Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 27(9): 1192-1205, 2023 05.
Article in English | MEDLINE | ID: mdl-37056054

ABSTRACT

Chronic kidney diseases affect a substantial percentage of the adult population worldwide. This observation emphasizes the need for novel insights into the molecular mechanisms that control the onset and progression of renal diseases. Recent advances in genomics have uncovered a previously unanticipated link between the non-coding genome and human kidney diseases. Here we screened and analysed long non-coding RNAs (lncRNAs) previously identified in mouse kidneys by genome-wide transcriptomic analysis, for conservation in humans and differential expression in renal tissue from healthy and diseased individuals. Our data suggest that LINC01187 is strongly down-regulated in human kidney tissues of patients with diabetic nephropathy and rapidly progressive glomerulonephritis, as well as in murine models of kidney diseases, including unilateral ureteral obstruction, nephrotoxic serum-induced glomerulonephritis and ischemia/reperfusion. Interestingly, LINC01187 overexpression in human kidney cells in vitro inhibits cell death indicating an anti-apoptotic function. Collectively, these data suggest a negative association of LINC01187 expression with renal diseases implying a potential protective role.


Subject(s)
Diabetic Nephropathies , Glomerulonephritis , RNA, Long Noncoding , Animals , Humans , Mice , Diabetic Nephropathies/metabolism , Down-Regulation/genetics , Glomerulonephritis/metabolism , Kidney/metabolism , RNA, Long Noncoding/metabolism
2.
Int J Mol Sci ; 24(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36769045

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a major cause of end-stage renal disease and remains without specific treatment. To identify new events during FSGS progression, we used an experimental model of FSGS associated with nephroangiosclerosis in rats injected with L-NAME (Nω-nitro-L-arginine methyl ester). After transcriptomic analysis we focused our study on the role of Isthmin-1 (ISM1, an anti-angiogenic protein involved in endothelial cell apoptosis. We studied the renal expression of ISM1 in L-NAME rats and other models of proteinuria, particularly at the glomerular level. In the L-NAME model, withdrawal of the stimulus partially restored basal ISM1 levels, along with an improvement in renal function. In other four animal models of proteinuria, ISM1 was overexpressed and localized in podocytes while the renal function was degraded. Together these facts suggest that the glomerular expression of ISM1 correlates directly with the progression-recovery of the disease. Further in vitro experiments demonstrated that ISM1 co-localized with its receptors GRP78 and integrin αvß5 on podocytes. Treatment of human podocytes with low doses of recombinant ISM1 decreased cell viability and induced caspase activation. Stronger ISM1 stimuli in podocytes dropped mitochondrial membrane potential and induced nuclear translocation of apoptosis-inducing factor (AIF). Our results suggest that ISM1 participates in the progression of glomerular diseases and promotes podocyte apoptosis in two different complementary ways: one caspase-dependent and one caspase-independent associated with mitochondrial destabilization.


Subject(s)
Glomerulosclerosis, Focal Segmental , Podocytes , Animals , Humans , Rats , Angiogenesis Inhibitors/therapeutic use , Caspases/metabolism , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/metabolism , NG-Nitroarginine Methyl Ester/metabolism , Podocytes/metabolism , Proteinuria/metabolism
3.
Cells ; 11(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35883655

ABSTRACT

Diabetic nephropathy (DN) remains the most common reason for end-stage renal disease and a leading cause of kidney replacement therapy. Multifactorial pathophysiological mechanisms underlie the development of DN. Among the signalling pathways involved, nuclear factor-κB (NF-κB) plays a key role in pathogenesis triggering inflammation, oxidative stress and fibrosis. Recent evidence shows that periostin, a matricellular protein, is involved in the development of renal glomerular diseases through interaction with NF-κB signalling. The aim of the present study is to investigate the contribution of periostin and its interaction with NF-κB in DN development. To this end, we used the BTBR ob/ob mice model of diabetes type 2, and we applied transcriptomic analysis, immunostaining and methods quantifying protein and mRNA expressions. We found that increased periostin expression was correlated with decreased renal function, advanced stage renal damage and fibrosis, and NF-κB activation. Subsequently, we identified novel pathways and genes regulated by the NF-κB-periostin interaction which are involved in the mechanisms of progression of DN. Some of these genes, such as FGF1 and GDF15, have the potential to be new biomarkers and/or targets for the therapy of DN.


