Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Sci Rep ; 13(1): 16919, 2023 10 07.
Article En | MEDLINE | ID: mdl-37805649

Type 2 diabetes (T2D) and its complications can have debilitating, sometimes fatal consequences for afflicted individuals. The disease can be difficult to control, and therapeutic strategies to prevent T2D-induced tissue and organ damage are needed. Here we describe the results of administering a potent and selective inhibitor of Protein Kinase C (PKC) family members PKCα and PKCß, Cmpd 1, in the ZSF1 obese rat model of hyperphagia-induced, obesity-driven T2D. Although our initial intent was to evaluate the effect of PKCα/ß inhibition on renal damage in this model setting, Cmpd 1 unexpectedly caused a marked reduction in the hyperphagic response of ZSF1 obese animals. This halted renal function decline but did so indirectly and indistinguishably from a pair feeding comparator group. However, above and beyond this food intake effect, Cmpd 1 lowered overall animal body weights, reduced liver vacuolation, and reduced inguinal adipose tissue (iWAT) mass, inflammation, and adipocyte size. Taken together, Cmpd 1 had strong effects on multiple disease parameters in this obesity-driven rodent model of T2D. Further evaluation for potential translation of PKCα/ß inhibition to T2D and obesity in humans is warranted.


Adiposity , Diabetes Mellitus, Type 2 , Humans , Rats , Animals , Adiposity/physiology , Protein Kinase C-alpha , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Obesity/complications , Obesity/drug therapy , Hyperphagia/complications , Hyperphagia/drug therapy , Kidney/physiology
2.
Nat Commun ; 14(1): 6493, 2023 10 14.
Article En | MEDLINE | ID: mdl-37838725

Misfolded protein aggregates may cause toxic proteinopathy, including autosomal dominant tubulointerstitial kidney disease due to uromodulin mutations (ADTKD-UMOD), a leading hereditary kidney disease. There are no targeted therapies. In our generated mouse model recapitulating human ADTKD-UMOD carrying a leading UMOD mutation, we show that autophagy/mitophagy and mitochondrial biogenesis are impaired, leading to cGAS-STING activation and tubular injury. Moreover, we demonstrate that inducible tubular overexpression of mesencephalic astrocyte-derived neurotrophic factor (MANF), a secreted endoplasmic reticulum protein, after the onset of disease stimulates autophagy/mitophagy, clears mutant UMOD, and promotes mitochondrial biogenesis through p-AMPK enhancement, thus protecting kidney function in our ADTKD mouse model. Conversely, genetic ablation of MANF in the mutant thick ascending limb tubular cells worsens autophagy suppression and kidney fibrosis. Together, we have discovered MANF as a biotherapeutic protein and elucidated previously unknown mechanisms of MANF in the regulation of organelle homeostasis, which may have broad therapeutic applications to treat various proteinopathies.


Polycystic Kidney Diseases , Humans , Mice , Animals , Autophagy/genetics , Homeostasis , Fibrosis , Nerve Growth Factors/genetics
3.
Sci Rep ; 13(1): 8922, 2023 06 01.
Article En | MEDLINE | ID: mdl-37264117

The intestinal epithelium comprises diverse cell types and executes many specialized functions as the primary interface between luminal contents and internal organs. A key function provided by the epithelium is maintenance of a barrier that protects the individual from pathogens, irritating luminal contents, and the microbiota. Disruption of this barrier can lead to inflammatory disease within the intestinal mucosa, and, in more severe cases, to sepsis. Animal models to study intestinal permeability are costly and not entirely predictive of human biology. Here we present a model of human colon barrier function that integrates primary human colon stem cells into Draper's PREDICT96 microfluidic organ-on-chip platform to yield a high-throughput system appropriate to predict damage and healing of the human colon epithelial barrier. We have demonstrated pharmacologically induced barrier damage measured by both a high throughput molecular permeability assay and transepithelial resistance. Using these assays, we developed an Inflammatory Bowel Disease-relevant model through cytokine induced damage that can support studies of disease mechanisms and putative therapeutics.


