Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Renal Physiol ; 321(2): F236-F244, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34251273

ABSTRACT

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.


Subject(s)
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
2.
Proc Natl Acad Sci U S A ; 114(33): 8758-8763, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28765370

ABSTRACT

The integrity of the endothelial barrier between circulating blood and tissue is important for blood vessel function and, ultimately, for organ homeostasis. Here, we developed a vessel-on-a-chip with perfused endothelialized channels lined with human bone marrow stromal cells, which adopt a mural cell-like phenotype that recapitulates barrier function of the vasculature. In this model, barrier function is compromised upon exposure to inflammatory factors such as LPS, thrombin, and TNFα, as has been observed in vivo. Interestingly, we observed a rapid physical withdrawal of mural cells from the endothelium that was accompanied by an inhibition of endogenous Rac1 activity and increase in RhoA activity in the mural cells themselves upon inflammation. Using a system to chemically induce activity in exogenously expressed Rac1 or RhoA within minutes of stimulation, we demonstrated RhoA activation induced loss of mural cell coverage on the endothelium and reduced endothelial barrier function, and this effect was abrogated when Rac1 was simultaneously activated. We further showed that N-cadherin expression in mural cells plays a key role in barrier function, as CRISPR-mediated knockout of N-cadherin in the mural cells led to loss of barrier function, and overexpression of N-cadherin in CHO cells promoted barrier function. In summary, this bicellular model demonstrates the continuous and rapid modulation of adhesive interactions between endothelial and mural cells and its impact on vascular barrier function and highlights an in vitro platform to study the biology of perivascular-endothelial interactions.


Subject(s)
Cadherins/metabolism , Endothelium, Vascular/metabolism , Endothelium/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Biomimetics/methods , CHO Cells , Cricetulus , Humans , Inflammation/metabolism , Thrombin/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Am J Pathol ; 188(12): 2745-2762, 2018 12.
Article in English | MEDLINE | ID: mdl-30268775

ABSTRACT

Mutations in mitochondrial DNA as well as nuclear-encoded mitochondrial proteins have been reported to cause tubulointerstitial kidney diseases and focal segmental glomerulosclerosis (FSGS). Recently, genes and pathways affecting mitochondrial turnover and permeability have been implicated in adult-onset FSGS. Furthermore, dysfunctioning mitochondria may be capable of engaging intracellular innate immune-sensing pathways. To determine the impact of mitochondrial dysfunction in FSGS and secondary innate immune responses, we generated Cre/loxP transgenic mice to generate a loss-of-function deletion mutation of the complex IV assembly cofactor heme A:farnesyltransferase (COX10) restricted to cells of the developing nephrons. These mice develop severe, early-onset FSGS with innate immune activation and die prematurely with kidney failure. Mutant kidneys showed loss of glomerular and tubular epithelial function, epithelial apoptosis, and, in addition, a marked interferon response. In vitro modeling of Cox10 deletion in primary kidney epithelium compromises oxygen consumption, ATP generation, and induces oxidative stress. In addition, loss of Cox10 triggers a selective interferon response, which may be caused by the leak of mitochondrial DNA into the cytosol activating the intracellular DNA sensor, stimulator of interferon genes. This new animal model provides a mechanism to study mitochondrial dysfunction in vivo and demonstrates a direct link between mitochondrial dysfunction and intracellular innate immune response.


Subject(s)
Alkyl and Aryl Transferases/physiology , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/etiology , Interferon Regulatory Factors/metabolism , Interferons/pharmacology , Membrane Proteins/physiology , Oxidative Stress , Sequence Deletion , Animals , Antiviral Agents/pharmacology , Female , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Interferon Regulatory Factors/genetics , Male , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/pathology
4.
J Am Soc Nephrol ; 29(6): 1690-1705, 2018 06.
Article in English | MEDLINE | ID: mdl-29739813

ABSTRACT

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.


Subject(s)
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
5.
J Am Soc Nephrol ; 28(6): 1769-1782, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28130402

ABSTRACT

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.


