Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Immunity ; 57(1): 68-85.e11, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38141610

ABSTRACT

Tissue factor (TF), which is a member of the cytokine receptor family, promotes coagulation and coagulation-dependent inflammation. TF also exerts protective effects through unknown mechanisms. Here, we showed that TF bound to interferon-α receptor 1 (IFNAR1) and antagonized its signaling, preventing spontaneous sterile inflammation and maintaining immune homeostasis. Structural modeling and direct binding studies revealed binding of the TF C-terminal fibronectin III domain to IFNAR1, which restricted the expression of interferon-stimulated genes (ISGs). Podocyte-specific loss of TF in mice (PodΔF3) resulted in sterile renal inflammation, characterized by JAK/STAT signaling, proinflammatory cytokine expression, disrupted immune homeostasis, and glomerulopathy. Inhibiting IFNAR1 signaling or loss of Ifnar1 expression in podocytes attenuated these effects in PodΔF3 mice. As a heteromer, TF and IFNAR1 were both inactive, while dissociation of the TF-IFNAR1 heteromer promoted TF activity and IFNAR1 signaling. These data suggest that the TF-IFNAR1 heteromer is a molecular switch that controls thrombo-inflammation.


Subject(s)
Signal Transduction , Thromboplastin , Animals , Mice , Inflammation , Interferon-alpha , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Thromboplastin/genetics
2.
Kidney Int ; 105(1): 65-83, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37774921

ABSTRACT

Glomerular-tubular crosstalk within the kidney has been proposed, but the paracrine signals enabling this remain largely unknown. The cold-shock protein Y-box binding protein 1 (YBX1) is known to regulate inflammation and kidney diseases but its role in podocytes remains undetermined. Therefore, we analyzed mice with podocyte specific Ybx1 deletion (Ybx1ΔPod). Albuminuria was increased in unchallenged Ybx1ΔPod mice, which surprisingly was associated with reduced glomerular, but enhanced tubular damage. Tubular toll-like receptor 4 (TLR4) expression, node-like receptor protein 3 (NLRP3) inflammasome activation and kidney inflammatory cell infiltrates were all increased in Ybx1ΔPod mice. In vitro, extracellular YBX1 inhibited NLRP3 inflammasome activation in tubular cells. Co-immunoprecipitation, immunohistochemical analyses, microscale cell-free thermophoresis assays, and blunting of the YBX1-mediated TLR4-inhibition by a unique YBX1-derived decapeptide suggests a direct interaction of YBX1 and TLR4. Since YBX1 can be secreted upon post-translational acetylation, we hypothesized that YBX1 secreted from podocytes can inhibit TLR4 signaling in tubular cells. Indeed, mice expressing a non-secreted YBX1 variant specifically in podocytes (Ybx1PodK2A mice) phenocopied Ybx1ΔPod mice, demonstrating a tubular-protective effect of YBX1 secreted from podocytes. Lipopolysaccharide-induced tubular injury was aggravated in Ybx1ΔPod and Ybx1PodK2A mice, indicating a pathophysiological relevance of this glomerular-tubular crosstalk. Thus, our data show that YBX1 is physiologically secreted from podocytes, thereby negatively modulating sterile inflammation in the tubular compartment, apparently by binding to and inhibiting tubular TLR4 signaling. Hence, we have uncovered an YBX1-dependent molecular mechanism of glomerular-tubular crosstalk.


Subject(s)
Kidney Diseases , Podocytes , Mice , Animals , Inflammasomes/metabolism , Toll-Like Receptor 4/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cold-Shock Response , Kidney/metabolism , Podocytes/metabolism , Kidney Diseases/metabolism , Inflammation/metabolism
3.
Kidney Int ; 103(2): 304-319, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36309126

ABSTRACT

Dysfunction of mesangial cells plays a major role in the pathogenesis of diabetic kidney disease (DKD), the leading cause of kidney failure. However, the underlying molecular mechanisms are incompletely understood. By unbiased gene expression analysis of glucose-exposed mesangial cells, we identified the transmembrane receptor CD248 as the most upregulated gene, and the maladaptive unfolded protein response (UPR) as one of the most stimulated pathways. Upregulation of CD248 was further confirmed in glucose-stressed mesangial cells in vitro, in kidney glomeruli isolated from diabetic mice (streptozotocin; STZ and db/db models, representing type 1 and type 2 diabetes mellitus, respectively) in vivo, and in glomerular kidney sections from patients with DKD. Time course analysis revealed that glomerular CD248 induction precedes the onset of albuminuria, mesangial matrix expansion and maladaptive UPR activation (hallmarked by transcription factor C/EBP homologous protein (CHOP) induction) but is paralleled by loss of the adaptive UPR regulator spliced X box binding protein (XBP1). Mechanistically, CD248 promoted maladaptive UPR signaling via inhibition of the inositol requiring enzyme 1α (IRE1α)-mediated transcription factor XBP1 splicing in vivo and in vitro. CD248 induced a multiprotein complex comprising heat shock protein 90, BH3 interacting domain death agonist (BID) and IRE1α, in which BID impedes IRE1α-mediated XBP1 splicing and induced CHOP mediated maladaptive UPR signaling. While CD248 knockout ameliorated DKD-associated glomerular dysfunction and reverses maladaptive unfolded protein response signaling, concomitant XBP1 deficiency abolished the protective effect in diabetic CD248 knockout mice, supporting a functional interaction of CD248 and XBP1 in vivo. Hence, CD248 is a novel mesangial cell receptor inducing maladaptive UPR signaling in DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Animals , Mice , Antigens, CD/metabolism , Antigens, Neoplasm , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Unfolded Protein Response , Humans
4.
BMC Plant Biol ; 23(1): 629, 2023 Dec 08.
Article in English | MEDLINE | ID: mdl-38062351

