Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Kidney Int ; 106(1): 24-34, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38614389

ABSTRACT

Kidney epithelial cells have very high energy requirements, which are largely met by fatty acid oxidation. Complex changes in lipid metabolism are observed in patients with kidney disease. Defects in fatty acid oxidation and increased lipid uptake, especially in the context of hyperlipidemia and proteinuria, contribute to this excess lipid build-up and exacerbate kidney disease development. Recent studies have also highlighted the role of increased de novo lipogenesis in kidney fibrosis. The defect in fatty acid oxidation causes energy starvation. Increased lipid uptake, synthesis, and lower fatty acid oxidation can cause toxic lipid build-up, reactive oxygen species generation, and mitochondrial damage. A better understanding of these metabolic processes may open new treatment avenues for kidney diseases by targeting lipid metabolism.


Subject(s)
Fatty Acids , Kidney Tubules , Lipid Metabolism , Humans , Kidney Tubules/metabolism , Kidney Tubules/pathology , Animals , Fatty Acids/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mitochondria/metabolism , Lipogenesis , Oxidation-Reduction , Fibrosis , Reactive Oxygen Species/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Energy Metabolism
2.
J Clin Invest ; 134(4)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38051585

ABSTRACT

Worldwide, over 800 million people are affected by kidney disease, yet its pathogenesis remains elusive, hindering the development of novel therapeutics. In this study, we used kidney-specific expression of quantitative traits and single-nucleus open chromatin analysis to show that genetic variants linked to kidney dysfunction on chromosome 20 target the acyl-CoA synthetase short-chain family 2 (ACSS2). By generating ACSS2-KO mice, we demonstrated their protection from kidney fibrosis in multiple disease models. Our analysis of primary tubular cells revealed that ACSS2 regulated de novo lipogenesis (DNL), causing NADPH depletion and increasing ROS levels, ultimately leading to NLRP3-dependent pyroptosis. Additionally, we discovered that pharmacological inhibition or genetic ablation of fatty acid synthase safeguarded kidney cells against profibrotic gene expression and prevented kidney disease in mice. Lipid accumulation and the expression of genes related to DNL were elevated in the kidneys of patients with fibrosis. Our findings pinpoint ACSS2 as a critical kidney disease gene and reveal the role of DNL in kidney disease.


Subject(s)
Acetate-CoA Ligase , Kidney Diseases , Lipogenesis , Animals , Humans , Mice , Acetate-CoA Ligase/genetics , Fibrosis , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Tubules/metabolism , Lipogenesis/genetics
3.
Nat Metab ; 5(3): 414-430, 2023 03.
Article in English | MEDLINE | ID: mdl-36914909

ABSTRACT

Our understanding of how global changes in cellular metabolism contribute to human kidney disease remains incompletely understood. Here we show that nicotinamide adenine dinucleotide (NAD+) deficiency drives mitochondrial dysfunction causing inflammation and kidney disease development. Using unbiased global metabolomics in healthy and diseased human kidneys, we identify NAD+ deficiency as a disease signature. Furthermore using models of cisplatin- or ischaemia-reperfusion induced kidney injury in male mice we observed NAD+ depletion Supplemental nicotinamide riboside or nicotinamide mononucleotide restores NAD+ levels and improved kidney function. We find that cisplatin exposure causes cytosolic leakage of mitochondrial RNA (mtRNA) and activation of the cytosolic pattern recognition receptor retinoic acid-inducible gene I (RIG-I), both of which can be ameliorated by restoring NAD+. Male mice with RIG-I knock-out (KO) are protected from cisplatin-induced kidney disease. In summary, we demonstrate that the cytosolic release of mtRNA and RIG-I activation is an NAD+-sensitive mechanism contributing to kidney disease.