Subject(s)
Cell Adhesion Molecules , Diabetes Mellitus , Diabetic Nephropathies , NF-kappa B , Animals , Cell Adhesion Molecules/metabolism , Diabetes Mellitus/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Fibrosis , Kidney/pathology , Mice , NF-kappa B/metabolism , Signal Transduction
4.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055068

ABSTRACT

BACKGROUND: Polycystic kidney disease (PKD) is a genetic disorder affecting millions of people worldwide that is characterized by fluid-filled cysts and leads to end-stage renal disease (ESRD). The hallmarks of PKD are proliferation and dedifferentiation of tubular epithelial cells, cellular processes known to be regulated by Notch signaling. METHODS: We found increased Notch3 expression in human PKD and renal cell carcinoma biopsies. To obtain insight into the underlying mechanisms and the functional consequences of this abnormal expression, we developed a transgenic mouse model with conditional overexpression of the intracellular Notch3 (ICN3) domain specifically in renal tubules. We evaluated the alterations in renal function (creatininemia, BUN) and structure (cysts, fibrosis, inflammation) and measured the expression of several genes involved in Notch signaling and the mechanisms of inflammation, proliferation, dedifferentiation, fibrosis, injury, apoptosis and regeneration. RESULTS: After one month of ICN3 overexpression, kidneys were larger with tubules grossly enlarged in diameter, with cell hypertrophy and hyperplasia, exclusively in the outer stripe of the outer medulla. After three months, mice developed numerous cysts in proximal and distal tubules. The cysts had variable sizes and were lined with a single- or multilayered, flattened, cuboid or columnar epithelium. This resulted in epithelial hyperplasia, which was observed as protrusions into the cystic lumen in some of the renal cysts. The pre-cystic and cystic epithelium showed increased expression of cytoskeletal filaments and markers of epithelial injury and dedifferentiation. Additionally, the epithelium showed increased proliferation with an aberrant orientation of the mitotic spindle. These phenotypic tubular alterations led to progressive interstitial inflammation and fibrosis. CONCLUSIONS: In summary, Notch3 signaling promoted tubular cell proliferation, the alignment of cell division, dedifferentiation and hyperplasia, leading to cystic kidney diseases and pre-neoplastic lesions.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules/metabolism , Polycystic Kidney Diseases/etiology , Polycystic Kidney Diseases/metabolism , Receptor, Notch3/metabolism , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Epithelial Cells/pathology , Fibrosis , Gene Expression , Immunohistochemistry , Kidney Neoplasms/etiology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules/pathology , Mice , Polycystic Kidney Diseases/pathology , Receptor, Notch3/genetics
5.
J Am Soc Nephrol ; 31(1): 85-100, 2020 01.
Article in English | MEDLINE | ID: mdl-31690575

ABSTRACT

BACKGROUND: The matricellular protein periostin has been associated with CKD progression in animal models and human biopsy specimens. Periostin functions by interacting with extracellular matrix components to drive collagen fibrillogenesis and remodeling or by signaling through cell-surface integrin receptors to promote cell adhesion, migration, and proliferation. However, its role in AKI is unknown. METHODS: We used mice with conditional tubule-specific overexpression of periostin or knockout mice lacking periostin expression in the renal ischemia-reperfusion injury model, and primary cultures of isolated tubular cells in a hypoxia-reoxygenation model. RESULTS: Tubular epithelial cells showed strong production of periostin during the repair phase of ischemia reperfusion. Periostin overexpression protected mice from renal injury compared with controls, whereas knockout mice showed increased tubular injury and deteriorated renal function. Periostin interacted with its receptor, integrin-ß1, to inhibit tubular cell cycle arrest and apoptosis in in vivo and in vitro models. After ischemia-reperfusion injury, periostin-overexpressing mice exhibited diminished expression of proinflammatory molecules and had more F4/80+ macrophages compared with knockout mice. Macrophages from periostin-overexpressing mice showed increased proliferation and expression of proregenerative factors after ischemia-reperfusion injury, whereas knockout mice exhibited the opposite. Coculturing a macrophage cell line with hypoxia-treated primary tubules overexpressing periostin, or treating such macrophages with recombinant periostin, directly induced macrophage proliferation and expression of proregenerative molecules. CONCLUSIONS: In contrast to the detrimental role of periostin in CKD, we discovered a protective role of periostin in AKI. Our findings suggest periostin may be a novel and important mediator of mechanisms controlling renal repair after AKI.