Subject(s)
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
6.
J Am Soc Nephrol ; 28(6): 1741-1752, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28062569

ABSTRACT

Ischemia-reperfusion injury (IRI) is a leading cause of AKI. This common clinical complication lacks effective therapies and can lead to the development of CKD. The αvß5 integrin may have an important role in acute injury, including septic shock and acute lung injury. To examine its function in AKI, we utilized a specific function-blocking antibody to inhibit αvß5 in a rat model of renal IRI. Pretreatment with this anti-αvß5 antibody significantly reduced serum creatinine levels, diminished renal damage detected by histopathologic evaluation, and decreased levels of injury biomarkers. Notably, therapeutic treatment with the αvß5 antibody 8 hours after IRI also provided protection from injury. Global gene expression profiling of post-ischemic kidneys showed that αvß5 inhibition affected established injury markers and induced pathway alterations previously shown to be protective. Intravital imaging of post-ischemic kidneys revealed reduced vascular leak with αvß5 antibody treatment. Immunostaining for αvß5 in the kidney detected evident expression in perivascular cells, with negligible expression in the endothelium. Studies in a three-dimensional microfluidics system identified a pericyte-dependent role for αvß5 in modulating vascular leak. Additional studies showed αvß5 functions in the adhesion and migration of kidney pericytes in vitro Initial studies monitoring renal blood flow after IRI did not find significant effects with αvß5 inhibition; however, future studies should explore the contribution of vasomotor effects. These studies identify a role for αvß5 in modulating injury-induced renal vascular leak, possibly through effects on pericyte adhesion and migration, and reveal αvß5 inhibition as a promising therapeutic strategy for AKI.


Subject(s)
Capillary Permeability/drug effects , Kidney/blood supply , Receptors, Vitronectin/antagonists & inhibitors , Reperfusion Injury/prevention & control , Animals , Male , Rats , Rats, Sprague-Dawley
7.
Am J Physiol Renal Physiol ; 312(3): F516-F532, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28052876

ABSTRACT

We have examined the pathogenic role of increased complement expression and activation during kidney fibrosis. Here, we show that PDGFRß-positive pericytes isolated from mice subjected to obstructive or folic acid injury secrete C1q. This was associated with increased production of proinflammatory cytokines, extracellular matrix components, collagens, and increased Wnt3a-mediated activation of Wnt/ß-catenin signaling, which are hallmarks of myofibroblast activation. Real-time PCR, immunoblots, immunohistochemistry, and flow cytometry analysis performed in whole kidney tissue confirmed increased expression of C1q, C1r, and C1s as well as complement activation, which is measured as increased synthesis of C3 fragments predominantly in the interstitial compartment. Flow studies localized increased C1q expression to PDGFRß-positive pericytes as well as to CD45-positive cells. Although deletion of C1qA did not prevent kidney fibrosis, global deletion of C3 reduced macrophage infiltration, reduced synthesis of C3 fragments, and reduced fibrosis. Clodronate mediated depletion of CD11bF4/80 high macrophages in UUO mice also reduced complement gene expression and reduced fibrosis. Our studies demonstrate local synthesis of complement by both PDGFRß-positive pericytes and CD45-positive cells in kidney fibrosis. Inhibition of complement activation represents a novel therapeutic target to ameliorate fibrosis and progression of chronic kidney disease.


Subject(s)
Complement Activation , Complement C1q/metabolism , Complement C3/metabolism , Kidney Tubules/metabolism , Macrophages/metabolism , Pericytes/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Cell Communication , Complement C1q/deficiency , Complement C1q/genetics , Complement C1q/immunology , Complement C3/deficiency , Complement C3/genetics , Complement C3/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Fibrosis , Folic Acid , Genotype , Inflammation Mediators/metabolism , Kidney Tubules/immunology , Kidney Tubules/pathology , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Pericytes/immunology , Pericytes/pathology , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Time Factors , Ureteral Obstruction/complications , Wnt Signaling Pathway , Wnt3A Protein/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L556-L567, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28188224