Subject(s)
Cisplatin , NAD , Animals , Humans , Male , Mice , Cisplatin/toxicity , Dietary Supplements , Inflammation , Kidney/metabolism , NAD/metabolism , RNA, Mitochondrial
4.
J Clin Invest ; 133(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36787250

ABSTRACT

Kidney disease is a major driver of mortality among patients with diabetes and diabetic kidney disease (DKD) is responsible for close to half of all chronic kidney disease cases. DKD usually develops in a genetically susceptible individual as a result of poor metabolic (glycemic) control. Molecular and genetic studies indicate the key role of podocytes and endothelial cells in driving albuminuria and early kidney disease in diabetes. Proximal tubule changes show a strong association with the glomerular filtration rate. Hyperglycemia represents a key cellular stress in the kidney by altering cellular metabolism in endothelial cells and podocytes and by imposing an excess workload requiring energy and oxygen for proximal tubule cells. Changes in metabolism induce early adaptive cellular hypertrophy and reorganization of the actin cytoskeleton. Later, mitochondrial defects contribute to increased oxidative stress and activation of inflammatory pathways, causing progressive kidney function decline and fibrosis. Blockade of the renin-angiotensin system or the sodium-glucose cotransporter is associated with cellular protection and slowing kidney function decline. Newly identified molecular pathways could provide the basis for the development of much-needed novel therapeutics.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Humans , Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Kidney/metabolism , Podocytes/metabolism , Albuminuria
5.
Nephrology (Carlton) ; 28(1): 60-71, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36217796

ABSTRACT

AIM: Podocytes, a vital component of the glomerular filtration barrier, are vulnerable to various noxious stimuli, including Hypoxic. HIF1α that transduces hypoxic adaptations induces Transglutaminase 2 (TG2), which catalyses cross-linking of extracellular matrix proteins. In this study, we investigated the mechanism of regulation of TG2 by HIF1α. METHODS: HIF1α was induced in podocytes by treating with FG4592 (Roxadustat) or hypoxia (1% oxygen) and in mice by treating with FG4592. Gene expression and protein analysis of ZEB2, TRPC6 and TG2 were performed in both experimental models. Histological and kidney function analyses were performed in mice. RESULTS: Data mining revealed co-expression of HIF1α, ZEB2, TRPC6 and TG2 in the chronic kidney diseases (CKD)-validated dataset. We observed elevated expression of ZEB2, TRPC6 and TG2 in FG4592-treated podocytes. Ectopic expression of ZEB2 resulted in high TRPC6 expression, elevated intracellular calcium levels and increased TG2 activity. Blocking the TRPC6 channel or inhibiting its expression partially attenuated FG4592-induced TG2 activity, whereas suppression of ZEB2 expression significantly abolished TG2 activity. Furthermore, we noticed the induction of the ZEB2/TRPC6/TG2 axis in podocytes in mice administered with FG-4592. Metformin ameliorated the HIF1α-induced podocyte injury and proteinuria in mice administered with FG-4592. CONCLUSION: This study demonstrates that HIF1α stimulates both TG2 expression and activity via ZEB2/TRPC6 axis, whereas abrogation of HIF1α by metformin prevented hypoxia-induced glomerular injury. Metformin could be explored to treat proteinuric diseases such as CKD, sleep apnea and renal Ischemia-reperfusion-injury, where hypoxia is considered a risk factor.


Subject(s)
Metformin , Podocytes , Renal Insufficiency, Chronic , Mice , Animals , Podocytes/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism , Metformin/pharmacology , Protein Glutamine gamma Glutamyltransferase 2 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/prevention & control , Renal Insufficiency, Chronic/metabolism
6.
Biochim Biophys Acta Mol Cell Res ; 1870(2): 119391, 2023 02.
Article in English | MEDLINE | ID: mdl-36400249

