Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
PLoS One ; 18(8): e0289778, 2023.
Article En | MEDLINE | ID: mdl-37540694

PKD1 is the most commonly mutated gene causing autosomal dominant polycystic kidney disease (ADPKD). It encodes Polycystin-1 (PC1), a putative membrane protein that undergoes a set of incompletely characterized post-transcriptional cleavage steps and has been reported to localize in multiple subcellular locations, including the primary cilium and mitochondria. However, direct visualization of PC1 and detailed characterization of its binding partners remain challenging. We now report a new mouse model with HA epitopes and eGFP knocked-in frame into the endogenous mouse Pkd1 gene by CRISPR/Cas9. Using this model, we sought to visualize endogenous PC1-eGFP and performed affinity-purification mass spectrometry (AP-MS) and network analyses. We show that the modified Pkd1 allele is fully functional but the eGFP-tagged protein cannot be detected without signal amplification by secondary antibodies. Using nanobody-coupled beads and large quantities of tissue, AP-MS identified an in vivo PC1 interactome, which is enriched for mitochondrial proteins and components of metabolic pathways. These studies suggest this mouse model and interactome data will be useful to understand PC1 function, but that new methods and brighter tags will be required to track endogenous PC1.


Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Mice , Animals , TRPP Cation Channels/chemistry , Polycystic Kidney, Autosomal Dominant/genetics , Disease Models, Animal
3.
Nature ; 610(7932): 547-554, 2022 10.
Article En | MEDLINE | ID: mdl-36198790

Loss of Paneth cells and their antimicrobial granules compromises the intestinal epithelial barrier and is associated with Crohn's disease, a major type of inflammatory bowel disease1-7. Non-classical lymphoid cells, broadly referred to as intraepithelial lymphocytes (IELs), intercalate the intestinal epithelium8,9. This anatomical position has implicated them as first-line defenders in resistance to infections, but their role in inflammatory disease pathogenesis requires clarification. The identification of mediators that coordinate crosstalk between specific IEL and epithelial subsets could provide insight into intestinal barrier mechanisms in health and disease. Here we show that the subset of IELs that express γ and δ T cell receptor subunits (γδ IELs) promotes the viability of Paneth cells deficient in the Crohn's disease susceptibility gene ATG16L1. Using an ex vivo lymphocyte-epithelium co-culture system, we identified apoptosis inhibitor 5 (API5) as a Paneth cell-protective factor secreted by γδ IELs. In the Atg16l1-mutant mouse model, viral infection induced a loss of Paneth cells and enhanced susceptibility to intestinal injury by inhibiting the secretion of API5 from γδ IELs. Therapeutic administration of recombinant API5 protected Paneth cells in vivo in mice and ex vivo in human organoids with the ATG16L1 risk allele. Thus, we identify API5 as a protective γδ IEL effector that masks genetic susceptibility to Paneth cell death.


Apoptosis Regulatory Proteins , Crohn Disease , Genetic Predisposition to Disease , Intraepithelial Lymphocytes , Nuclear Proteins , Paneth Cells , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Cell Death , Crohn Disease/genetics , Crohn Disease/metabolism , Crohn Disease/pathology , Genetic Predisposition to Disease/genetics , Intestinal Mucosa/pathology , Nuclear Proteins/metabolism , Paneth Cells/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Cell Survival , Organoids , Alleles
4.
Blood ; 135(26): 2388-2401, 2020 06 25.
Article En | MEDLINE | ID: mdl-32232483

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Graft vs Host Disease/prevention & control , Intestinal Diseases/prevention & control , Organoids , T-Lymphocytes/immunology , Acrylamides/pharmacology , Animals , Autophagy , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bone Marrow Transplantation/adverse effects , Coculture Techniques , Colon/abnormalities , Female , Genetic Predisposition to Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inflammatory Bowel Diseases/pathology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Necroptosis/drug effects , Nitriles , Paneth Cells/pathology , Precision Medicine , Pyrazoles/pharmacology , Pyrimidines , Radiation Chimera , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Sulfonamides/pharmacology , T-Lymphocytes/transplantation
5.
J Am Soc Nephrol ; 31(3): 560-577, 2020 03.
Article En | MEDLINE | ID: mdl-31996409