Colon , Inflammatory Bowel Diseases , Animals , Humans , Disease Models, Animal , Colon/metabolism , Inflammatory Bowel Diseases/metabolism , Intestines , Intestinal Mucosa/metabolism , Permeability
4.
bioRxiv ; 2023 Jan 12.
Article En | MEDLINE | ID: mdl-36711449

Misfolded protein aggregates may cause toxic proteinopathy, including autosomal dominant tubulointerstitial kidney disease due to uromodulin mutations (ADTKD- UMOD ), one of the leading hereditary kidney diseases, and Alzheimer’s disease etc. There are no targeted therapies. ADTKD is also a genetic form of renal fibrosis and chronic kidney disease, which affects 500 million people worldwide. For the first time, in our newly generated mouse model recapitulating human ADTKD- UMOD carrying a leading UMOD deletion mutation, we show that autophagy/mitophagy and mitochondrial biogenesis are severely impaired, leading to cGAS- STING activation and tubular injury. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a novel endoplasmic reticulum stress-regulated secreted protein. We provide the first study that inducible tubular overexpression of MANF after the onset of disease stimulates autophagy/mitophagy and clearance of the misfolded UMOD, and promotes mitochondrial biogenesis through p-AMPK enhancement, resulting in protection of kidney function. Conversely, genetic ablation of endogenous MANF upregulated in the mutant mouse and human tubular cells worsens autophagy suppression and kidney fibrosis. Together, we discover MANF as a novel biotherapeutic protein and elucidate previously unknown mechanisms of MANF in regulating organelle homeostasis to treat ADTKD, which may have broad therapeutic application to treat various proteinopathies.

5.
Inflamm Bowel Dis ; 29(2): 195-206, 2023 02 01.
Article En | MEDLINE | ID: mdl-36356046

BACKGROUND: Patient-derived organoid (PDO) models offer potential to transform drug discovery for inflammatory bowel disease (IBD) but are limited by inconsistencies with differentiation and functional characterization. We profiled molecular and cellular features across a range of intestinal organoid models and examined differentiation and establishment of a functional epithelial barrier. METHODS: Patient-derived organoids or monolayers were generated from control or IBD patient-derived colon or ileum and were molecularly or functionally profiled. Biological or technical replicates were examined for transcriptional responses under conditions of expansion or differentiation. Cell-type composition was determined by deconvolution of cell-associated gene signatures and histological features. Differentiated control or IBD-derived monolayers were examined for establishment of transepithelial electrical resistance (TEER), loss of barrier integrity in response to a cocktail of interferon (IFN)-γ and tumor necrosis factor (TNF)-α, and prevention of cytokine-induced barrier disruption by the JAK inhibitor, tofacitinib. RESULTS: In response to differentiation media, intestinal organoids and monolayers displayed gene expression patterns consistent with maturation of epithelial cell types found in the human gut. Upon differentiation, both colon- and ileum-derived monolayers formed functional barriers, with sustained TEER. Barrier integrity was compromised by inflammatory cytokines IFN-γ and TNF-α, and damage was inhibited in a dose-dependent manner by tofacitinib. CONCLUSIONS: We describe the generation and characterization of human colonic or ileal organoid models capable of functional differentiation to mature epithelial cell types. In monolayer culture, these cells formed a robust epithelial barrier with sustained TEER and responses to pharmacological modulation. Our findings demonstrate that control and IBD patient-derived organoids possess consistent transcriptional and functional profiles that can enable development of epithelial-targeted therapies.