ABSTRACT

Pituitary growth hormone (GH) is essential for growth, metabolism, and renal function. Overactive GH signaling is associated with impaired kidney function. Glomerular podocytes, a key kidney cell type, play an indispensable role in the renal filtration and express GH receptors (GHR), suggesting the direct action of GH on these cells. However, the precise mechanism and the downstream signaling events by which GH leads to diabetic nephropathy remain to be elucidated. Here we performed proteome analysis of the condition media from human podocytes and confirmed that GH-induces TGF-ß1. Inhibition of GH/GHR stimulated-JAK2 signaling abrogates GH-induced TGF-ß1 secretion. Mice administered with GH showed glomerular manifestations concomitant with proteinuria. Pharmacological inhibition of TGF-ßR1 in mice prevented GH-induced TGF-ß dependent SMAD signaling and proteinuria. Conditional deletion of GHR in podocytes protected mice from streptozotocin-induced diabetic nephropathy. GH and TGF-ß1 signaling components expression was elevated in the kidneys of human diabetic nephropathy patients. Our study identifies that GH induces TGF-ß1 in podocytes, contributing to diabetic nephropathy.


Subject(s)
Diabetic Nephropathies , Human Growth Hormone , Podocytes , Humans , Mice , Animals , Podocytes/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Growth Hormone/genetics , Growth Hormone/metabolism , Growth Hormone/pharmacology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Human Growth Hormone/genetics , Human Growth Hormone/pharmacology , Human Growth Hormone/metabolism , Proteinuria/genetics , Proteinuria/metabolism
7.
FASEB J ; 36(12): e22622, 2022 12.
Article in English | MEDLINE | ID: mdl-36421039

ABSTRACT

Diabetes shortens the life expectancy by more than a decade, and the excess mortality in diabetes is correlated with the incidence of kidney disease. Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease. Macrophage accumulation predicts the severity of kidney injury in human biopsies and experimental models of DKD. However, the mechanism underlying macrophage recruitment in diabetes glomeruli is unclear. Elevated plasma growth hormone (GH) levels in type I diabetes and acromegalic individuals impaired glomerular biology. In this study, we examined whether GH-stimulated podocytes contribute to macrophage accumulation. RNA-seq analysis revealed elevated TNF-α signaling in GH-treated human podocytes. Conditioned media from GH-treated podocytes (GH-CM) induced differentiation of monocytes to macrophages. On the other hand, neutralization of GH-CM with the TNF-α antibody diminished GH-CM's action on monocytes. The treatment of mice with GH resulted in increased macrophage recruitment, podocyte injury, and proteinuria. Furthermore, we noticed the activation of TNF-α signaling, macrophage accumulation, and fibrosis in DKD patients' kidney biopsies. Our findings suggest that podocytes could secrete TNF-α and contribute to macrophage migration, resulting in DKD-related renal inflammation. Inhibition of either GH action or TNF-α expression in podocytes could be a novel therapeutic approach for DKD treatment.


Subject(s)
Diabetic Nephropathies , Monocytes , Podocytes , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Monocytes/cytology , Podocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Differentiation
8.
Nat Commun ; 13(1): 4018, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35821371

ABSTRACT

The kidney has tremendous capacity to repair after acute injury, however, pathways guiding adaptive and fibrotic repair are poorly understood. We developed a model of adaptive and fibrotic kidney regeneration by titrating ischemic injury dose. We performed detailed biochemical and histological analysis and profiled transcriptomic changes at bulk and single-cell level (> 110,000 cells) over time. Our analysis highlights kidney proximal tubule cells as key susceptible cells to injury. Adaptive proximal tubule repair correlated with fatty acid oxidation and oxidative phosphorylation. We identify a specific maladaptive/profibrotic proximal tubule cluster after long ischemia, which expresses proinflammatory and profibrotic cytokines and myeloid cell chemotactic factors. Druggability analysis highlights pyroptosis/ferroptosis as vulnerable pathways in these profibrotic cells. Pharmacological targeting of pyroptosis/ferroptosis in vivo pushed cells towards adaptive repair and ameliorates fibrosis. In summary, our single-cell analysis defines key differences in adaptive and fibrotic repair and identifies druggable pathways for pharmacological intervention to prevent kidney fibrosis.