BACKGROUND: Prolyl hydroxylase domain (PHD) inhibitors, which stimulate erythropoietin production through the activation of hypoxia-inducible factor (HIF), are novel therapeutic agents used for treating renal anemia. Several PHD inhibitors, including enarodustat, are currently undergoing phase 2 or phase 3 clinical trials. Because HIF regulates a broad spectrum of genes, PHD inhibitors are expected to have other effects in addition to erythropoiesis, such as protection against metabolic disorders. However, whether such beneficial effects would extend to metabolic disorder-related kidney disease is largely unknown. METHODS: We administered enarodustat or vehicle without enarodustat in feed to diabetic black and tan brachyury (BTBR) ob/ob mice from 4 to 22 weeks of age. To elucidate molecular changes induced by enarodustat, we performed transcriptome analysis of isolated glomeruli and in vitro experiments using murine mesangial cells. RESULTS: Compared with BTBR ob/ob mice that received only vehicle, BTBR ob/ob mice treated with enarodustat displayed lower body weight, reduced blood glucose levels with improved insulin sensitivity, lower total cholesterol levels, higher adiponectin levels, and less adipose tissue, as well as a tendency for lower macrophage infiltration. Enarodustat-treated mice also exhibited reduced albuminuria and amelioration of glomerular epithelial and endothelial damage. Transcriptome analysis of isolated glomeruli revealed reduced expression of C-C motif chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) in enarodustat-treated mice compared with the vehicle-only group, accompanied by reduced glomerular macrophage infiltration. In vitro experiments demonstrated that both local HIF-1 activation and restoration of adiponectin by enarodustat contributed to CCL2/MCP-1 reduction in mesangial cells. CONCLUSIONS: These results indicate that the PHD inhibitor enarodustat has potential renoprotective effects in addition to its potential to protect against metabolic disorders.


Chemokine CCL2/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/prevention & control , Prolyl-Hydroxylase Inhibitors/pharmacology , Animals , Chemokine CCL2/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/metabolism , Disease Models, Animal , Insulin Resistance , Metabolic Diseases/etiology , Metabolic Diseases/prevention & control , Mice , Mice, Obese , N-substituted Glycines/pharmacology , Prolyl Hydroxylases/metabolism , Pyridines/pharmacology , Random Allocation , Reference Values , Treatment Outcome , Triazoles/pharmacology
6.
J Clin Invest ; 130(4): 2111-2128, 2020 04 01.
Article En | MEDLINE | ID: mdl-31961824

Although Western diet and dysbiosis are the most prominent environmental factors associated with inflammatory bowel diseases (IBDs), the corresponding host factors and cellular mechanisms remain poorly defined. Here we report that the TSC1/mTOR pathway in the gut epithelium represents a metabolic and innate immune checkpoint for intestinal dysfunction and inflammation. mTOR hyperactivation triggered by Western diet or Tsc1 ablation led to epithelium necroptosis, barrier disruption, and predisposition to dextran sulfate sodium-induced colitis and inflammation-associated colon cancer. Mechanistically, our results uncovered a critical role for TSC1/mTOR in restraining the expression and activation of RIPK3 in the gut epithelium through TRIM11-mediated ubiquitination and autophagy-dependent degradation. Notably, microbiota depletion by antibiotics or gnotobiotics attenuated RIPK3 expression and activation, thereby alleviating epithelial necroptosis and colitis driven by mTOR hyperactivation. mTOR primarily impinged on RIPK3 to potentiate necroptosis induced by TNF and by microbial pathogen-associated molecular patterns (PAMPs), and hyperactive mTOR and aberrant necroptosis were intertwined in human IBDs. Together, our data reveal a previously unsuspected link between the Western diet, microbiota, and necroptosis and identify the mTOR/RIPK3/necroptosis axis as a driving force for intestinal inflammation and cancer.


Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Neoplasms/metabolism , Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Animals , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 1 Protein/genetics
7.
J Clin Invest ; 129(8): 3387-3400, 2019 07 22.
Article En | MEDLINE | ID: mdl-31329164

Fibroblasts from patients with Tangier disease carrying ATP-binding cassette A1 (ABCA1) loss-of-function mutations are characterized by cardiolipin accumulation, a mitochondrial-specific phospholipid. Suppression of ABCA1 expression occurs in glomeruli from patients with diabetic kidney disease (DKD) and in human podocytes exposed to DKD sera collected prior to the development of DKD. We demonstrated that siRNA ABCA1 knockdown in podocytes led to reduced oxygen consumption capabilities associated with alterations in the oxidative phosphorylation (OXPHOS) complexes and with cardiolipin accumulation. Podocyte-specific deletion of Abca1 (Abca1fl/fl) rendered mice susceptible to DKD, and pharmacological induction of ABCA1 improved established DKD. This was not mediated by free cholesterol, as genetic deletion of sterol-o-acyltransferase-1 (SOAT1) in Abca1fl/fl mice was sufficient to cause free cholesterol accumulation but did not cause glomerular injury. Instead, cardiolipin mediates ABCA1-dependent susceptibility to podocyte injury, as inhibition of cardiolipin peroxidation with elamipretide improved DKD in vivo and prevented ABCA1-dependent podocyte injury in vitro and in vivo. Collectively, we describe a pathway definitively linking ABCA1 deficiency to cardiolipin-driven mitochondrial dysfunction. We demonstrated that this pathway is relevant to DKD and that ABCA1 inducers or inhibitors of cardiolipin peroxidation may each represent therapeutic strategies for the treatment of established DKD.


ATP Binding Cassette Transporter 1/deficiency , Cardiolipins/metabolism , Diabetic Nephropathies/metabolism , Lipid Peroxidation , Mitochondria/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Cardiolipins/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Podocytes , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
8.
Nat Microbiol ; 4(10): 1737-1749, 2019 10.
Article En | MEDLINE | ID: mdl-31182797

Products derived from bacterial members of the gut microbiota evoke immune signalling pathways of the host that promote immunity and barrier function in the intestine. How immune reactions to enteric viruses support intestinal homeostasis is unknown. We recently demonstrated that infection by murine norovirus (MNV) reverses intestinal abnormalities following depletion of bacteria, indicating that an intestinal animal virus can provide cues to the host that are typically attributed to the microbiota. Here, we elucidate mechanisms by which MNV evokes protective responses from the host. We identify an important role for the viral protein NS1/2 in establishing local replication and a type I interferon (IFN-I) response in the colon. We further show that IFN-I acts on intestinal epithelial cells to increase the proportion of CCR2-dependent macrophages and interleukin (IL)-22-producing innate lymphoid cells, which in turn promote pSTAT3 signalling in intestinal epithelial cells and protection from intestinal injury. In addition, we demonstrate that MNV provides a striking IL-22-dependent protection against early-life lethal infection by Citrobacter rodentium. These findings demonstrate novel ways in which a viral member of the microbiota fortifies the intestinal barrier during chemical injury and infectious challenges.


Gastrointestinal Microbiome/immunology , Interferon Type I/metabolism , Interleukins/metabolism , Intestines/immunology , Intestines/virology , Animals , Anti-Bacterial Agents/toxicity , Cell Proliferation , Citrobacter rodentium/physiology , Colon/cytology , Colon/immunology , Colon/metabolism , Colon/virology , Dextran Sulfate/toxicity , Enterobacteriaceae Infections/prevention & control , Interleukins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/drug effects , Lymphocytes/cytology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Norovirus/immunology , Norovirus/physiology , Signal Transduction/genetics , Specific Pathogen-Free Organisms , Viral Nonstructural Proteins/genetics , Virus Replication , Interleukin-22
9.
Kidney Int ; 95(3): 577-589, 2019 03.
Article En | MEDLINE | ID: mdl-30639234