Inflammatory Bowel Diseases , Intestines , Organoids , Humans , Cytokines/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Organoids/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Intestines/physiology
6.
Am J Physiol Renal Physiol ; 321(2): F236-F244, 2021 08 01.
Article En | MEDLINE | ID: mdl-34251273

Autosomal dominant tubulointerstitial kidney disease (ADTKD)-uromodulin (UMOD) is the most common nonpolycystic genetic kidney disease, but it remains unrecognized due to its clinical heterogeneity and lack of screening test. Moreover, the fact that the clinical feature is a poor predictor of disease outcome further highlights the need for the development of mechanistic biomarkers in ADTKD. However, low abundant urinary proteins secreted by thick ascending limb cells, where UMOD is synthesized, have posed a challenge for the detection of biomarkers in ADTKD-UMOD. In the CRISPR/Cas9-generated murine model and patients with ADTKD-UMOD, we found that immunoglobulin heavy chain-binding protein (BiP), an endoplasmic reticulum chaperone, was exclusively upregulated by mutant UMOD in the thick ascending limb and easily detected by Western blot analysis in the urine at an early stage of disease. However, even the most sensitive ELISA failed to detect urinary BiP in affected individuals. We therefore developed an ultrasensitive, plasmon-enhanced fluorescence-linked immunosorbent assay (p-FLISA) to quantify urinary BiP concentration by harnessing the newly invented ultrabright fluorescent nanoconstruct, termed "plasmonic Fluor." p-FLISA demonstrated that urinary BiP excretion was significantly elevated in patients with ADTKD-UMOD compared with unaffected controls, which may have potential utility in risk stratification, disease activity monitoring, disease progression prediction, and guidance of endoplasmic reticulum-targeted therapies in ADTKD.NEW & NOTEWORTHY Autosomal dominant tubulointerstitial kidney disease (ADTKD)-uromodulin (UMOD) is an underdiagnosed cause of chronic kidney disease (CKD). Lack of ultrasensitive bioanalytical tools has hindered the discovery of low abundant urinary biomarkers in ADTKD. Here, we developed an ultrasensitive plasmon-enhanced fluorescence-linked immunosorbent assay (p-FLISA). p-FLISA demonstrated that secreted immunoglobulin heavy chain-binding protein is an early urinary endoplasmic reticulum stress biomarker in ADTKD-UMOD, which will be valuable in monitoring disease progression and the treatment response in ADTKD.


Biomarkers/urine , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/urine , Immunosorbent Techniques , Nephritis, Interstitial/urine , Animals , Endoplasmic Reticulum Chaperone BiP , Humans , Mice , Nephritis, Interstitial/genetics , Uromodulin/genetics
7.
J Am Soc Nephrol ; 29(6): 1690-1705, 2018 06.
Article En | MEDLINE | ID: mdl-29739813

Background Kidney injury is characterized by persisting inflammation and fibrosis, yet mechanisms by which inflammatory signals drive fibrogenesis remain poorly defined.Methods RNA sequencing of fibrotic kidneys from patients with CKD identified a metabolic gene signature comprising loss of mitochondrial and oxidative phosphorylation gene expression with a concomitant increase in regulators and enzymes of glycolysis under the control of PGC1α and MYC transcription factors, respectively. We modeled this metabolic switch in vivo, in experimental murine models of kidney injury, and in vitro in human kidney stromal cells (SCs) and human kidney organoids.Results In mice, MYC and the target genes thereof became activated in resident SCs early after kidney injury, suggesting that acute innate immune signals regulate this transcriptional switch. In vitro, stimulation of purified human kidney SCs and human kidney organoids with IL-1ß recapitulated the molecular events observed in vivo, inducing functional metabolic derangement characterized by increased MYC-dependent glycolysis, the latter proving necessary to drive proliferation and matrix production. MYC interacted directly with sequestosome 1/p62, which is involved in proteasomal degradation, and modulation of p62 expression caused inverse effects on MYC expression. IL-1ß stimulated autophagy flux, causing degradation of p62 and accumulation of MYC. Inhibition of the IL-1R signal transducer kinase IRAK4 in vivo or inhibition of MYC in vivo as well as in human kidney organoids in vitro abrogated fibrosis and reduced tubular injury.Conclusions Our findings define a connection between IL-1ß and metabolic switch in fibrosis initiation and progression and highlight IL-1ß and MYC as potential therapeutic targets in tubulointerstitial diseases.