Subject(s)
Acute Kidney Injury , Kidney , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Fibrosis , Humans , Kidney/metabolism , Regeneration , Single-Cell Analysis
9.
Compr Physiol ; 12(2): 3167-3192, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35578969

ABSTRACT

The increased production of derivatives of molecular oxygen and nitrogen in the form of reactive oxygen species (ROS) and reactive nitrogen species (RNS) lead to molecular damage called oxidative stress. Under normal physiological conditions, the ROS generation is tightly regulated in different cells and cellular compartments. Any disturbance in the balance between the cellular generation of ROS and antioxidant balance leads to oxidative stress. In this article, we discuss the sources of ROS (endogenous and exogenous) and antioxidant mechanisms. We also focus on the pathophysiological significance of oxidative stress in various cell types of the liver. Oxidative stress is implicated in the development and progression of various liver diseases. We narrate the master regulators of ROS-mediated signaling and their contribution to liver diseases. Nonalcoholic fatty liver diseases (NAFLD) are influenced by a "multiple parallel-hit model" in which oxidative stress plays a central role. We highlight the recent findings on the role of oxidative stress in the spectrum of NAFLD, including fibrosis and liver cancer. Finally, we provide a brief overview of oxidative stress biomarkers and their therapeutic applications in various liver-related disorders. Overall, the article sheds light on the significance of oxidative stress in the pathophysiology of the liver. © 2022 American Physiological Society. Compr Physiol 12:3167-3192, 2022.


Subject(s)
Antioxidants , Non-alcoholic Fatty Liver Disease , Antioxidants/metabolism , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
Cell Mol Gastroenterol Hepatol ; 13(4): 1057-1072, 2022.
Article in English | MEDLINE | ID: mdl-34902628

ABSTRACT

BACKGROUND & AIMS: Compelling evidence shows that glucagon-like peptide-1 (GLP-1) has a profound effect in restoring normoglycemia in type 2 diabetic patients by increasing pancreatic insulin secretion. Although L-cells are the primary source of circulating GLP-1, the current therapies do not target L-cells to increase GLP-1 levels. Our study aimed to determine the molecular underpinnings of GLP-1 secretion as an impetus to identify new interventions to target endogenous L-cells. METHODS: We used genetic mouse models of intestine-specific overexpression of hypoxia-inducible factor (HIF)-1α and HIF-2α (VhlΔIE), conditional overexpression of intestinal HIF-2α (Hif-2αLSL;Vilin-Cre/ERT2), and intestine-specific HIF-2α knockout mice (Hif-2αΔIE) to show that HIF signaling, especially HIF-2α, regulates GLP-1 secretion. RESULTS: Our data show that intestinal HIF signaling improved glucose homeostasis in a GLP-1-dependent manner. Intestinal HIF potentiated GLP-1 secretion via the lipid sensor G-protein-coupled receptor (GPR)40 enriched in L-cells. We show that HIF-2α regulates GPR40 in L-cells and potentiates fatty acid-induced GLP-1 secretion via extracellular regulated kinase (ERK). Using a genetic model of intestine-specific overexpression of HIF-2α, we show that HIF-2α is sufficient to increase GLP-1 levels and attenuate diet-induced metabolic perturbations such as visceral adiposity, glucose intolerance, and hepatic steatosis. Lastly, we show that intestinal HIF-2α signaling acts as a priming mechanism crucial for postprandial lipid-mediated GLP-1 secretion. Thus, disruption of intestinal HIF-2α decreases GLP-1 secretion. CONCLUSIONS: In summary, we show that intestinal HIF signaling, particularly HIF-2α, regulates the lipid sensor GPR40, which is crucial for the lipid-mediated GLP-1 secretion, and suggest that HIF-2α is a potential target to induce endogenous GLP-1 secretion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucagon-Like Peptide 1 , Intestines , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Glucagon-Like Peptide 1/metabolism , Humans , L Cells , Lipids , Mice
11.
Immunity ; 54(11): 2632-2649.e6, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34715018