Tubulointerstitial fibrosis is a strong predictor of progression in patients with chronic kidney disease, and is often accompanied by lipid accumulation in renal tubules. However, the molecular mechanisms modulating the relationship between lipotoxicity and tubulointerstitial fibrosis remain obscure. ATF6α, a transcription factor of the unfolded protein response, is reported to be an upstream regulator of fatty acid metabolism. Owing to their high energy demand, proximal tubular cells (PTCs) use fatty acids as their main energy source. We therefore hypothesized that ATF6α regulates PTC fatty acid metabolism, contributing to lipotoxicity-induced tubulointerstitial fibrosis. Overexpression of activated ATF6α transcriptionally downregulated peroxisome proliferator-activated receptor-α (PPARα), the master regulator of lipid metabolism, leading to reduced activity of fatty acid ß-oxidation and cytosolic accumulation of lipid droplets in a human PTC line (HK-2). ATF6α-induced lipid accumulation caused mitochondrial dysfunction, enhanced apoptosis, and increased expression of connective tissue growth factor (CTGF), as well as reduced cell viability. Atf6α-/- mice had sustained expression of PPARα and less tubular lipid accumulation following unilateral ischemia-reperfusion injury (uIRI), resulting in the amelioration of apoptosis; reduced expression of CTGF, α-smooth muscle actin, and collagen I; and less tubulointerstitial fibrosis. Administration of fenofibrate, a PPARα agonist, reduced lipid accumulation and tubulointerstitial fibrosis in the uIRI model. Taken together, these findings suggest that ATF6α deranges fatty acid metabolism in PTCs, which leads to lipotoxicity-mediated apoptosis and CTGF upregulation, both of which promote tubulointerstitial fibrosis.


Activating Transcription Factor 6/metabolism , Kidney Tubules, Proximal/pathology , PPAR alpha/metabolism , Renal Insufficiency, Chronic/pathology , Activating Transcription Factor 6/genetics , Animals , Cell Line , Disease Models, Animal , Down-Regulation , Fatty Acids/metabolism , Fibrosis , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Lipid Metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Renal Insufficiency, Chronic/etiology
10.
Clin Exp Pharmacol Physiol ; 45(11): 1097-1105, 2018 11.
Article En | MEDLINE | ID: mdl-30051924

End-stage renal disease is a leading cause of morbidity and mortality worldwide. The prevalence of the disease and the number of patients who receive renal replacement therapy are expected to increase in the next decade. Accumulating evidence suggests that chronic hypoxia in the tubulointerstitium represents the final common pathway to end-stage renal failure, and that reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the key players in kidney injury. However, ROS and RNS that exceed the physiological levels associated with the pathophysiology of most kidney diseases. The molecules that comprise ROS and RNS play an important role in regulating solute and water reabsorption in the kidney, which is vital for maintaining electrolyte homeostasis and the volume of extracellular fluid. This article reviews the physiological and pathophysiological role of ROS and RNS in normal kidney function and in various kidney diseases.


Kidney/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Humans , Kidney/physiology , Kidney/physiopathology , Oxidative Stress , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology
11.
Kidney Int ; 94(3): 536-550, 2018 09.
Article En | MEDLINE | ID: mdl-29887316

Lipotoxicity plays an important role in the progression of chronic kidney damage via various mechanisms, such as endoplasmic reticulum stress. Several studies proposed renal lipotoxicity in glomerular and tubular cells but the effect of lipid on renal erythropoietin (EPO)-producing (REP) cells in the interstitium has not been elucidated. Since renal anemia is caused by derangement of EPO production in REP cells, we evaluated the effect of palmitate, a representative long-chain saturated fatty acid, on EPO production and the endoplasmic reticulum stress pathway. EPO production was suppressed by palmitate (palmitate-conjugated bovine serum albumin [BSA]) or a high palmitate diet, but not oleic acid-conjugated BSA or a high oleic acid diet, especially under cobalt-induced pseudo-hypoxia both in vitro and in vivo. Importantly, suppression of EPO production was not induced by a decrease in transcription factor HIF activity, while it was significantly associated with endoplasmic reticulum stress, particularly transcription factor ATF4 activation, which suppresses 3'-enhancer activity of the EPO gene. ATF4 knockdown by siRNA significantly attenuated the suppressive effect of palmitate on EPO production. Studies utilizing inherited super-anemic mice (ISAM) mated with EPO-Cre mice (ISAM-REC mice) for lineage-labeling of REP cells showed that ATF4 activation by palmitate suppressed EPO production in REP cells. Laser capture microdissection confirmed ATF4 activation in the interstitial area of ISAM-REC mice treated with palmitate-conjugated BSA. Thus, endoplasmic reticulum stress induced by palmitate suppressed EPO expression by REP cells in a manner independent of HIF activation. The link between endoplasmic reticulum stress, dyslipidemia, and hypoxia may contribute to development and progression of anemia in CKD.