Acute Kidney Injury/pathology , Interleukin-1beta/pharmacology , Kidney/cytology , Kidney/pathology , Proto-Oncogene Proteins c-myc/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Acute Kidney Injury/metabolism , Animals , Autophagy/drug effects , Azepines/pharmacology , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Progression , Extracellular Matrix/metabolism , Fibrosis , Glycolysis/drug effects , Humans , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Kidney Tubules, Proximal/pathology , Male , Membrane Proteins/metabolism , Mice , Organoids , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Stromal Cells/metabolism , Thyroid Hormones/metabolism , Triazoles/pharmacology , Thyroid Hormone-Binding Proteins
8.
J Clin Invest ; 127(11): 3954-3969, 2017 Nov 01.
Article En | MEDLINE | ID: mdl-28990932

Uromodulin-associated kidney disease (UAKD) is caused by mutations in the uromodulin (UMOD) gene that result in a misfolded form of UMOD protein, which is normally secreted by nephrons. In UAKD patients, mutant UMOD is poorly secreted and accumulates in the ER of distal kidney epithelium, but its role in disease progression is largely unknown. Here, we modeled UMOD accumulation in mice by expressing the murine equivalent of the human UMOD p.Cys148Trp point mutation (UmodC147W/+ mice). Like affected humans, these UmodC147W/+ mice developed spontaneous and progressive kidney disease with organ failure over 24 weeks. Analysis of diseased kidneys and purified UMOD-producing cells revealed early activation of the PKR-like ER kinase/activating transcription factor 4 (PERK/ATF4) ER stress pathway, innate immune mediators, and increased apoptotic signaling, including caspase-3 activation. Unexpectedly, we also detected autophagy deficiency. Human cells expressing UMOD p.Cys147Trp recapitulated the findings in UmodC147W/+ mice, and autophagy activation with mTOR inhibitors stimulated the intracellular removal of aggregated mutant UMOD. Human cells producing mutant UMOD were susceptible to TNF-α- and TRAIL-mediated apoptosis due to increased expression of the ER stress mediator tribbles-3. Blocking TNF-α in vivo with the soluble recombinant fusion protein TNFR:Fc slowed disease progression in UmodC147W/+ mice by reducing active caspase-3, thereby preventing tubule cell death and loss of epithelial function. These findings reveal a targetable mechanism for disease processes involved in UAKD.


Apoptosis , Endoplasmic Reticulum Stress , Renal Insufficiency/genetics , Uromodulin/genetics , Animals , Autophagy , Base Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Disease Progression , Female , Genetic Association Studies , Humans , Immunity, Innate , Male , Mice, Transgenic , Mutation, Missense , Point Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Renal Insufficiency/immunology , Renal Insufficiency/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uromodulin/metabolism
9.
Biomaterials ; 141: 314-329, 2017 Oct.
Article En | MEDLINE | ID: mdl-28711779

Loss of the microvascular (MV) network results in tissue ischemia, loss of tissue function, and is a hallmark of chronic diseases. The incorporation of a functional vascular network with that of the host remains a challenge to utilizing engineered tissues in clinically relevant therapies. We showed that vascular-bed-specific endothelial cells (ECs) exhibit differing angiogenic capacities, with kidney microvascular endothelial cells (MVECs) being the most deficient, and sought to explore the underlying mechanism. Constitutive activation of the phosphatase PTEN in kidney MVECs resulted in impaired PI3K/AKT activity in response to vascular endothelial growth factor (VEGF). Suppression of PTEN in vivo resulted in microvascular regeneration, but was insufficient to improve tissue function. Promoter analysis of the differentially regulated genes in KMVECs suggests that the transcription factor FOXO1 is highly active and RNAseq analysis revealed that hyperactive FOXO1 inhibits VEGF-Notch-dependent tip-cell formation by direct and indirect inhibition of DLL4 expression in response to VEGF. Inhibition of FOXO1 enhanced angiogenesis in human bio-engineered capillaries, and resulted in microvascular regeneration and improved function in mouse models of injury-repair.