Subject(s)
Acute Kidney Injury , Cell Adhesion Molecules/physiology , Cell Proliferation , Macrophages/physiology , Acute Kidney Injury/etiology , Animals , Disease Models, Animal , Kidney/blood supply , Male , Mice , Mice, Knockout , Reperfusion Injury/complications , Reperfusion Injury/pathology
6.
JACC Basic Transl Sci ; 4(6): 717-732, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31709320

ABSTRACT

Acute kidney injury is associated with increased risk of heart failure and mortality. This study demonstrates that acute kidney injury induces remote cardiac dysfunction, damage, injury, and fibrosis via a galectin-3 (Gal-3) dependent pathway. Gal-3 originates from bone marrow-derived immune cells. Cardiac damage could be prevented by blocking this pathway.

7.
J Histochem Cytochem ; 67(9): 701-715, 2019 09.
Article in English | MEDLINE | ID: mdl-31116064

ABSTRACT

Renal fibrosis is an important component of chronic kidney disease, an incurable pathology with increasing prevalence worldwide. With a lack of available therapeutic options, end-stage renal disease is currently treated with renal replacement therapy through dialysis or transplantation. In recent years, many efforts have been made to identify novel targets for therapy of renal diseases, with special focus on the characterization of unknown mediators and pathways participating in renal fibrosis development. Using experimental models of renal disease and patient biopsies, we identified four novel mediators of renal fibrosis with potential to constitute future therapeutic targets against kidney disease: discoidin domain receptor 1, periostin, connexin 43, and cannabinoid receptor 1. The four candidates were highly upregulated in different models of renal disease and were localized at the sites of injury. Subsequent studies showed that they are centrally involved in the underlying mechanisms of renal fibrosis progression. Interestingly, inhibition of either of these proteins by different strategies, including gene deletion, antisense administration, or specific blockers, delayed the progression of renal disease and preserved renal structure and function, even when the inhibition started after initiation of the disease. This review will summarize the current findings on these candidates emphasizing on their potential to constitute future targets of therapy.


Subject(s)
Extracellular Matrix/pathology , Kidney/pathology , Molecular Targeted Therapy , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Animals , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Connexin 43/analysis , Connexin 43/metabolism , Discoidin Domain Receptor 1/analysis , Discoidin Domain Receptor 1/metabolism , Drug Discovery/methods , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibrosis , Humans , Kidney/drug effects , Kidney/metabolism , Molecular Targeted Therapy/methods , Receptor, Cannabinoid, CB1/analysis , Receptor, Cannabinoid, CB1/metabolism , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/metabolism
8.
Cell Physiol Biochem ; 49(3): 985, 2018.
Article in English | MEDLINE | ID: mdl-30196298

ABSTRACT

Chronic kidney disease is an incurable to date pathology with a continuously growing incidence that contributes to the increase of the number of deaths worldwide. With currently no efficient prognostic or therapeutic options being available, the only possibility for treatment of end-stage renal disease is renal replacement therapy through dialysis or transplantation. Understanding the molecular mechanisms participating in the progression of renal diseases and uncovering the pathways implicated will permit the identification of novel and more efficient targets of therapy. Connexin43 was recently identified as a novel player in the development of chronic kidney disease. It was found de novo expressed and/or differentially localized in various renal cell populations during progression of renal disease, indicating an abnormal connexin signaling, both in patients and animal models. Subsequent in vivo studies demonstrated that connexin43 is involved in mediating inflammatory and fibrotic processes contributing to renal damage. Genetic, pharmaco-genetic or peptide-based inhibition of connexin43 in animal models and cell culture systems was successful in preventing the progression of the pathology and preserving the cell phenotypes. This review will summarize the recent advances on connexin43 in the field of kidney diseases and discuss the potential of future connexin43-based therapies against chronic kidney disease.