Activating Transcription Factor 4/metabolism , Anemia/pathology , Erythropoietin/metabolism , Kidney/metabolism , Palmitates/metabolism , Activating Transcription Factor 4/genetics , Anemia/blood , Anemia/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Endoplasmic Reticulum Stress , Erythropoietin/blood , Erythropoietin/genetics , Gene Knockdown Techniques , Hep G2 Cells , Humans , Laser Capture Microdissection , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology , Unfolded Protein Response
12.
Sci Rep ; 8(1): 2743, 2018 02 09.
Article En | MEDLINE | ID: mdl-29426897

Recent studies have reported intrinsic metabolic reprogramming in Pkd1 knock-out cells, implicating dysregulated cellular metabolism in the pathogenesis of polycystic kidney disease. However, the exact nature of the metabolic changes and their underlying cause remains controversial. We show herein that Pkd1 k o /ko renal epithelial cells have impaired fatty acid utilization, abnormal mitochondrial morphology and function, and that mitochondria in kidneys of ADPKD patients have morphological alterations. We further show that a C-terminal cleavage product of polycystin-1 (CTT) translocates to the mitochondria matrix and that expression of CTT in Pkd1 ko/ko cells rescues some of the mitochondrial phenotypes. Using Drosophila to model in vivo effects, we find that transgenic expression of mouse CTT results in decreased viability and exercise endurance but increased CO2 production, consistent with altered mitochondrial function. Our results suggest that PC1 may play a direct role in regulating mitochondrial function and cellular metabolism and provide a framework to understand how impaired mitochondrial function could be linked to the regulation of tubular diameter in both physiological and pathological conditions.


Kidney , Mitochondria , Mitochondrial Proteins/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Proteolysis , TRPP Cation Channels/metabolism , Aged , Animals , Animals, Genetically Modified , Dogs , Drosophila melanogaster , Embryo, Mammalian , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fatty Acids/metabolism , Gene Knockdown Techniques , Humans , Kidney/metabolism , Kidney/pathology , Madin Darby Canine Kidney Cells , Male , Mice , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , TRPP Cation Channels/genetics
13.
Annu Rev Immunol ; 36: 73-101, 2018 04 26.
Article En | MEDLINE | ID: mdl-29144836

The cellular degradative pathway of autophagy has a fundamental role in immunity. Here, we review the function of autophagy and autophagy proteins in inflammation. We discuss how the autophagy machinery controls the burden of infectious agents while simultaneously limiting inflammatory pathologies, which often involves processes that are distinct from conventional autophagy. Among the newly emerging processes we describe are LC3-associated phagocytosis and targeting by autophagy proteins, both of which require many of the same proteins that mediate conventional autophagy. We also discuss how autophagy contributes to differentiation of myeloid and lymphoid cell types, coordinates multicellular immunity, and facilitates memory responses. Together, these functions establish an intimate link between autophagy, mucosal immunity, and chronic inflammatory diseases. Finally, we offer our perspective on current challenges and barriers to translation.


Autophagy , Disease Susceptibility , Inflammation/etiology , Animals , Biomarkers , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunomodulation , Inflammation/diagnosis , Inflammation/metabolism , Signal Transduction
14.
J Exp Med ; 214(12): 3687-3705, 2017 Dec 04.
Article En | MEDLINE | ID: mdl-29089374

A variant of the autophagy gene ATG16L1 is associated with Crohn's disease, an inflammatory bowel disease (IBD), and poor survival in allogeneic hematopoietic stem cell transplant recipients. We demonstrate that ATG16L1 in the intestinal epithelium is essential for preventing loss of Paneth cells and exaggerated cell death in animal models of virally triggered IBD and allogeneic hematopoietic stem cell transplantation. Intestinal organoids lacking ATG16L1 reproduced this loss in Paneth cells and displayed TNFα-mediated necroptosis, a form of programmed necrosis. This cytoprotective function of ATG16L1 was associated with the role of autophagy in promoting mitochondrial homeostasis. Finally, therapeutic blockade of necroptosis through TNFα or RIPK1 inhibition ameliorated disease in the virally triggered IBD model. These findings indicate that, in contrast to tumor cells in which autophagy promotes caspase-independent cell death, ATG16L1 maintains the intestinal barrier by inhibiting necroptosis in the epithelium.