ABSTRACT

Pericytes are perivascular PDGF receptor-ß+ (PDGFRß+) stromal cells required for vasculogenesis and maintenance of microvascular homeostasis in many organs. Because of their unique juxtaposition to microvascular endothelium, lung PDGFRß+ cells are well situated to detect proinflammatory molecules released following epithelial injury and promote acute inflammatory responses. Thus we hypothesized that these cells represent an unrecognized immune surveillance or injury-sentinel interstitial cell. To evaluate this hypothesis, we isolated PDGFRß+ cells from murine lung and demonstrated that they have characteristics consistent with a pericyte population (referred to as pericyte-like cells for simplicity hereafter). We showed that pericyte-like cells expressed functional Toll-like receptors and upregulated chemokine expression following exposure to bronchoalveolar lavage fluid (BALF) collected from mice with sterile lung injury. Interestingly, BALF from mice without lung injury also induced chemokine expression in pericyte-like cells, suggesting that pericyte-like cells are primed to sense epithelial injury (permeability changes). Following LPS-induced lung inflammation, increased numbers of pericyte-like cells expressed IL-6, chemokine (C-X-C motif) ligand-1, chemokine (C-C motif) ligand 2/ monocyte chemotactic protein-1, and ICAM-1 in vivo. Sterile lung injury in pericyte-ablated mice was associated with decreased inflammation compared with normal mice. In summary, we found that pericyte-like cells are immune responsive and express diverse chemokines in response to lung injury in vitro and in vivo. Furthermore, pericyte-like cell ablation attenuated inflammation in sterile lung injury, suggesting that these cells play an important functional role in mediating lung inflammatory responses. We propose a model in which pericyte-like cells function as interstitial immune sentinels, detecting proinflammatory molecules released following epithelial barrier damage and participating in recruitment of circulating leukocytes.


Subject(s)
Immune System/cytology , Lung/cytology , Pericytes/cytology , Animals , Bronchoalveolar Lavage Fluid , Cells, Cultured , Inflammation/pathology , Inflammation Mediators/metabolism , Lung Injury/pathology , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stromal Cells/metabolism
9.
PLoS Med ; 14(2): e1002248, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28245243

ABSTRACT

BACKGROUND: Chronic liver scarring from any cause leads to cirrhosis, portal hypertension, and a progressive decline in renal blood flow and renal function. Extreme renal vasoconstriction characterizes hepatorenal syndrome, a functional and potentially reversible form of acute kidney injury in patients with advanced cirrhosis, but current therapy with systemic vasoconstrictors is ineffective in a substantial proportion of patients and is limited by ischemic adverse events. Serelaxin (recombinant human relaxin-2) is a peptide molecule with anti-fibrotic and vasoprotective properties that binds to relaxin family peptide receptor-1 (RXFP1) and has been shown to increase renal perfusion in healthy human volunteers. We hypothesized that serelaxin could ameliorate renal vasoconstriction and renal dysfunction in patients with cirrhosis and portal hypertension. METHODS AND FINDINGS: To establish preclinical proof of concept, we developed two independent rat models of cirrhosis that were characterized by progressive reduction in renal blood flow and glomerular filtration rate and showed evidence of renal endothelial dysfunction. We then set out to further explore and validate our hypothesis in a phase 2 randomized open-label parallel-group study in male and female patients with alcohol-related cirrhosis and portal hypertension. Forty patients were randomized 1:1 to treatment with serelaxin intravenous (i.v.) infusion (for 60 min at 80 µg/kg/d and then 60 min at 30 µg/kg/d) or terlipressin (single 2-mg i.v. bolus), and the regional hemodynamic effects were quantified by phase contrast magnetic resonance angiography at baseline and after 120 min. The primary endpoint was the change from baseline in total renal artery blood flow. Therapeutic targeting of renal vasoconstriction with serelaxin in the rat models increased kidney perfusion, oxygenation, and function through reduction in renal vascular resistance, reversal of endothelial dysfunction, and increased activation of the AKT/eNOS/NO signaling pathway in the kidney. In the randomized clinical study, infusion of serelaxin for 120 min increased total renal arterial blood flow by 65% (95% CI 40%, 95%; p < 0.001) from baseline. Administration of serelaxin was safe and well tolerated, with no detrimental effect on systemic blood pressure or hepatic perfusion. The clinical study's main limitations were the relatively small sample size and stable, well-compensated population. CONCLUSIONS: Our mechanistic findings in rat models and exploratory study in human cirrhosis suggest the therapeutic potential of selective renal vasodilation using serelaxin as a new treatment for renal dysfunction in cirrhosis, although further validation in patients with more advanced cirrhosis and renal dysfunction is required. TRIAL REGISTRATION: ClinicalTrials.gov NCT01640964.