Subject(s)
Connexin 43/metabolism , Renal Insufficiency, Chronic/pathology , Connexin 43/antagonists & inhibitors , Connexin 43/genetics , Humans , Kidney/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Oligonucleotides, Antisense/therapeutic use , Peptides/chemistry , Peptides/metabolism , Peptides/therapeutic use , Podocytes/cytology , Podocytes/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism
9.
J Cell Mol Med ; 22(9): 4139-4149, 2018 09.
Article in English | MEDLINE | ID: mdl-29956451

ABSTRACT

Chronic kidney disease, the end result of most renal and some systemic diseases, is a common condition where renal function is compromised due to fibrosis. During renal fibrosis, calreticulin, a multifunctional chaperone of the endoplasmic reticulum (ER) is up-regulated in tubular epithelial cells (TECs) both in vitro and in vivo. Proteomic analysis of cultured TECs overexpressing calreticulin led to the identification of the family of 14-3-3 proteins as key proteins overexpressed as well. Furthermore, an increased expression in the majority of 14-3-3 family members was observed in 3 different animal models of renal pathologies: the unilateral ureteric obstruction, the nephrotoxic serum administration and the ischaemia-reperfusion. In all these models, the 14-3-3σ isoform (also known as stratifin) was predominantly overexpressed. As in all these models ischaemia is a common denominator, we showed that the ischaemia-induced transcription factor HIF1α is specifically associated with the promoter region of the 14-3-3σ gene. Finally, we evaluated the expression of the family of 14-3-3 proteins and specifically 14-3-3σ in biopsies from IgA nephropathy and membranous nephropathy patients. These results propose an involvement of 14-3-3σ in renal pathology and provide evidence for the first time that hypoxia may be responsible for its altered expression.


Subject(s)
14-3-3 Proteins/genetics , Biomarkers, Tumor/genetics , Exoribonucleases/genetics , Glomerulonephritis, IGA/genetics , Glomerulonephritis, Membranous/genetics , Renal Insufficiency, Chronic/genetics , Reperfusion Injury/genetics , Ureteral Obstruction/genetics , 14-3-3 Proteins/metabolism , Animals , Biomarkers, Tumor/metabolism , Calreticulin/genetics , Calreticulin/metabolism , Cell Line , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Exoribonucleases/metabolism , Fibrosis , Gene Expression Regulation , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Proteomics/methods , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
10.
Kidney Int ; 94(1): 126-138, 2018 07.
Article in English | MEDLINE | ID: mdl-29751972

ABSTRACT

Acute kidney injury is a major risk factor for subsequent chronic renal and/or cardiovascular complications. Previous studies have shown that Notch3 was de novo expressed in the injured renal epithelium in the early phases of chronic kidney disease. Here we examined whether Notch3 is involved in the inflammatory response and the epithelial cell damage that typifies ischemic kidneys using Notch3 knockout mice and mice with short-term activated Notch3 signaling (N3ICD) in renal epithelial cells. After ischemia/reperfusion, N3ICD mice showed exacerbated infiltration of inflammatory cells and severe tubular damage compared to control mice. Inversely, Notch3 knockout mice were protected against ischemia/reperfusion injury. Renal macrophages derived from Notch3 knockout mice failed to activate proinflammatory cytokines. Chromatin immunoprecipitation analysis of the Notch3 promoter identified NF-κB as the principal inducer of Notch3 in ischemia/reperfusion. Thus, Notch3 induced by NF-κB in the injured epithelium sustains a proinflammatory environment attracting activated macrophages to the site of injury leading to a rapid deterioration of renal function and structure. Hence, targeting Notch3 may provide a novel therapeutic strategy against ischemia/reperfusion and acute kidney injury by preservation of epithelial structure and disruption of proinflammatory signaling.


