Dnmt3a, and Dnmt3b) knockout mice, optical projection tomography, three-dimensional reconstructions of the nephrogenic niche, and transcriptome and DNA methylation analysis to characterize the role of DNA methylation for kidney development. RESULTS: We demonstrate that DNA hypomethylation is a key feature of nutritional kidney growth restriction in vitro and in vivo, and that DNA methyltransferases Dnmt1 and Dnmt3a are highly enriched in the nephrogenic zone of the developing kidneys. Deletion of Dnmt1 in nephron progenitor cells (in contrast to deletion of Dnmt3a or Dnm3b) mimics nutritional models of kidney growth restriction and results in a substantial reduction of nephron number as well as renal hypoplasia at birth. In Dnmt1-deficient mice, optical projection tomography and three-dimensional reconstructions uncovered a significant reduction of stem cell niches and progenitor cells. RNA sequencing analysis revealed that global DNA hypomethylation interferes in the progenitor cell regulatory network, leading to downregulation of genes crucial for initiation of nephrogenesis, Wt1 and its target Wnt4. Derepression of germline genes, protocadherins, Rhox genes, and endogenous retroviral elements resulted in the upregulation of IFN targets and inhibitors of cell cycle progression. CONCLUSIONS: These findings establish DNA methylation as a key regulatory event of prenatal renal programming, which possibly represents a fundamental link between maternal nutritional factors during gestation and reduced nephron number.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Kidney/embryology , Organogenesis/genetics , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cells, Cultured , DNA Methylation , Gene Expression Regulation, Developmental/genetics , Immunohistochemistry , Mice , Mice, Knockout , Nephrons/cytology , Nephrons/physiology , Rats , Rats, Wistar , Sensitivity and Specificity , Stem Cells/physiology , DNA Methyltransferase 3B
7.
Kidney Int ; 96(4): 850-861, 2019 10.
Article in English | MEDLINE | ID: mdl-31420194

ABSTRACT

The podocyte is a key component of the glomerular filtration barrier. Podocyte dysfunction is central to the underlying pathophysiology of many common glomerular diseases, including diabetic nephropathy, glomerulonephritis and genetic forms of nephrotic syndrome. Collectively, these conditions affect millions of people worldwide, and account for the majority of kidney diseases requiring dialysis and transplantation. The 12th International Podocyte Conference was held in Montreal, Canada from May 30 to June 2, 2018. The primary aim of this conference was to bring together nephrologists, clinician scientists, basic scientists and their trainees from all over the world to present their research and to establish networks with the common goal of developing new therapies for glomerular diseases based on the latest advances in podocyte biology. This review briefly highlights recent advances made in understanding podocyte structure and metabolism, experimental systems in which to study podocytes and glomerular disease, disease mediators, genetic and immune origins of glomerulopathies, and the development of novel therapeutic agents to protect podocyte and glomerular injury.


Subject(s)
Diabetic Nephropathies/therapy , Glomerular Filtration Barrier/physiopathology , Glomerulonephritis/therapy , Nephrotic Syndrome/therapy , Podocytes/pathology , Animals , Canada , Congresses as Topic , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Humans , Nephrotic Syndrome/pathology , Nephrotic Syndrome/physiopathology
8.
Am J Physiol Renal Physiol ; 315(6): F1526-F1535, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30207169

ABSTRACT

The role of CX3CR1, also known as fractalkine receptor, in hypertension is unknown. The present study determined the role of the fractalkine receptor CX3CR1 in hypertensive renal and cardiac injury. Expression of CX3CR1 was determined using CX3CR1GFP/+ mice that express a green fluorescent protein (GFP) reporter in CX3CR1+ cells. FACS analysis of leukocytes isolated from the kidney showed that 34% of CD45+ cells expressed CX3CR1. Dendritic cells were the majority of positive cells (67%) followed by macrophages (10%), NK cells (6%), and T cells (10%). With the use of confocal microscopy, the receptor was detected in the kidney only on infiltrating cells but not on resident renal cells. To evaluate the role of CX3CR1 in hypertensive end-organ injury, an aggravated model of hypertension was used. Unilateral nephrectomy was performed followed by infusion of angiotensin II (ANG II, 1.5 ng·g-1·min-1) and a high-salt diet in wild-type ( n = 15) and CX3CR1-deficient mice ( n = 18). CX3CR1 deficiency reduced the number of renal dendritic cells and increased the numbers of renal CD11b/F4/80+ macrophages and CD11b/Ly6G+ neutrophils in ANG II-infused mice. Surprisingly, CX3CR1-deficient mice exhibited increased albuminuria, glomerular injury, and reduced podocyte density in spite of similar levels of arterial hypertension. In contrast, cardiac damage as assessed by increased heart weight, cardiac fibrosis, and expression of fetal genes, and matrix components were not different between both genotypes. Our findings suggest that CX3CR1 exerts protective properties by modulating the invasion of inflammatory cells in hypertensive renal injury. CX3CR1 inhibition should be avoided in hypertension because it may promote hypertensive renal injury.