Forkhead Box Protein O1/metabolism , Kidney/blood supply , Kidney/physiopathology , Microvessels/physiopathology , Neovascularization, Physiologic , Adult , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Kidney/injuries , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
PLoS One ; 12(7): e0181861, 2017.
Article En | MEDLINE | ID: mdl-28746409

ZSF1 rats exhibit spontaneous nephropathy secondary to obesity, hypertension, and diabetes, and have gained interest as a model system with potentially high translational value to progressive human disease. To thoroughly characterize this model, and to better understand how closely it recapitulates human disease, we performed a high resolution longitudinal analysis of renal disease progression in ZSF1 rats spanning from early disease to end stage renal disease. Analyses included metabolic endpoints, renal histology and ultrastructure, evaluation of a urinary biomarker of fibrosis, and transcriptome analysis of glomerular-enriched tissue over the course of disease. Our findings support the translational value of the ZSF1 rat model, and are provided here to assist researchers in the determination of the model's suitability for testing a particular mechanism of interest, the design of therapeutic intervention studies, and the identification of new targets and biomarkers for type 2 diabetic nephropathy.


Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/genetics , Kidney Failure, Chronic/complications , Kidney/metabolism , Animals , Cluster Analysis , Collagen/genetics , Collagen/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Immunohistochemistry , Kidney/pathology , Kidney/ultrastructure , Kidney Failure, Chronic/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Microscopy, Electron, Transmission , Obesity/complications , Rats , Reverse Transcriptase Polymerase Chain Reaction
11.
J Am Soc Nephrol ; 28(6): 1769-1782, 2017 Jun.
Article En | MEDLINE | ID: mdl-28130402

Connective tissue growth factor (CTGF), a matrix-associated protein with four distinct cytokine binding domains, has roles in vasculogenesis, wound healing responses, and fibrogenesis and is upregulated in fibroblasts and myofibroblasts in disease. Here, we investigated the role of CTGF in fibrogenic cells. In mice, tissue-specific inducible overexpression of CTGF by kidney pericytes and fibroblasts had no bearing on nephrogenesis or kidney homeostasis but exacerbated inflammation and fibrosis after ureteral obstruction. These effects required the WNT receptor LDL receptor-related protein 6 (LRP6). Additionally, pericytes isolated from these mice became hypermigratory and hyperproliferative on overexpression of CTGF. CTGF is cleaved in vivo into distinct domains. Treatment with recombinant domain 1, 1+2 (N terminus), or 4 (C terminus) independently activated myofibroblast differentiation and wound healing responses in cultured pericytes, but domain 4 showed the broadest profibrotic activity. Domain 4 exhibited low-affinity binding to LRP6 in in vitro binding assays, and inhibition of LRP6 or critical signaling cascades downstream of LRP6, including JNK and WNT/ß-catenin, inhibited the biologic activity of domain 4. Administration of blocking antibodies specifically against CTGF domain 4 or recombinant Dickkopf-related protein-1, an endogenous inhibitor of LRP6, effectively inhibited inflammation and fibrosis associated with ureteral obstruction in vivo Therefore, domain 4 of CTGF and the WNT signaling pathway are important new targets in fibrosis.