Subject(s)
Kidney Diseases/drug therapy , Kidney/drug effects , Liver Cirrhosis/drug therapy , Relaxin/pharmacology , Relaxin/therapeutic use , Adolescent , Adult , Aged , Animals , Female , Glomerular Filtration Rate/drug effects , Humans , Kidney/blood supply , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Regional Blood Flow/drug effects , Scotland , Young Adult
10.
Kidney Int ; 91(3): 628-641, 2017 03.
Article in English | MEDLINE | ID: mdl-27927603

ABSTRACT

The expansion of the fibroblast pool is a critical step in organ fibrosis, but the mechanisms driving expansion remain to be fully clarified. We previously showed that lysophosphatidic acid (LPA) signaling through its receptor LPA1 expressed on fibroblasts directly induces the recruitment of these cells. Here we tested whether LPA-LPA1 signaling drives fibroblast proliferation and activation during the development of renal fibrosis. LPA1-deficient (LPA1-/-) or -sufficient (LPA1+/+) mice were crossed to mice with green fluorescent protein expression (GFP) driven by the type I procollagen promoter (Col-GFP) to identify fibroblasts. Unilateral ureteral obstruction-induced increases in renal collagen were significantly, though not completely, attenuated in LPA1-/-Col-GFP mice, as were the accumulations of both fibroblasts and myofibroblasts. Connective tissue growth factor was detected mainly in tubular epithelial cells, and its levels were suppressed in LPA1-/-Col-GFP mice. LPA-LPA1 signaling directly induced connective tissue growth factor expression in primary proximal tubular epithelial cells, through a myocardin-related transcription factor-serum response factor pathway. Proximal tubular epithelial cell-derived connective tissue growth factor mediated renal fibroblast proliferation and myofibroblast differentiation. Administration of an inhibitor of myocardin-related transcription factor/serum response factor suppressed obstruction-induced renal fibrosis. Thus, targeting LPA-LPA1 signaling and/or myocardin-related transcription factor/serum response factor-induced transcription could be promising therapeutic strategies for renal fibrosis.


Subject(s)
Cell Communication/drug effects , Connective Tissue Growth Factor/metabolism , Epithelial Cells/drug effects , Fibroblasts/drug effects , Kidney Diseases/metabolism , Kidney/drug effects , Lysophospholipids/pharmacology , Receptors, Lysophosphatidic Acid/agonists , Signal Transduction/drug effects , Ureteral Obstruction/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Mice, Knockout , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Promoter Regions, Genetic , RNA Interference , Receptors, Lysophosphatidic Acid/deficiency , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transfection , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
11.
Am J Pathol ; 186(10): 2519-31, 2016 10.
Article in English | MEDLINE | ID: mdl-27555112

ABSTRACT

Pericytes, resident fibroblasts, and mesenchymal stem cells are poorly described cell populations. They have recently been characterized in much greater detail in rodent lungs and have been shown to play important roles in development, homeostasis, response to injury and pathogens, as well as recovery from damage. These closely related mesenchymal cell populations form extensive connections to the lung's internal structure, as well as its internal and external surfaces. They generate and remodel extracellular matrix, coregulate the vasculature, help maintain and restore the epithelium, and act as sentries for the immune system. In this review, we revisit these functions in light of significant advances in characterizing and tracking lung fibroblast populations in rodents. Lineage tracing experiments have mapped the heritage, identified functions that discriminate lung pericytes from resident fibroblasts, identified a subset of mesenchymal stem cells, and shown these populations to be the predominant progenitors of pathological fibroblasts and myofibroblasts in lung diseases. These findings point to the importance of resident lung mesenchymal populations as therapeutic targets in acute lung injury as well as fibrotic and degenerative diseases. Far from being passive and quiescent, pericytes and resident fibroblasts are busily sensing and responding, through diverse mechanisms, to changes in lung health and function.