Subject(s)
Acute Kidney Injury/pathology , Kidney Tubules/pathology , Receptor, Notch3/metabolism , Reperfusion Injury/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/immunology , Animals , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Epithelium/pathology , Humans , Kidney Tubules/immunology , Kidney Tubules/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Receptor, Notch3/genetics
11.
Cell Mol Life Sci ; 74(23): 4315-4320, 2017 12.
Article in English | MEDLINE | ID: mdl-28884334

ABSTRACT

Chronic kidney disease is an incurable to date pathology, with renal replacement therapy through dialysis or transplantation being the only available option for end-stage patients. A deeper understanding of the molecular mechanisms governing the progression of kidney diseases will permit the identification of unknown mediators and potential novel markers or targets of therapy which promise more efficient diagnostic and therapeutic applications. Over the last years, periostin was established by several studies as a novel key player in the progression of renal disease. Periostin is de novo expressed focally by the injured kidney cells during the development of renal disease. In diverse cohorts of renal disease patients, the expression levels of periostin in the kidney and urine were highly correlated with the stage of the pathology and the decline of renal function. Subsequent studies in animal models demonstrated that periostin is centrally involved in mediating renal inflammation and fibrosis, contributing to the deterioration of renal structure and function. Genetic or pharmaco-genetic inhibition of periostin in animal models of renal disease was efficient in arresting the progression of the pathology. This review will summarize the recent advances on periostin in the field of kidney diseases and will discuss its utility of as a novel target of therapy for chronic kidney disease.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Molecular Targeted Therapy/methods , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/genetics , Animals , Antibodies, Monoclonal/therapeutic use , Cell Adhesion Molecules/immunology , Collagen Type I/genetics , Collagen Type I/immunology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Disease Progression , Fibrosis , Gene Expression Regulation , Humans , Kidney/drug effects , Kidney/immunology , Kidney/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Signal Transduction
12.
J Am Soc Nephrol ; 28(10): 2915-2930, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667079

ABSTRACT

GN refers to a variety of renal pathologies that often progress to ESRD, but the molecular mechanisms underlying this progression remain incompletely characterized. Here, we determined whether dysregulated expression of the gap junction protein connexin 43, which has been observed in the progression of renal disease, contributes to GN progression. Immunostaining revealed de novo expression of connexin 43 in damaged glomeruli in patients with glomerular diseases as well as in mice after induction of experimental GN. Notably, 2 weeks after the induction of GN with nephrotoxic serum, mice with a heterozygous deletion of the connexin 43 gene (connexin 43+/-) had proteinuria, BUN, and serum creatinine levels significantly lower than those of wild-type animals. Additionally, the connexin 43+/- mice showed less crescent formation, tubular dilation, monocyte infiltration, and interstitial renal fibrosis. Treatment of cultured podocytes with connexin 43-specific blocking peptides attenuated TGF-ß-induced cytoskeletal and morphologic changes and apoptosis as did treatment with the purinergic blocker suramin. Finally, therapeutic treatment of GN mice with connexin 43-specific antisense oligodeoxynucleotide improved functional and structural renal parameters. These findings suggest that crosstalk between connexin 43 and purinergic signaling contributes to podocyte damage in GN. Given that this protein is highly induced in individuals with glomerular diseases, connexin 43 may be a novel target for therapeutic treatment of GN.


Subject(s)
Connexin 43/metabolism , Glomerulonephritis/metabolism , Animals , Apoptosis , Cell Dedifferentiation , Cell Line , Disease Models, Animal , Disease Progression , Female , Fibrosis , Glomerulonephritis/pathology , Humans , Kidney/pathology , Mice , Podocytes/metabolism , Receptors, Purinergic/metabolism
13.
Front Med (Lausanne) ; 4: 52, 2017.
Article in English | MEDLINE | ID: mdl-28536691

ABSTRACT

Chronic kidney disease (CKD) can be a life-threatening condition, which eventually requires renal replacement therapy through dialysis or transplantation. A lot of effort and resources have been invested the last years in the identification of novel markers of progression and targets for therapy, in order to achieve a more efficient prognosis, diagnosis, and treatment of renal diseases. Using experimental models of renal disease, we identified and studied two promising candidates: periostin, a matricellular protein with high expression in bone and dental tissues, and discoidin domain receptor 1 (DDR1), a transmembrane collagen receptor of the tyrosine kinase family. Both proteins are inactive in physiological conditions, while they are highly upregulated during development of renal disease and are primarily expressed at the sites of injury. Further studies demonstrated that both periostin and DDR1 are involved in the regulation of inflammation and fibrosis, two major processes implicated in the development of renal disease. Targeting of either protein by genetic deletion or pharmacogenetic inhibition via antisense oligonucleotides highly attenuates renal damage and preserves renal structure and function in several animal models. The scope of this review is to summarize the existing evidence supporting the role of periostin and DDR1 as novel biomarkers and therapeutic targets in CKD.