Connective Tissue Growth Factor/physiology , Kidney Diseases/etiology , Kidney/pathology , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Animals , Connective Tissue Growth Factor/antagonists & inhibitors , Fibroblasts , Fibrosis/etiology , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pericytes
12.
J Clin Invest ; 127(1): 321-334, 2017 01 03.
Article En | MEDLINE | ID: mdl-27869651

Fibrotic disease is associated with matrix deposition that results in the loss of organ function. Pericytes, the precursors of myofibroblasts, are a source of pathological matrix collagens and may be promising targets for treating fibrogenesis. Here, we have shown that pericytes activate a TLR2/4- and MyD88-dependent proinflammatory program in response to tissue injury. Similarly to classic immune cells, pericytes activate the NLRP3 inflammasome, leading to IL-1ß and IL-18 secretion. Released IL-1ß signals through pericyte MyD88 to amplify this response. Unexpectedly, we found that MyD88 and its downstream effector kinase IRAK4 intrinsically control pericyte migration and conversion to myofibroblasts. Specific ablation of MyD88 in pericytes or pharmacological inhibition of MyD88 signaling by an IRAK4 inhibitor in vivo protected against kidney injury by profoundly attenuating tissue injury, activation, and differentiation of myofibroblasts. Our data show that in pericytes, MyD88 and IRAK4 are key regulators of 2 major injury responses: inflammatory and fibrogenic. Moreover, these findings suggest that disruption of this MyD88-dependent pathway in pericytes might be a potential therapeutic approach to inhibit fibrogenesis and promote regeneration.


Acute Kidney Injury/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Myeloid Differentiation Factor 88/metabolism , Pericytes/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Cells, Cultured , Fibrosis , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pericytes/pathology , Signal Transduction/genetics
13.
JCI Insight ; 1(20): e87446, 2016 12 08.
Article En | MEDLINE | ID: mdl-27942582

Pentraxin-2 (PTX-2), also known as serum amyloid P component (SAP/APCS), is a constitutive, antiinflammatory, innate immune plasma protein whose circulating level is decreased in chronic human fibrotic diseases. Here we show that recombinant human PTX-2 (rhPTX-2) retards progression of chronic kidney disease in Col4a3 mutant mice with Alport syndrome, reducing blood markers of kidney failure, enhancing lifespan by 20%, and improving histological signs of disease. Exogenously delivered rhPTX-2 was detected in macrophages but also in tubular epithelial cells, where it counteracted macrophage activation and was cytoprotective for the epithelium. Computational analysis of genes regulated by rhPTX-2 identified the transcriptional regulator c-Jun along with its activator protein-1 (AP-1) binding partners as a central target for the function of rhPTX-2. Accordingly, PTX-2 attenuates c-Jun and AP-1 activity, and reduces expression of AP-1-dependent inflammatory genes in both monocytes and epithelium. Our studies therefore identify rhPTX-2 as a potential therapy for chronic fibrotic disease of the kidney and an important inhibitor of pathological c-Jun signaling in this setting.


C-Reactive Protein/pharmacology , Kidney/pathology , Nephritis, Hereditary/therapy , Nerve Tissue Proteins/pharmacology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Signal Transduction , Transcription Factor AP-1/antagonists & inhibitors , Animals , Cells, Cultured , Fibrosis , Humans , Macrophage Activation , Mice , Mice, 129 Strain , Mice, Knockout , Monocytes , Nephritis, Hereditary/pathology , Recombinant Proteins/pharmacology
14.
J Am Soc Nephrol ; 27(12): 3639-3652, 2016 Dec.
Article En | MEDLINE | ID: mdl-27026366

The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.


Kidney Diseases/etiology , Kidney/pathology , Myofibroblasts/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction , Tumor Necrosis Factors/physiology , Animals , Cytokine TWEAK , Disease Progression , Fibrosis/etiology , Mice , TWEAK Receptor
15.
J Clin Invest ; 125(1): 141-56, 2015 Jan.
Article En | MEDLINE | ID: mdl-25415439