Subject(s)
Fibroblasts/physiology , Lung Injury/therapy , Lung/pathology , Mesenchymal Stem Cells/physiology , Pericytes/physiology , Pulmonary Fibrosis/pathology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Disease Models, Animal , Extracellular Matrix/pathology , Humans , Lung/embryology , Lung Injury/pathology , Myofibroblasts/physiology , Phenotype , Pulmonary Fibrosis/therapy , Rodentia
12.
Nephrol Dial Transplant ; 32(suppl_1): i89-i97, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28391346

ABSTRACT

Fibrosis, a characteristic of all chronic kidney diseases, is now recognized to be an independent predictor of disease progression. Deposition of pathological matrix in the walls of glomerular capillaries, the interstitial space and around arterioles both predicts and contributes to functional demise of the nephron and its surrounding vasculature. Recent identification of the major cell populations of fibroblast precursors in the kidney interstitium as pericytes and tissue-resident mesenchymal stem cells, and in the glomerulus as podocytes, parietal epithelial and mesangial cells, has enabled the study of the fibrogenic process in much greater depth directly in the fibroblast precursors. These cells are not only matrix-producing cells, but are also important innate immune surveillance cells that regulate the inflammatory process, exacerbate tissue damage by release of radicals and cytokines, and contribute to parenchymal and microvascular dysfunction by aberrant wound-healing responses. Innate immune signaling in fibroblasts and their precursors is intimately intertwined with the process of fibrogenesis. In addition, genomic and genetic studies also point to defective responses in loci close to genes involved in solute transport, metabolism, autophagy, protein handling and vascular homeostasis, principally in the epithelium and endothelium, as upstream drivers of the fibrotic process, indicating that cellular crosstalk is vital for development of fibrosis. As we move beyond TGFß inhibition as a central target for fibrosis, targeting innate immune signaling and metabolic dysfunction appear increasingly tenable alternative targets for novel therapies.


Subject(s)
Fibrosis/pathology , Fibrosis/prevention & control , Kidney/pathology , Animals , Humans
13.
J Pathol ; 240(4): 410-424, 2016 12.
Article in English | MEDLINE | ID: mdl-27569721

ABSTRACT

Fibrosis is a characteristic of Duchenne muscular dystrophy (DMD), yet the cellular and molecular mechanisms responsible for DMD fibrosis are poorly understood. Utilizing the Collagen1a1-GFP transgene to identify cells producing Collagen-I matrix in wild-type mice exposed to toxic injury or those mutated at the dystrophin gene locus (mdx) as a model of DMD, we studied mechanisms of skeletal muscle injury/repair and fibrosis. PDGFRα is restricted to Sca1+, CD45- mesenchymal progenitors. Fate-mapping experiments using inducible CreER/LoxP somatic recombination indicate that these progenitors expand in injury or DMD to become PDGFRα+, Col1a1-GFP+ matrix-forming fibroblasts, whereas muscle fibres do not become fibroblasts but are an important source of the PDGFRα ligand, PDGF-AA. While in toxin injury/repair of muscle PDGFRα, signalling is transiently up-regulated during the regenerative phase in the DMD model and in human DMD it is chronically overactivated. Conditional expression of the constitutively active PDGFRα D842V mutation in Collagen-I+ fibroblasts, during injury/repair, hindered the repair phase and instead promoted fibrosis. In DMD, treatment of mdx mice with crenolanib, a highly selective PDGFRα/ß tyrosine kinase inhibitor, reduced fibrosis, improved muscle strength, and was associated with decreased activity of Src, a downstream effector of PDGFRα signalling. These observations are consistent with a model in which PDGFRα activation of mesenchymal progenitors normally regulates repair of the injured muscle, but in DMD persistent and excessive activation of this pathway directly drives fibrosis and hinders repair. The PDGFRα pathway is a potential new target for treatment of progressive DMD. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Collagen Type I/biosynthesis , Muscular Dystrophy, Duchenne/pathology , Receptor, Platelet-Derived Growth Factor alpha/physiology , Animals , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cells, Cultured , Disease Models, Animal , Dystrophin/genetics , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Male , Mice, Transgenic , Muscle Strength/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Mutation , Piperidines/pharmacology , Piperidines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/genetics , Regeneration/drug effects , Regeneration/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
14.
J Am Soc Nephrol ; 27(12): 3639-3652, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27026366

ABSTRACT

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.