ABSTRACT

Chromium (Cr) toxicity significantly threatens sunflower growth and productivity by interfering with enzymatic activity and generating reactive oxygen species (ROS). Zinc quantum dot biochar (ZQDB) and arbuscular mycorrhizal fungi (AMF) have become popular to resolve this issue. AMF can facilitate root growth, while biochar tends to minimize Cr mobility in soil. The current study aimed to explore AMF and ZQDB combined effects on sunflower plants in response to Cr toxicity. Four treatments were applied, i.e. NoAMF + NoZQDB, AMF + 0.40%ZQDB, AMF + 0.80%ZQDB, and AMF + 1.20%ZQDB, under different stress levels of Cr, i.e. no Cr (control), 150 and 200 mg Cr/kg soil. Results showed that AMF + 1.20%ZQDB was the treatment that caused the greatest improvement in plant height, stem diameter, head diameter, number of leaves per plant, achenes per head, 1000 achenes weight, achene yield, biological yield, transpiration rate, stomatal conductance, chlorophyll content and oleic acid, relative to the condition NoAMF + No ZQDB at 200 mg Cr/kg soil. A significant decline in peroxidase (POD), superoxide dismutase (SOD), and catalase (CAT) while improvement in ascorbate peroxidase (APx), oil content, and protein content further supported the effectiveness of AMF + 1.20%ZQDB against Cr toxicity. Our results suggest that the treatment AMF + 1.20%ZQDB can efficiently alleviate Cr stress in sunflowers.


Subject(s)
Helianthus , Mycorrhizae , Quantum Dots , Mycorrhizae/physiology , Antioxidants/metabolism , Helianthus/metabolism , Chromium/toxicity , Chromium/metabolism , Soil , Plant Roots/metabolism
5.
Circ Res ; 128(4): 513-529, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33353373

ABSTRACT

RATIONALE: While thrombin is the key protease in thrombus formation, other coagulation proteases, such as fXa (factor Xa) or aPC (activated protein C), independently modulate intracellular signaling via partially distinct receptors. OBJECTIVES: To study the differential effects of fXa or fIIa (factor IIa) inhibition on gene expression and inflammation in myocardial ischemia-reperfusion injury. METHODS AND RESULTS: Mice were treated with a direct fIIa inhibitor (fIIai) or direct fXa inhibitor (fXai) at doses that induced comparable anticoagulant effects ex vivo and in vivo (tail-bleeding assay and FeCl3-induced thrombosis). Myocardial ischemia-reperfusion injury was induced via left anterior descending ligation. We determined infarct size and in vivo aPC generation, analyzed gene expression by RNA sequencing, and performed immunoblotting and ELISA. The signaling-only 3K3A-aPC variant and inhibitory antibodies that blocked all or only the anticoagulant function of aPC were used to determine the role of aPC. Doses of fIIai and fXai that induced comparable anticoagulant effects resulted in a comparable reduction in infarct size. However, unbiased gene expression analyses revealed marked differences, including pathways related to sterile inflammation and inflammasome regulation. fXai but not fIIai inhibited sterile inflammation by reducing the expression of proinflammatory cytokines (IL [interleukin]-1ß, IL-6, and TNFα [tumor necrosis factor alpha]), as well as NF-κB (nuclear factor kappa B) and inflammasome activation. This anti-inflammatory effect was associated with reduced myocardial fibrosis 28 days post-myocardial ischemia-reperfusion injury. Mechanistically, in vivo aPC generation was higher with fXai than with fIIai. Inhibition of the anticoagulant and signaling properties of aPC abolished the anti-inflammatory effect associated with fXai, while inhibiting only the anticoagulant function of aPC had no effect. Combining 3K3A-aPC with fIIai reduced the inflammatory response, mimicking the fXai-associated effect. CONCLUSIONS: We showed that specific inhibition of coagulation via direct oral anticoagulants had differential effects on gene expression and inflammation, despite comparable anticoagulant effects and infarct sizes. Targeting individual coagulation proteases induces specific cellular responses unrelated to their anticoagulant effect.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Factor Xa Inhibitors/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Protein C/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Factor Xa Inhibitors/pharmacology , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Protein C/pharmacology
6.
Kidney Int ; 102(4): 766-779, 2022 10.
Article in English | MEDLINE | ID: mdl-35779608