14.
J Am Soc Nephrol ; 28(5): 1475-1490, 2017 May.
Article in English | MEDLINE | ID: mdl-27920156

ABSTRACT

De novo expression in the kidney of periostin, a protein involved in odontogenesis and osteogenesis, has been suggested as a biomarker of renal disease. In this study, we investigated the mechanism(s) of induction and the role of periostin in renal disease. Using a combination of bioinformatics, reporter assay, and chromatin immunoprecipitation analyses, we found that NFκB and other proinflammatory transcription factors induce periostin expression in vitro and that binding of these factors on the periostin promoter is enriched in glomeruli during experimental GN. Mice lacking expression of periostin displayed preserved renal function and structure during GN. Furthermore, delayed administration of periostin antisense oligonucleotides in wild-type animals with GN reversed already established proteinuria, diminished tissue inflammation, and improved renal structure. Lack of periostin expression also blunted the de novo renal expression of integrin-ß3 and phosphorylation of focal adhesion kinase and AKT, known mediators of integrin-ß3 signaling that affect cell motility and survival, observed during GN in wild-type animals. In vitro, recombinant periostin increased the expression of integrin-ß3 and the concomitant phosphorylation of focal adhesion kinase and AKT in podocytes. Notably, periostin and integrin-ß3 were highly colocalized in biopsy specimens from patients with inflammatory GN. These results demonstrate that interplay between periostin and renal inflammation orchestrates inflammatory and fibrotic responses, driving podocyte damage through downstream activation of integrin-ß3 signaling. Targeting periostin may be a novel therapeutic strategy for treating CKD.


Subject(s)
Cell Adhesion Molecules/physiology , Integrin beta3/physiology , Kidney Diseases/etiology , NF-kappa B/physiology , Animals , Female , Glomerulonephritis/complications , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
15.
Biochim Biophys Acta ; 1862(9): 1774-85, 2016 09.
Article in English | MEDLINE | ID: mdl-27345266

ABSTRACT

BACKGROUND: Renal fibrosis is a common histological finding present in many pathologies; however, key signaling pathways and molecular determinants involved in the development of fibrosis are not fully known yet. Previous findings have established a causative role of calreticulin's up-regulation during the development of renal fibrosis while its down-regulation exhibited a protective effect against fibrosis. Therefore, the mechanism of its up-regulation needs to be explored. METHODS: Bioinformatics analyses of the calreticulin gene promoter combined with transcriptional assays and in vivo chromatin immunoprecipitation experiments in the Unilateral Ureteric Obstruction (UUO) model of renal fibrosis, indicated that NR5A2 is a critical regulator of calreticulin expression. To confirm this finding, and further study post-translational modifications of NR5A2, real time RT-qPCR, immunohistochemistry and Western blotting experiments were performed. RESULTS: NR5A2 is up-regulated at both mRNA and protein level during kidney fibrosis in the UUO model. The post-translational modification of SUMOylation was identified as a critical parameter in this phenomenon and SUMOylation was observed to be up-regulated during the development of renal fibrosis. The enzyme Ubc9, critical for the process of SUMOylation was also upregulated at mRNA and protein level during the process. CONCLUSION: These data establish for the first time a role for NR5A2 and its SUMOylation on the transcriptional regulation of the calreticulin gene in a rodent model of renal fibrosis and raise the possibility that NR5A2 might be a novel target for future anti-fibrotic interventions.