Subject(s)
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 Am Soc Nephrol ; 27(8): 2370-81, 2016 08.
Article in English | MEDLINE | ID: mdl-26657868

ABSTRACT

Human kidney peritubular capillaries are particularly susceptible to injury, resulting in dysregulated angiogenesis, capillary rarefaction and regression, and progressive loss of kidney function. However, little is known about the structure and function of human kidney microvasculature. Here, we isolated, purified, and characterized human kidney peritubular microvascular endothelial cells (HKMECs) and reconstituted a three-dimensional human kidney microvasculature in a flow-directed microphysiologic system. By combining epithelial cell depletion and cell culture in media with high concentrations of vascular endothelial growth factor, we obtained HKMECs of high purity in large quantity. Unlike other endothelial cells, isolated HKMECs depended on high vascular endothelial growth factor concentration for survival and growth and exhibited high tubulogenic but low angiogenic potential. Furthermore, HKMECs had a different transcriptional profile. Under flow, HKMECs formed a thin fenestrated endothelium with a functional permeability barrier. In conclusion, this three-dimensional HKMEC-specific microphysiologic system recapitulates human kidney microvascular structure and function and shows phenotypic characteristics different from those of other microvascular endothelial cells.


Subject(s)
Capillaries/cytology , Endothelial Cells , Kidney Tubules/cytology , Cells, Cultured , Disease Progression , Humans , Kidney Diseases/etiology
16.
J Biol Chem ; 290(23): 14656-67, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25931125

ABSTRACT

The physiological roles of macrophages and dendritic cells (DCs) in lean white adipose tissue homeostasis have received little attention. Because DCs are generated from bone marrow progenitors in the presence of granulocyte/macrophage colony-stimulating factor (GM-CSF), we used GM-CSF-deficient (Csf2(-/-)) mice fed a low fat diet to test the hypothesis that adipose tissue DCs regulate the development of adipose tissue. At 4 weeks of age, Csf2(-/-) mice had 75% fewer CD45(+)Cd11b(+)Cd11c(+)MHCII(+) F4/80(-) DCs in white adipose tissue than did wild-type controls. Furthermore, the Csf2(-/-) mice showed a 30% increase in whole body adiposity, which persisted to adulthood. Adipocytes from Csf2(-/-) mice were 50% larger by volume and contained higher levels of adipogenesis gene transcripts, indicating enhanced adipocyte differentiation. In contrast, adipogenesis/adipocyte lipid accumulation was inhibited when preadipocytes were co-cultured with CD45(+)Cd11b(+)Cd11c(+)MHCII(+)F4/80(-) DCs. Medium conditioned by DCs, but not by macrophages, also inhibited adipocyte lipid accumulation. Proteomic analysis revealed that matrix metalloproteinase 12 and fibronectin 1 were greatly enriched in the medium conditioned by DCs compared with that conditioned by macrophages. Silencing fibronectin or genetic deletion of matrix metalloproteinase 12 in DCs partially reversed the inhibition of adipocyte lipid accumulation. Our observations indicate that DCs residing in adipose tissue play a critical role in suppressing normal adipose tissue expansion.