ABSTRACT

The incidence and severity of sepsis is higher among individuals of African versus European ancestry. We found that genetic risk variants (RVs) in the trypanolytic factor apolipoprotein L1 (APOL1), present only in individuals of African ancestry, were associated with increased sepsis incidence and severity. Serum APOL1 levels correlated with sepsis and COVID-19 severity, and single-cell sequencing in human kidneys revealed high expression of APOL1 in endothelial cells. Analysis of mice with endothelial-specific expression of RV APOL1 and in vitro studies demonstrated that RV APOL1 interfered with mitophagy, leading to cytosolic release of mitochondrial DNA and activation of the inflammasome (NLRP3) and the cytosolic nucleotide sensing pathways (STING). Genetic deletion or pharmacological inhibition of NLRP3 and STING protected mice from RV APOL1-induced permeability defects and proinflammatory endothelial changes in sepsis. Our studies identify the inflammasome and STING pathways as potential targets to reduce APOL1-associated health disparities in sepsis and COVID-19.


Subject(s)
Apolipoprotein L1/genetics , Black People/genetics , COVID-19/genetics , Genetic Predisposition to Disease/genetics , Sepsis/genetics , Animals , Apolipoprotein L1/blood , Black People/statistics & numerical data , COVID-19/pathology , DNA, Mitochondrial/metabolism , Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitophagy/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Risk Factors , Sepsis/pathology , Severity of Illness Index , White People/genetics , White People/statistics & numerical data
12.
Cell Death Dis ; 12(4): 342, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795655

ABSTRACT

Glomerular podocytes are integral members of the glomerular filtration barrier in the kidney and are crucial for glomerular permselectivity. These highly differentiated cells are vulnerable to an array of noxious stimuli that prevail in several glomerular diseases. Elevated circulating growth hormone (GH) levels are associated with podocyte injury and proteinuria in diabetes. However, the precise mechanism(s) by which excess GH elicits podocytopathy remains to be elucidated. Previous studies have shown that podocytes express GH receptor (GHR) and induce Notch signaling when exposed to GH. In the present study, we demonstrated that GH induces TGF-ß1 signaling and provokes cell cycle reentry of otherwise quiescent podocytes. Though differentiated podocytes reenter the cell cycle in response to GH and TGF-ß1, they cannot accomplish cytokinesis, despite karyokinesis. Owing to this aberrant cell cycle event, GH- or TGF-ß1-treated cells remain binucleated and undergo mitotic catastrophe. Importantly, inhibition of JAK2, TGFBR1 (TGF-ß receptor 1), or Notch prevented cell cycle reentry of podocytes and protected them from mitotic catastrophe associated with cell death. Inhibition of Notch activation prevents GH-dependent podocyte injury and proteinuria. Similarly, attenuation of GHR expression abated Notch activation in podocytes. Kidney biopsy sections from patients with diabetic nephropathy (DN) show activation of Notch signaling and binucleated podocytes. These data indicate that excess GH induced TGF-ß1-dependent Notch1 signaling contributes to the mitotic catastrophe of podocytes. This study highlights the role of aberrant GH signaling in podocytopathy and the potential application of TGF-ß1 or Notch inhibitors, as a therapeutic agent for DN.


Subject(s)
Cell Cycle/drug effects , Growth Hormone/pharmacology , Kidney Glomerulus/drug effects , Podocytes/drug effects , Proteinuria/drug therapy , Cell Death/drug effects , Cell Differentiation/drug effects , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Growth Hormone/metabolism , Humans , Kidney Glomerulus/metabolism , Mitosis/drug effects , Podocytes/metabolism , Proteinuria/metabolism , Signal Transduction/drug effects
13.
Kidney Int ; 98(5): 1079-1081, 2020 11.
Article in English | MEDLINE | ID: mdl-33126968

ABSTRACT

Aging is the strongest independent risk factor for chronic kidney disease. Glomerular epithelial cells are unable to proliferate and have limited ability to renew; therefore, podocytes must maintain a delicate intracellular homeostasis that enables them to function and adapt to stress endured during the human life span. Here, Wang et al. performed unbiased transcriptomic analysis of aging podocytes and identified important novel regulators.