Subject(s)
Calreticulin/genetics , Kidney/metabolism , Kidney/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Line , Disease Models, Animal , Fibrosis , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , SUMO-1 Protein/metabolism , Sumoylation , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Up-Regulation , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
16.
Am J Pathol ; 183(5): 1474-1487, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035512

ABSTRACT

Renal fibrosis is the common anatomical feature underlying the progression of chronic kidney disease, a leading cause of morbidity and mortality worldwide. In a previous study, we demonstrated that during development of renal fibrosis in a rat model of unilateral ureteric obstruction, calreticulin (CRT) is up-regulated in tubular epithelial cells (TECs). In the present study, we used in vitro and in vivo approaches to examine the role of CRT in TECs and its contribution to the progression of fibrosis. In cultured renal TECs, CRT overexpression induced acquisition of an altered, profibrotic cellular phenotype. Consistently, the opposite effects were observed for CRT knockdown. Subsequently, we confirmed that critical changes observed in vitro were also apparent in tubular cells in vivo in the animal model of unilateral ureteric obstruction. In agreement with these results, we demonstrate that substantial (50%) reduction in the expression of CRT reduced the development of tubulointerstitial fibrosis at a comparable level through regulation of inflammation, transcriptional activation, transforming growth factor ß1-associated effects, and apoptosis. In summary, our findings establish that CRT is critically involved in the molecular mechanisms that drive renal fibrosis progression and indicate that inhibition of CRT expression might be a therapeutic target for reduction of fibrosis and chronic kidney disease development.


Subject(s)
Calreticulin/metabolism , Epithelial Cells/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Up-Regulation , Animals , Apoptosis , Biomarkers/metabolism , Cell Line , Cell Movement , Cell Proliferation , Collagen/metabolism , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum Stress , Epithelial Cells/pathology , Fibrosis , Gene Knockdown Techniques , Heterozygote , Humans , Male , Mesoderm/metabolism , Mesoderm/pathology , Mice , Rats , Signal Transduction , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
17.
PLoS One ; 8(6): e66887, 2013.
Article in English | MEDLINE | ID: mdl-23840546

ABSTRACT

Fibrosis is a complex and multifactorial process, affecting the structure and compromising the function of several organs. Among those, renal fibrosis is an important pathological change, eventually leading to renal failure. Proteomic analysis of the renal parenchyma in the well-established rat model of unilateral ureteral obstruction (UUO model) suggested that transgelin was up-regulated during the development of fibrosis. Transgelin up-regulation was confirmed both at the protein and at the mRNA level. It was observed that at early stages of fibrosis transgelin was mainly expressed in the interstitial compartment and, more specifically, in cells surrounding the glomeruli. Subsequently, it was confirmed that transgelin expressing cells were activated fibroblasts, based on their extensive co-expression of α-SMA and their complete lack of co-distribution with markers of other cell types (endothelial, epithelial and cells of the immune system). These periglomerular fibroblasts exhibited staining for transgelin mainly cytoplasmic but occasionally nuclear as well. In addition, transgelin expression in periglomerular fibroblasts was absent in renal fibrosis developed in a hypertensive model, compared to the UUO model. Promoter analysis indicated that there are several conserved motifs for transcription factor binding. Among those, Kruppel-like factor 6 was found to be up-regulated in transgelin positive periglomerular activated fibroblasts, suggesting a possible involvement in the mechanism of transgelin up-regulation. These data strongly suggest that transgelin is up-regulated in the obstructive nephropathy and could be used as a novel marker for renal fibrosis in the future.


Subject(s)
Kidney Diseases/complications , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Up-Regulation , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Animals , Biomarkers/metabolism , Fibrosis , Intracellular Space/metabolism , Kidney/pathology , Male , Promoter Regions, Genetic/genetics , Protein Transport , Rats , Rats, Wistar , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
18.
Proteomics ; 11(4): 805-15, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21229585

ABSTRACT

Chronic kidney disease (CKD) is the end-point of a number of renal and systemic diseases. The high incidence and financial burden of CKD makes it imperative to diagnose CKD at early stages when therapeutic interventions are far more effective. A key component of CKD is the development of renal fibrosis. Renal fibrosis is a complex process, associated with many cell types and pathways, resulting in structural and functional alterations. Identification of specific biomarkers of renal fibrosis may thus not only help us to understand the pathophysiological mechanisms involved in this process, but also improve diagnosis in the clinic. In this review, the existing literature on proteomic approaches to study renal fibrosis is presented and evaluated. The importance of using animal models along with patient material is discussed and future directions, considered key to this field, are proposed.


Subject(s)
Disease Models, Animal , Kidney Diseases/metabolism , Kidney/pathology , Proteomics , Animals , Biomarkers/metabolism , Chronic Disease , Fibrosis , Humans , Kidney/metabolism , Kidney Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...