Subject(s)
Angiotensin II , Arterial Pressure , CX3C Chemokine Receptor 1/metabolism , Dendritic Cells/metabolism , Hypertension/metabolism , Kidney Diseases/prevention & control , Kidney/metabolism , Leukocytes/metabolism , Macrophages/metabolism , Albuminuria/metabolism , Albuminuria/physiopathology , Albuminuria/prevention & control , Animals , CX3C Chemokine Receptor 1/deficiency , CX3C Chemokine Receptor 1/genetics , Chemotaxis, Leukocyte , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Killer Cells, Natural/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
10.
J Am Soc Nephrol ; 28(6): 1707-1715, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28404664

ABSTRACT

Podocyte and glomerular research is center stage for the development of improved preventive and therapeutic strategies for chronic progressive kidney diseases. Held April 3-6, 2016, the 11th International Podocyte Conference took place in Haifa and Jerusalem, Israel, where participants from all over the world presented their work on new developments in podocyte research. In this review, we briefly highlight the advances made in characterizing the mechanisms involved in podocyte development, metabolism, acquired injury, and repair, including progress in determining the roles of genetic variants and microRNA in particular, as well as the advances made in diagnostic techniques and therapeutics.


Subject(s)
Kidney Diseases/etiology , Kidney Glomerulus/physiology , Podocytes/physiology , Animals , Biomedical Research , Humans , Kidney Diseases/diagnosis , Kidney Diseases/therapy , MicroRNAs/physiology , Podocytes/ultrastructure , Signal Transduction , Stem Cells
11.
J Am Soc Nephrol ; 28(1): 230-241, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27297946

ABSTRACT

Renal proximal tubular cells constantly recycle nutrients to ensure minimal loss of vital substrates into the urine. Although most of the transport mechanisms have been discovered at the molecular level, little is known about the factors regulating these processes. Here, we show that mTORC1 and mTORC2 specifically and synergistically regulate PTC endocytosis and transport processes. Using a conditional mouse genetic approach to disable nonredundant subunits of mTORC1, mTORC2, or both, we showed that mice lacking mTORC1 or mTORC1/mTORC2 but not mTORC2 alone develop a Fanconi-like syndrome of glucosuria, phosphaturia, aminoaciduria, low molecular weight proteinuria, and albuminuria. Interestingly, proteomics and phosphoproteomics of freshly isolated kidney cortex identified either reduced expression or loss of phosphorylation at critical residues of different classes of specific transport proteins. Functionally, this resulted in reduced nutrient transport and a profound perturbation of the endocytic machinery, despite preserved absolute expression of the main scavenger receptors, MEGALIN and CUBILIN. Our findings highlight a novel mTOR-dependent regulatory network for nutrient transport in renal proximal tubular cells.


Subject(s)
Endocytosis/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Protein Transport
12.
J Am Soc Nephrol ; 28(7): 2144-2157, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28270414

ABSTRACT

Mammalian target of rapamycin (mTOR) signaling is involved in a variety of kidney diseases. Clinical trials administering mTOR inhibitors to patients with FSGS, a prototypic podocyte disease, led to conflicting results, ranging from remission to deterioration of kidney function. Here, we combined complex genetic titration of mTOR complex 1 (mTORC1) levels in murine glomerular disease models, pharmacologic studies, and human studies to precisely delineate the role of mTOR in FSGS. mTORC1 target genes were significantly induced in microdissected glomeruli from both patients with FSGS and a murine FSGS model. Furthermore, a mouse model with constitutive mTORC1 activation closely recapitulated human FSGS. Notably, the complete knockout of mTORC1 by induced deletion of both Raptor alleles accelerated the progression of murine FSGS models. However, lowering mTORC1 signaling by deleting just one Raptor allele ameliorated the progression of glomerulosclerosis. Similarly, low-dose treatment with the mTORC1 inhibitor rapamycin efficiently diminished disease progression. Mechanistically, complete pharmacologic inhibition of mTOR in immortalized podocytes shifted the cellular energy metabolism toward reduced rates of oxidative phosphorylation and anaerobic glycolysis, which correlated with increased production of reactive oxygen species. Together, these data suggest that podocyte injury and loss is commonly followed by adaptive mTOR activation. Prolonged mTOR activation, however, results in a metabolic podocyte reprogramming leading to increased cellular stress and dedifferentiation, thus offering a treatment rationale for incomplete mTOR inhibition.