Apoptosis , Autophagy , Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Animals , Autophagy-Related Proteins , Caliciviridae Infections/pathology , Caliciviridae Infections/virology , Cell Survival , Cytoprotection , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Deletion , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation , Homeostasis , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Necrosis , Norovirus/physiology , Organoids/pathology , Paneth Cells/metabolism , Paneth Cells/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Mol Cell Biol ; 37(24)2017 12 15.
Article En | MEDLINE | ID: mdl-28993480

Autosomal dominant polycystic kidney disease (ADPKD) constitutes the most inherited kidney disease. Mutations in the PKD1 and PKD2 genes, encoding the polycystin 1 and polycystin 2 Ca2+ ion channels, respectively, result in tubular epithelial cell-derived renal cysts. Recent clinical studies demonstrate oxidative stress to be present early in ADPKD. Mitochondria comprise the primary reactive oxygen species source and also their main effector target; however, the pathophysiological role of mitochondria in ADPKD remains uncharacterized. To clarify this function, we examined the mitochondria of cyst-lining cells in ADPKD model mice (Ksp-Cre PKD1flox/flox) and rats (Han:SPRD Cy/+), demonstrating obvious tubular cell morphological abnormalities. Notably, the mitochondrial DNA copy number and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) expression were decreased in ADPKD model animal kidneys, with PGC-1α expression inversely correlated with oxidative stress levels. Consistent with these findings, human ADPKD cyst-derived cells with heterozygous and homozygous PKD1 mutation exhibited morphological and functional abnormalities, including increased mitochondrial superoxide. Furthermore, PGC-1α expression was suppressed by decreased intracellular Ca2+ levels via calcineurin, p38 mitogen-activated protein kinase (MAPK), and nitric oxide synthase deactivation. Moreover, the mitochondrion-specific antioxidant MitoQuinone (MitoQ) reduced intracellular superoxide and inhibited cyst epithelial cell proliferation through extracellular signal-related kinase/MAPK inactivation. Collectively, these results indicate that mitochondrial abnormalities facilitate cyst formation in ADPKD.


Cysts/pathology , Mitochondria/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Calcineurin/metabolism , Calcium/metabolism , Cell Proliferation/physiology , Cells, Cultured , Cysts/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mitochondria/metabolism , Mutation/genetics , Nitric Oxide Synthase/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Rats , Signal Transduction/physiology , Superoxides/metabolism , TRPP Cation Channels/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Kidney Int ; 92(4): 782-784, 2017 10.
Article En | MEDLINE | ID: mdl-28938947

Podocyte biology is an important key factor for the maintenance of glomerular function and structure. Nephronectin is a podocyte-derived extracellular matrix protein in the glomerular basement membrane. Suppression of nephronectin expression by microRNA-378a-3p significantly induced proteinuria due to loss of integrity of the glomerular filtration barrier in zebrafish and mice, demonstrating the relevance of the microRNA-nephronectin axis in regulation of the podocyte-glomerular basement membrane interaction and integrity of the glomerular filtration barrier.


MicroRNAs , Podocytes , Animals , Extracellular Matrix Proteins , Glomerular Basement Membrane , Mice , Proteinuria
17.
Blood Purif ; 43(1-3): 97-100, 2017.
Article En | MEDLINE | ID: mdl-27951582

BACKGROUND: Randomized trials have demonstrated that a phosphate binder ferric citrate (FeC) increases iron parameters in comparison with other phosphate binders, but the doses for FeC to improve iron stores safely have not been clarified. METHODS: We examined changes of iron parameters and blood hemoglobin (Hb) in 7 iron-deficient hemodialysis (HD) patients taking FeC 750 mg/day as a phosphate binder. RESULTS: The median serum transferrin saturation and ferritin increased from 13% (interquartile range (IQR) 7-18) to 28% (IQR 22-31; p = 0.010) and from 17 ng/ml (IQR 11-60) to 106 ng/ml (IQR 58-176; p = 0.015) by 2 and 3 months respectively. With the persistence of these levels thereafter, the FeC administration reduced the usage of erythropoiesis-stimulating agents while maintaining adequate blood Hb levels. CONCLUSION: Oral FeC 750 mg/day improves iron stores without inducing iron overload in hyperphosphatemic HD patients.