Subject(s)
Kidney Diseases , Podocytes , Aging/genetics , Animals , Humans , Kidney Glomerulus , Mice , Transcriptome
14.
J Cell Sci ; 133(18)2020 09 15.
Article in English | MEDLINE | ID: mdl-32843579

ABSTRACT

Emerging evidence indicates that proper mitochondrial dynamics are critical for adipocyte differentiation and functional thermogenic capacity. We found that the mitochondrial fission protein dynamin-related protein 1 (DRP1, also known as DNML1) is highly expressed in brown adipose tissue compared to expression in white adipose tissue, and these expression levels increase during brown adipocyte differentiation. Our results reveal that the inhibition of DRP1 using mdivi-1 mitigates beige adipocyte differentiation and differentiation-associated mitochondrial biogenesis. We found that DRP1 is essential for the induction of the early-phase beige adipogenic transcriptional program. Intriguingly, inhibition of DRP1 is dispensable following the induction of beige adipogenesis and adipogenesis-associated mitochondrial biogenesis. Altogether, we demonstrate that DRP1 in preadipocytes plays an essential role in beige and brown adipogenesis.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adipogenesis , Adipose Tissue, Brown , Adipogenesis/genetics , Adipose Tissue, White , Cell Differentiation , Dynamins/genetics , Humans , Thermogenesis
15.
Metabolism ; 107: 154225, 2020 06.
Article in English | MEDLINE | ID: mdl-32275973

ABSTRACT

BACKGROUND: Caloric restriction (CR) delays the onset of metabolic and age-related disorders. Recent studies have demonstrated that formation of beige adipocytes induced by CR is strongly associated with extracellular remodeling in adipose tissue, decrease in adipose tissue inflammation, and improved systemic metabolic homeostasis. However, beige adipocytes rapidly transition to white upon CR withdrawal through unclear mechanisms. MATERIALS AND METHODS: Six-week old C57BL6 mice were fed with 40% CR chow diet for 6 weeks. Subsequently, one group of mice was switched back to ad libitum chow diet, which was continued for additional 2 weeks. Adipose tissues were assessed histologically and biochemically for beige adipocytes. RESULTS: Beige adipocytes induced by CR rapidly transition to white adipocytes when CR is withdrawn independent of parkin-mediated mitophagy. We demonstrate that the involution of mitochondria during CR withdrawal is strongly linked with a decrease in mitochondrial biogenesis. We further demonstrate that beige-to-white fat transition upon ß3-AR agonist-withdrawal could be attenuated by CR, partly via maintenance of mitochondrial biogenesis. CONCLUSION: In the model of CR, our study highlights the dominant role of mitochondrial biogenesis in the maintenance of beige adipocytes. We propose that loss of beige adipocytes upon ß3-AR agonist withdrawal could be attenuated by CR.


Subject(s)
Adipocytes, Beige/physiology , Caloric Restriction , Organelle Biogenesis , Adipocytes, White/physiology , Adipose Tissue/cytology , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Body Composition , Cell Fusion , Diet , Insulin/blood , Mice , Mice, Inbred C57BL , Mitophagy , Oxygen Consumption , Primary Cell Culture
16.
Hepatology ; 72(6): 2165-2181, 2020 12.
Article in English | MEDLINE | ID: mdl-32190913