Subject(s)
Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/prevention & control , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/physiology , Signal Transduction/drug effects , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology , Animals , Disease Progression , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice
13.
Cell Tissue Res ; 369(1): 75-92, 2017 07.
Article in English | MEDLINE | ID: mdl-28286899

ABSTRACT

DNA methylation and histone modifications determine renal programming and the development and progression of renal disease. The identification of the way in which the renal cell epigenome is altered by environmental modifiers driving the onset and progression of renal diseases has extended our understanding of the pathophysiology of kidney disease progression. In this review, we focus on current knowledge concerning the implications of epigenetic modifications during renal disease from early development to chronic kidney disease progression including renal fibrosis, diabetic nephropathy and the translational potential of identifying new biomarkers and treatments for the prevention and therapy of chronic kidney disease and end-stage kidney disease.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Kidney Failure, Chronic/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Fibrosis , Humans , Kidney/pathology , Kidney Failure, Chronic/pathology , Renal Insufficiency, Chronic/pathology
14.
Proc Natl Acad Sci U S A ; 111(27): E2817-26, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-24958889

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cell metabolism and autophagy. Despite widespread clinical use of mTORC1 inhibitors, the role of mTORC1 in renal tubular function and kidney homeostasis remains elusive. By using constitutive and inducible deletion of conditional Raptor alleles in renal tubular epithelial cells, we discovered that mTORC1 deficiency caused a marked concentrating defect, loss of tubular cells, and slowly progressive renal fibrosis. Transcriptional profiling revealed that mTORC1 maintains renal tubular homeostasis by controlling mitochondrial metabolism and biogenesis as well as transcellular transport processes involved in countercurrent multiplication and urine concentration. Although mTORC2 partially compensated for the loss of mTORC1, exposure to ischemia and reperfusion injury exaggerated the tubular damage in mTORC1-deficient mice and caused pronounced apoptosis, diminished proliferation rates, and delayed recovery. These findings identify mTORC1 as an important regulator of tubular energy metabolism and as a crucial component of ischemic stress responses.


Subject(s)
Homeostasis/physiology , Ischemia/physiopathology , Kidney Tubules/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Blotting, Western , Kidney Tubules/blood supply , Magnetic Resonance Imaging , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Polyuria/genetics , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
15.
J Am Soc Nephrol ; 25(4): 707-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24408871

ABSTRACT

Podocyte loss is a major determinant of progressive CKD. Although recent studies showed that a subset of parietal epithelial cells can serve as podocyte progenitors, the role of podocyte turnover and regeneration in repair, aging, and nephron loss remains unclear. Here, we combined genetic fate mapping with highly efficient podocyte isolation protocols to precisely quantify podocyte turnover and regeneration. We demonstrate that parietal epithelial cells can give rise to fully differentiated visceral epithelial cells indistinguishable from resident podocytes and that limited podocyte renewal occurs in a diphtheria toxin model of acute podocyte ablation. In contrast, the compensatory programs initiated in response to nephron loss evoke glomerular hypertrophy, but not de novo podocyte generation. In addition, no turnover of podocytes could be detected in aging mice under physiologic conditions. In the absence of podocyte replacement, characteristic features of aging mouse kidneys included progressive accumulation of oxidized proteins, deposits of protein aggregates, loss of podocytes, and glomerulosclerosis. In summary, quantitative investigation of podocyte regeneration in vivo provides novel insights into the mechanism and capacity of podocyte turnover and regeneration in mice. Our data reveal that podocyte generation is mainly confined to glomerular development and may occur after acute glomerular injury, but it fails to regenerate podocytes in aging kidneys or in response to nephron loss.


Subject(s)
Aging/pathology , Kidney Glomerulus/pathology , Podocytes/physiology , Animals , Flow Cytometry , Hypertrophy , Mice , Regeneration
16.
Am J Pathol ; 183(2): 333-5, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23727347

ABSTRACT

This Commentary highlights the article by Pippin et al that showed that renin lineage cells may enhance glomerular regeneration by serving as progenitors for glomerular epithelial cells in glomerular disease characterized by podocyte depletion.