MicroRNA-21 (miR-21) contributes to the pathogenesis of fibrogenic diseases in multiple organs, including the kidneys, potentially by silencing metabolic pathways that are critical for cellular ATP generation, ROS production, and inflammatory signaling. Here, we developed highly specific oligonucleotides that distribute to the kidney and inhibit miR-21 function when administered subcutaneously and evaluated the therapeutic potential of these anti-miR-21 oligonucleotides in chronic kidney disease. In a murine model of Alport nephropathy, miR-21 silencing did not produce any adverse effects and resulted in substantially milder kidney disease, with minimal albuminuria and dysfunction, compared with vehicle-treated mice. miR-21 silencing dramatically improved survival of Alport mice and reduced histological end points, including glomerulosclerosis, interstitial fibrosis, tubular injury, and inflammation. Anti-miR-21 enhanced PPARα/retinoid X receptor (PPARα/RXR) activity and downstream signaling pathways in glomerular, tubular, and interstitial cells. Moreover, miR-21 silencing enhanced mitochondrial function, which reduced mitochondrial ROS production and thus preserved tubular functions. Inhibition of miR-21 was protective against TGF-ß-induced fibrogenesis and inflammation in glomerular and interstitial cells, likely as the result of enhanced PPARα/RXR activity and improved mitochondrial function. Together, these results demonstrate that inhibition of miR-21 represents a potential therapeutic strategy for chronic kidney diseases including Alport nephropathy.


MicroRNAs/genetics , Nephritis, Hereditary/therapy , Oligoribonucleotides, Antisense/genetics , Animals , Autoantigens/genetics , Collagen Type IV/deficiency , Collagen Type IV/genetics , Disease Progression , Fibrosis/metabolism , Kidney/metabolism , Kidney/pathology , Metabolic Networks and Pathways/genetics , Mice, 129 Strain , MicroRNAs/metabolism , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , Reactive Oxygen Species/metabolism , Transcriptome , Up-Regulation
16.
Proc Natl Acad Sci U S A ; 110(4): 1440-5, 2013 Jan 22.
Article En | MEDLINE | ID: mdl-23302695

Fibrosis of vital organs is a major public health problem with limited therapeutic options. Mesenchymal cells including microvascular mural cells (pericytes) are major progenitors of scar-forming myofibroblasts in kidney and other organs. Here we show pericytes in healthy kidneys have active WNT/ß-catenin signaling responses that are markedly up-regulated following kidney injury. Dickkopf-related protein 1 (DKK-1), a ligand for the WNT coreceptors low-density lipoprotein receptor-related proteins 5 and 6 (LRP-5 and LRP-6) and an inhibitor of WNT/ß-catenin signaling, effectively inhibits pericyte activation, detachment, and transition to myofibroblasts in vivo in response to kidney injury, resulting in attenuated fibrogenesis, capillary rarefaction, and inflammation. DKK-1 blocks activation and proliferation of established myofibroblasts in vitro and blocks pericyte proliferation to PDGF, pericyte migration, gene activation, and cytoskeletal reorganization to TGF-ß or connective tissue growth factor. These effects are largely independent of inhibition of downstream ß-catenin signaling. DKK-1 acts predominantly by inhibiting PDGF-, TGF-ß-, and connective tissue growth factor-activated MAPK and JNK signaling cascades, acting via LRP-6 with associated WNT ligand. Biochemically, LRP-6 interacts closely with PDGF receptor ß and TGF-ß receptor 1 at the cell membrane, suggesting that it may have roles in pathways other than WNT/ß-catenin. In summary, DKK-1 blocks many of the changes in pericytes required for myofibroblast transition and attenuates established myofibroblast proliferation/activation by mechanisms dependent on LRP-6 and WNT ligands but not the downstream ß-catenin pathway.


Intercellular Signaling Peptides and Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pericytes/metabolism , Pericytes/pathology , Animals , Becaplermin , Cell Proliferation/drug effects , Connective Tissue Growth Factor/pharmacology , Fibrosis , G1 Phase Cell Cycle Checkpoints , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pericytes/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , Transforming Growth Factor beta/pharmacology , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
...