ABSTRACT

BACKGROUND AND AIMS: Hepatic crisis is an emergent complication affecting patients with sickle cell disease (SCD); however, the molecular mechanism of sickle cell hepatobiliary injury remains poorly understood. Using the knock-in humanized mouse model of SCD and SCD patient blood, we sought to mechanistically characterize SCD-associated hepato-pathophysiology applying our recently developed quantitative liver intravital imaging, RNA sequence analysis, and biochemical approaches. APPROACH AND RESULTS: SCD mice manifested sinusoidal ischemia, progressive hepatomegaly, liver injury, hyperbilirubinemia, and increased ductular reaction under basal conditions. Nuclear factor kappa B (NF-κB) activation in the liver of SCD mice inhibited farnesoid X receptor (FXR) signaling and its downstream targets, leading to loss of canalicular bile transport and altered bile acid pool. Intravital imaging revealed impaired bile secretion into the bile canaliculi, which was secondary to loss of canalicular bile transport and bile acid metabolism, leading to intrahepatic bile accumulation in SCD mouse liver. Blocking NF-κB activation rescued FXR signaling and partially ameliorated liver injury and sinusoidal ischemia in SCD mice. CONCLUSIONS: These findings identify that NF-κB/FXR-dependent impaired bile secretion promotes intrahepatic bile accumulation, which contributes to hepatobiliary injury of SCD. Improved understanding of these processes could potentially benefit the development of therapies to treat sickle cell hepatic crisis.


Subject(s)
Anemia, Sickle Cell/complications , Bile/metabolism , Cholestasis/etiology , Hepatic Insufficiency/etiology , Liver/pathology , Adolescent , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Bile Ducts, Intrahepatic/diagnostic imaging , Bile Ducts, Intrahepatic/pathology , Cholestasis/pathology , Cholestasis/prevention & control , Disease Models, Animal , Female , Gene Knock-In Techniques , Hemoglobin, Sickle/genetics , Hepatic Insufficiency/pathology , Hepatic Insufficiency/prevention & control , Humans , Intravital Microscopy , Liver/diagnostic imaging , Male , Mice , Middle Aged , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Young Adult
17.
Sci Rep ; 9(1): 17897, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784544

ABSTRACT

Podocytes are specialized cells of the glomerulus and key component of the glomerular filtration apparatus (GFA). GFA regulates the permselectivity and ultrafiltration of blood. The mechanism by which the integrity of the GFA is compromised and manifest in proteinuria during ischemic stroke remains enigmatic. We investigated the mechanism of ischemic hypoxia-induced proteinuria in a middle cerebral artery occlusion (MCAO) model. Ischemic hypoxia resulted in the accumulation of HIF1α in the podocytes that resulted in the increased expression of ZEB2 (Zinc finger E-box-binding homeobox 2). ZEB2, in turn, induced TRPC6 (transient receptor potential cation channel, subfamily C, member 6), which has increased selectivity for calcium. Elevated expression of TRPC6 elicited increased calcium influx and aberrant activation of focal adhesion kinase (FAK) in podocytes. FAK activation resulted in the stress fibers reorganization and podocyte foot process effacement. Our study suggests overactive HIF1α/ZEB2 axis during ischemic-hypoxia raises intracellular calcium levels via TRPC6 and consequently altered podocyte structure and function thus contributes to proteinuria.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infarction, Middle Cerebral Artery/pathology , Proteinuria/etiology , TRPC Cation Channels/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Actin Cytoskeleton , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , Disease Models, Animal , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Permeability/drug effects , Phosphorylation , Podocytes/cytology , Podocytes/metabolism , Rats , Rats, Sprague-Dawley
18.
J Biol Chem ; 294(44): 16109-16122, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31511328