Subject(s)
Adipogenesis , Adipose Tissue/cytology , Dendritic Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , 3T3-L1 Cells , Adipose Tissue/metabolism , Aging , Animals , Dendritic Cells/cytology , Energy Metabolism , Female , Gene Deletion , Glucose/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Homeostasis , Male , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism
17.
Am J Physiol Renal Physiol ; 310(10): F931-44, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26911854

ABSTRACT

MicroRNAs (miRs), a class of small noncoding RNAs that act as post-transcriptional regulators of gene expression, have attracted increasing attention as critical regulators of organogenesis, cancer, and disease. Interest has been spurred by development of a novel class of synthetic RNA oligonucleotides with excellent drug-like properties that hybridize to a specific miR, preventing its action. In kidney disease, a small number of miRs are dysregulated. These overlap with regulated miRs in nephrogenesis and kidney cancers. Several dysregulated miRs have been identified in fibrotic diseases of other organs, representing a "fibrotic signature," and some of these fibrotic miRs contribute remarkably to the pathogenesis of kidney disease. Chronic kidney disease, affecting ∼10% of the population, leads to kidney failure, with few treatment options. Here, we will explore the pathological mechanism of miR-21, whose pre-eminent role in amplifying kidney disease and fibrosis by suppressing mitochondrial biogenesis and function is established. Evolving roles for miR-214, -199, -200, -155, -29, -223, and -126 in kidney disease will be discussed, and we will demonstrate how studying functions of distinct miRs has led to new mechanistic insights for kidney disease progression. Finally, the utility of anti-miR oligonucleotides as potential novel therapeutics to treat chronic disease will be highlighted.


Subject(s)
MicroRNAs/metabolism , Molecular Targeted Therapy , Oligonucleotides/therapeutic use , Organelle Biogenesis , Renal Insufficiency/metabolism , Animals , Fatty Acids/metabolism , Fibrosis , Humans , Kidney/pathology , MicroRNAs/antagonists & inhibitors , Oligonucleotides/pharmacology , Oxidation-Reduction , Renal Insufficiency/drug therapy , Renal Insufficiency/etiology
18.
Am J Physiol Renal Physiol ; 311(6): F1230-F1242, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27335372

ABSTRACT

Pericytes are tissue-resident mesenchymal progenitor cells anatomically associated with the vasculature that have been shown to participate in tissue regeneration. Here, we tested the hypothesis that kidney pericytes, derived from FoxD1+ mesodermal progenitors during embryogenesis, are necessary for postnatal kidney homeostasis. Diphtheria toxin delivery to FoxD1Cre::RsDTR transgenic mice resulted in selective ablation of >90% of kidney pericytes but not other cell lineages. Abrupt increases in plasma creatinine, blood urea nitrogen, and albuminuria within 96 h indicated acute kidney injury in pericyte-ablated mice. Loss of pericytes led to a rapid accumulation of neutral lipid vacuoles, swollen mitochondria, and apoptosis in tubular epithelial cells. Pericyte ablation led to endothelial cell swelling, reduced expression of vascular homeostasis markers, and peritubular capillary loss. Despite the observed injury, no signs of the acute inflammatory response were observed. Pathway enrichment analysis of genes expressed in kidney pericytes in vivo identified basement membrane proteins, angiogenic factors, and factors regulating vascular tone as major regulators of vascular function. Using novel microphysiological devices, we recapitulated human kidney peritubular capillaries coated with pericytes and showed that pericytes regulate permeability, basement membrane deposition, and microvascular tone. These findings suggest that through the active support of the microvasculature, pericytes are essential to adult kidney homeostasis.


Subject(s)
Acute Kidney Injury/metabolism , Capillaries/metabolism , Endothelium, Vascular/metabolism , Kidney/blood supply , Pericytes/metabolism , Animals , Kidney/metabolism , Mice , Mice, Transgenic , Microvessels/metabolism , Permeability
19.
Crit Care Med ; 44(8): e594-603, 2016 08.
Article in English | MEDLINE | ID: mdl-26757163