Ferric Compounds/administration & dosage , Hyperphosphatemia , Iron/blood , Renal Dialysis/adverse effects , Dose-Response Relationship, Drug , Ferric Compounds/pharmacology , Ferritins/blood , Hematinics/administration & dosage , Hemoglobins/analysis , Humans , Hyperphosphatemia/drug therapy , Transferrin/analysis
19.
Nephrol Dial Transplant ; 31(7): 1062-9, 2016 07.
Article En | MEDLINE | ID: mdl-26333547

Acute kidney injury (AKI) is a common clinical entity that is associated with high mortality and morbidity. It is a risk factor for the development and progression of chronic kidney disease. Presently, no effective treatment for AKI is available, and novel therapeutic approaches are desperately needed. Accumulating evidence highlights mitochondrial dysfunction as an important factor in the pathogenesis of AKI. Recent advances in our understanding of the molecules involved in mitochondrial biogenesis, fusion/fission, mitophagy and their pathophysiological roles will lead to the development of drugs that target mitochondria for the treatment of various diseases, including AKI. In this review, we summarize current knowledge of the contribution of mitochondria-related pathophysiology in AKI and the prospective benefits of mitochondria-targeting therapeutic approaches against AKI.


Acute Kidney Injury/drug therapy , Mitochondria/drug effects , Pharmaceutical Preparations/administration & dosage , Acute Kidney Injury/physiopathology , Animals , Humans , Mitochondria/metabolism , Mitochondria/pathology
20.
J Am Soc Nephrol ; 26(8): 1939-59, 2015 Aug.
Article En | MEDLINE | ID: mdl-25424328

Recent studies have highlighted the renoprotective effect of sirtuin1 (SIRT1), a deacetylase that contributes to cellular regulation. However, the pathophysiologic role of SIRT1 in podocytes remains unclear. Here, we investigated the function of SIRT1 in podocytes. We first established podocyte-specific Sirt1 knockout (SIRT1(pod-/-)) mice. We then induced glomerular disease by nephrotoxic serum injection. The increase in urinary albumin excretion and BUN and the severity of glomerular injury were all significantly greater in SIRT1(pod-/-) mice than in wild-type mice. Western blot analysis and immunofluorescence showed a significant decrease in podocyte-specific proteins in SIRT1(pod-/-) mice, and electron microscopy showed marked exacerbation of podocyte injury, including actin cytoskeleton derangement in SIRT1(pod-/-) mice compared with wild-type mice. Protamine sulfate-induced podocyte injury was also exacerbated by podocyte-specific SIRT1 deficiency. In vitro, actin cytoskeleton derangement in H2O2-treated podocytes became prominent when the cells were pretreated with SIRT1 inhibitors. Conversely, this H2O2-induced derangement was ameliorated by SIRT1 activation. Furthermore, SIRT1 activation deacetylated the actin-binding and -polymerizing protein cortactin in the nucleus and facilitated deacetylated cortactin localization in the cytoplasm. Cortactin knockdown or inhibition of the nuclear export of cortactin induced actin cytoskeleton derangement and dissociation of cortactin from F-actin, suggesting the necessity of cytoplasmic cortactin for maintenance of the actin cytoskeleton. Taken together, these findings indicate that SIRT1 protects podocytes and prevents glomerular injury by deacetylating cortactin and thereby, maintaining actin cytoskeleton integrity.


Actin Cytoskeleton/metabolism , Cortactin/metabolism , Podocytes/physiology , Sirtuin 1/physiology , Animals , Cell Movement , Cell Nucleus/metabolism , Glomerulonephritis/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Podocytes/ultrastructure
...