ABSTRACT

Growth hormone (GH) plays a significant role in normal renal function and overactive GH signaling has been implicated in proteinuria in diabetes and acromegaly. Previous results have shown that the glomerular podocytes, which play an essential role in renal filtration, express the GH receptor, suggesting the direct action of GH on these cells. However, the exact mechanism and the downstream pathways by which excess GH leads to diabetic nephropathy is not established. In the present article, using immortalized human podocytes in vitro and a mouse model in vivo, we show that excess GH activates Notch1 signaling in a γ-secretase-dependent manner. Pharmacological inhibition of Notch1 by γ-secretase inhibitor DAPT (N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenyl glycine t-butylester) abrogates GH-induced epithelial to mesenchymal transition (EMT) and is associated with a reduction in podocyte loss. More importantly, our results show that DAPT treatment blocks cytokine release and prevents glomerular fibrosis, all of which are induced by excess GH. Furthermore, DAPT prevented glomerular basement membrane thickening and proteinuria induced by excess GH. Finally, using kidney biopsy sections from people with diabetic nephropathy, we show that Notch signaling is indeed up-regulated in such settings. All these results confirm that excess GH induces Notch1 signaling in podocytes, which contributes to proteinuria through EMT as well as renal fibrosis. Our studies highlight the potential application of γ-secretase inhibitors as a therapeutic target in people with diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Growth Hormone/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cells, Cultured , Cytokines/metabolism , Diabetic Nephropathies/drug therapy , Dipeptides/pharmacology , Dipeptides/therapeutic use , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Podocytes/drug effects , Podocytes/pathology , Proteinuria/drug therapy , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics
19.
J Cell Physiol ; 234(5): 6503-6518, 2019 05.
Article in English | MEDLINE | ID: mdl-30238984

ABSTRACT

The glomerular filtration barrier (GFB) plays a critical role in ensuing protein free urine. The integrity of the GFB is compromised during hypoxia that prevails during extreme physiological conditions. However, the mechanism by which glomerular permselectivity is compromised during hypoxia remains enigmatic. Rats exposed to hypoxia showed a decreased glomerular filtration rate, podocyte foot-processes effacement, and proteinuria. Accumulation of hypoxia-inducible factor-1α (HIF1α) in podocytes resulted in elevated expression of zinc finger E-box binding homeobox 2 (ZEB2) and decreased expression of E- and P-cadherin. We also demonstrated that HIF1α binds to hypoxia response element localized in the ZEB2 promoter. Furthermore, HIF1α also induced the expression of ZEB2-natural antisense transcript, which is known to increase the efficiency of ZEB2 translation. Ectopic expression of ZEB2 induced loss of E- and P-cadherin and is associated with enhanced motility of podocytes during hypoxic conditions. ZEB2 knockdown abrogated hypoxia-induced decrease in podocyte permselectivity. This study suggests that hypoxia leads to activation of HIF1α-ZEB2 axis, resulting in podocyte injury and poor renal outcome.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Proteinuria/physiopathology , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Hypoxia/metabolism , Hypoxia/physiopathology , Podocytes/pathology , Rats , Rats, Wistar
20.
J Cell Biochem ; 120(5): 7667-7678, 2019 May.
Article in English | MEDLINE | ID: mdl-30387200

ABSTRACT

Glomerular podocytes are the major components of the renal filtration barrier, and altered podocyte permselectivity is a key event in the pathogenesis of proteinuric conditions. Clinical conditions such as ischemia and sleep apnea and extreme physiological conditions such as high-altitude sickness are presented with renal hypoxia and are associated with significant proteinuria. Hypoxia is considered as an etiological factor in the progression of acute renal injury. A sustained increase in hypoxia-inducible factor 1α (HIF1α) is a major adaptive stimulus to the hypoxic conditions. Although the temporal association between hypoxia and proteinuria is known, the mechanism by which hypoxia elicits proteinuria remains to be investigated. Furthermore, stabilization of HIF1α is being considered as a therapeutic option to treat anemia in patients with chronic kidney disease. Therefore, in this study, we induced stabilization of HIF1α in glomerular regions in vivo and in podocytes in vitro upon exposure to cobalt chloride. The elevated HIF1α expression is concurrence with diminished expression of nephrin and podocin, podocyte foot-processes effacement, and significant proteinuria. Podocytes exposed to cobalt chloride lost their arborized morphology and cell-cell connections and also displayed cytoskeletal derangements. Elevation in expression of HIF1α is in concomitance with loss of nephrin and podocin in patients with diabetic nephropathy and chronic kidney disease. In summary, the current study suggests that HIF1α stabilization impairs podocyte function vis-à-vis glomerular permselectivity.

SELECTION OF CITATIONS
SEARCH DETAIL
...