ABSTRACT

OBJECTIVES: Peroxisome proliferator-activated receptor-α is significantly down-regulated in circulating leukocytes from children with sepsis. Peroxisome proliferator-activated receptor-α null (Ppara) mice have greater mortality than wild-type mice when subjected to sepsis by cecal ligation and puncture. We sought to characterize the role of peroxisome proliferator-activated receptor-α in sepsis and to identify the mechanism whereby peroxisome proliferator-activated receptor-α confers a survival advantage. DESIGN: Prospective randomized preclinical study. SETTING: Laboratory investigation. SUBJECTS: Male C57Bl/6J and Ppara mice (B6.129S4-Ppara/J), aged 12-16 weeks. INTERVENTIONS: Bone marrow chimeric mice were generated and subjected to cecal ligation and puncture. Survival was measured for 7 days. Separate groups of nontransplanted mice underwent cecal ligation and puncture and were euthanized 24 hours later for plasma and tissue analyses. MEASUREMENTS AND MAIN RESULTS: Ppara mice had dramatically reduced survival compared with wild-type mice irrespective of the peroxisome proliferator-activated receptor-α status of the bone marrow they received (3% vs 63%; p < 0.0001). No difference in survival was observed between Ppara mice that received wild-type versus Ppara marrow or in wild-type mice receiving wild-type versus Ppara marrow. In septic, nontransplanted mice at 24 hours, Ppara mice had elevated cardiac troponin levels compared with wild-type mice. Cardiac histologic injury scores were greater in Ppara versus wild-type mice. Expression of transcription factors and enzymes related to fatty acid oxidation in the heart were profoundly down-regulated in both wild-type and Ppara mice, but more so in the Ppara mice. CONCLUSIONS: Peroxisome proliferator-activated receptor-α expression in nonhematopoietic tissues plays a critical role in determining clinical outcome in experimental polymicrobial sepsis and is more important to survival in sepsis than hematopoietic peroxisome proliferator-activated receptor-α expression. Cardiac injury due to inadequate energy production from fatty acid substrate is a probable mechanism of decreased survival in Ppara mice. These results suggest that altered peroxisome proliferator-activated receptor-α-mediated cellular metabolism may play an important role in sepsis-related end-organ injury and dysfunction, especially in the heart.


Subject(s)
PPAR alpha/biosynthesis , PPAR alpha/genetics , Sepsis/genetics , Sepsis/mortality , Animals , Behavior, Animal , Blood Glucose , Body Weight , Bone Marrow Transplantation , Disease Models, Animal , Down-Regulation , Gene Expression , Health Status , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Prospective Studies , Random Allocation , Sepsis/physiopathology , Troponin I/biosynthesis
20.
BMC Nephrol ; 17: 5, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26746687

ABSTRACT

BACKGROUND: Recent studies indicate that mural cells of the preglomerular vessels, known as cells of renin lineage (CoRL), contribute to repair and regeneration of injured kidney glomeruli. However, their potential roles in tubulointerstitial disease are less understood. The aim of this study was to better understand CoRL number and distribution following UUO so that future mechanistic studies could be undertaken. METHODS: We mapped the fate of CoRL in adult Ren1cCreER x Rs-tdTomato-R reporter mice that underwent UUO. Kidney biopsies from sham and UUO-subjected mice on days 3, 7, and 14 were evaluated by immunohistochemistry. RESULTS: In sham animals, CoRL were restricted to juxtaglomerular location. At day 7 following UUO, CoRL increased two-fold, were perivascular in location, and co-expressed pericyte markers (PDGFßR, NG2), but did not express renin. At day 14 post UUO, labeled CoRL detached from vessels and were present in the interstitium, in areas of fibrosis, where they now expressed the myofibroblast marker alpha-smooth muscle actin. The increase in CoRL was likely due to proliferation as marked by BrdU labeling, and migration from the cortex. Following UUO starting from day 3, active hypoxia inducible factor-2α was detected in nuclei in labeled CoRL, in the cortex, but not those cells found in medulla. CONCLUSIONS: We have demonstrated that arteriolar CoRL are potential kidney progenitors that may contribute to the initial vascular regeneration. However, in chronic kidney injury (≥14 days post UUO), perivascular CoRL transition to myofibroblast-like cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Kidney/cytology , Kidney/metabolism , Pericytes/metabolism , Renal Insufficiency, Chronic/metabolism , Actins/metabolism , Animals , Antigens/metabolism , Cell Differentiation , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Female , Fibrosis , Mice , Pericytes/cytology , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Renal Insufficiency, Chronic/etiology , Renin/metabolism , Ureteral Obstruction/complications
SELECTION OF CITATIONS
SEARCH DETAIL