Subject(s)
Glomerulosclerosis, Focal Segmental/pathology , Podocytes/physiology , Renin/metabolism , Stem Cells/physiology , Animals , Female , Male
17.
Nephrol Dial Transplant ; 29 Suppl 1: i9-i18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24493874

ABSTRACT

Renal epithelial function is the cornerstone of key excretory processes performed by our kidneys. Most of these tasks need to be tightly controlled to keep our internal environment in balance. Recently, the mTOR signalling network emerged as a key pathway controlling renal epithelial cells from the glomerular tuft along the entire nephron. Both mTOR complexes, mTORC1 and mTORC2, regulate such diverse processes as glomerular filtration and the fine tuning of tubular electrolyte balance. Most importantly, dysregulation of mTOR signalling contributes to prevalent kidney diseases like diabetic nephropathy and cystic kidney disease. The following review shall summarize our current knowledge of the renal epithelial mTOR signalling system under physiological and pathophysiological conditions.


Subject(s)
Epithelial Cells/metabolism , Kidney Diseases/physiopathology , Kidney/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Humans , Kidney Glomerulus/metabolism
18.
J Am Soc Nephrol ; 24(11): 1756-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23949798

ABSTRACT

In CKD, the risk of kidney failure and death depends on the severity of proteinuria, which correlates with the extent of podocyte loss and glomerular scarring. We investigated whether proteinuria contributes directly to progressive glomerulosclerosis through the suppression of podocyte regeneration and found that individual components of proteinuria exert distinct effects on renal progenitor survival and differentiation toward a podocyte lineage. In particular, albumin prevented podocyte differentiation from human renal progenitors in vitro by sequestering retinoic acid, thus impairing retinoic acid response element (RARE)-mediated transcription of podocyte-specific genes. In mice with Adriamycin nephropathy, a model of human FSGS, blocking endogenous retinoic acid synthesis increased proteinuria and exacerbated glomerulosclerosis. This effect was related to a reduction in podocyte number, as validated through genetic podocyte labeling in NPHS2.Cre;mT/mG transgenic mice. In RARE-lacZ transgenic mice, albuminuria reduced retinoic acid bioavailability and impaired RARE activation in renal progenitors, inhibiting their differentiation into podocytes. Treatment with retinoic acid restored RARE activity and induced the expression of podocyte markers in renal progenitors, decreasing proteinuria and increasing podocyte number, as demonstrated in serial biopsy specimens. These results suggest that albumin loss through the damaged filtration barrier impairs podocyte regeneration by sequestering retinoic acid and promotes the generation of FSGS lesions. Our findings may explain why reducing proteinuria delays CKD progression and provide a biologic rationale for the clinical use of pharmacologic modulators to induce regression of glomerular diseases.


Subject(s)
Albuminuria/complications , Podocytes/physiology , Regeneration , Tretinoin/metabolism , Albuminuria/pathology , Animals , Cells, Cultured , Female , Glomerulosclerosis, Focal Segmental/etiology , Humans , Mice , Mice, SCID , Response Elements/physiology , Tretinoin/pharmacology
19.
J Clin Invest ; 134(17)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39225099

ABSTRACT

Adeno-associated virus (AAV) is a promising in vivo gene delivery platform showing advantages in delivering therapeutic molecules to difficult or undruggable cells. However, natural AAV serotypes have insufficient transduction specificity and efficiency in kidney cells. Here, we developed an evolution-directed selection protocol for renal glomeruli and identified what we believe to be a new vector termed AAV2-GEC that specifically and efficiently targets the glomerular endothelial cells (GEC) after systemic administration and maintains robust GEC tropism in healthy and diseased rodents. AAV2-GEC-mediated delivery of IdeS, a bacterial antibody-cleaving proteinase, provided sustained clearance of kidney-bound antibodies and successfully treated antiglomerular basement membrane glomerulonephritis in mice. Taken together, this study showcases the potential of AAV as a gene delivery platform for challenging cell types. The development of AAV2-GEC and its successful application in the treatment of antibody-mediated kidney disease represents a significant step forward and opens up promising avenues for kidney medicine.


Subject(s)
Dependovirus , Genetic Therapy , Genetic Vectors , Animals , Dependovirus/genetics , Mice , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Endothelial Cells/metabolism , Kidney Glomerulus/pathology , Glomerulonephritis/therapy , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Anti-Glomerular Basement Membrane Disease/therapy , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/immunology
SELECTION OF CITATIONS
SEARCH DETAIL