ABSTRACT

Efficient therapies for diabetic kidney disease (DKD), now the leading cause of kidney failure, are lacking. One hallmark of DKD is sterile inflammation (inflammation in absence of microorganisms), but the underlying molecular mechanisms remain poorly understood. The NLRP3 inflammasome (innate immune system receptors and sensors regulating activation of caspase-1) is a mechanism of sterile inflammation known to be activated by metabolic stimuli and reactive metabolites associated with DKD, including inflammasome activation in podocytes. However, whether NLRP3 inflammasome activation in podocytes contributes to sterile inflammation and glomerular damage in DKD remains unknown. Here, we found that kidney damage, as reflected by increased albuminuria, glomerular mesangial expansion and glomerular basement membrane thickness was aggravated in hyperglycemic mice with podocyte-specific expression of an Nlrp3 gain-of-function mutant (Nlrp3A350V). In contrast, hyperglycemic mice with podocyte-specific Nlrp3 or Caspase-1 deficiency showed protection against DKD. Intriguingly, podocyte-specific Nlrp3 deficiency was fully protective, while podocyte-specific caspase-1 deficiency was only partially protective. Podocyte-specific Nlrp3, but not caspase-1 deficiency, maintained glomerular autophagy in hyperglycemic mice, suggesting that podocyte Nlrp3 exerts both canonical and non-canonical effects. Thus, podocyte NLRP3 inflammasome activation is both sufficient and required for DKD and supports the concept that podocytes exert some immune cell-like functions. Hence, as podocyte NLRP3 exerts non-canonical and canonical effects, targeting NLRP3 may be a promising therapeutic approach in DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Animals , Diabetes Mellitus/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Mice , Mice, Obese , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Podocytes/metabolism
7.
J Am Soc Nephrol ; 32(12): 3066-3079, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34479965

ABSTRACT

BACKGROUND: Maladaptive endoplasmic reticulum stress signaling in diabetic kidney disease (DKD) is linked to increased glomerular and tubular expression of the cell-death-promoting transcription factor C/EBP homologous protein (CHOP). Here, we determined whether locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) targeting CHOP ameliorate experimental DKD. METHODS: We determined the efficacy of CHOP-ASO in the early and late stages of experimental DKD (in 8- or 16-week-old db/db mice, respectively) alone or with an angiotensin-converting enzyme inhibitor (ACEi), after an in vivo dose-escalation study. We used renal functional parameters and morphologic analyses to assess the effect of CHOP-ASO and renal gene-expression profiling to identify differentially regulated genes and pathways. Several human CHOP-ASOs were tested in hyperglycemia-exposed human kidney cells. RESULTS: CHOP-ASOs efficiently reduced renal CHOP expression in diabetic mice and reduced markers of DKD at the early and late stages. Early combined intervention (CHOP-ASO and ACEi) efficiently prevented interstitial damage. At the later timepoint, the combined treatment reduced indices of both glomerular and tubular damage more efficiently than either intervention alone. CHOP-ASO affected a significantly larger number of genes and disease pathways, including reduced sodium-glucose transport protein 2 (Slc5a2) and PROM1 (CD133). Human CHOP-ASOs efficiently reduced glucose-induced CHOP and prevented death of human kidney cells in vitro . CONCLUSIONS: The ASO-based approach efficiently reduced renal CHOP expression in a diabetic mouse model, providing an additional benefit to an ACEi, particularly at later timepoints. These studies demonstrate that ASO-based therapies efficiently reduce maladaptive CHOP expression and ameliorate experimental DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Mice , Humans , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Diabetic Nephropathies/prevention & control , Diabetes Mellitus, Experimental/complications , Kidney Glomerulus , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Kidney , Oligonucleotides, Antisense/pharmacology
8.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576036

ABSTRACT

Procoagulant extracellular vesicles (EV) and platelet activation have been associated with gestational vascular complications. EV-induced platelet-mediated placental inflammasome activation has been shown to cause preeclampsia-like symptoms in mice. However, the effect of EV-mediated placental thrombo-inflammation on trophoblast differentiation remains unknown. Here, we identify that the EV-induced thrombo-inflammatory pathway modulates trophoblast morphology and differentiation. EVs and platelets reduce syncytiotrophoblast differentiation while increasing giant trophoblast and spongiotrophoblast including the glycogen-rich cells. These effects are platelet-dependent and mediated by the NLRP3 inflammasome. In humans, inflammasome activation was negatively correlated with trophoblast differentiation marker GCM1 and positively correlated with blood pressure. These data identify a crucial role of EV-induced placental thrombo-inflammation on altering trophoblast differentiation and suggest platelet activation or inflammasome activation as a therapeutic target in order to achieve successful placentation.


Subject(s)
Extracellular Vesicles/genetics , Inflammation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pregnancy Complications, Cardiovascular/genetics , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Extracellular Vesicles/metabolism , Female , Humans , Inflammasomes/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Platelet Activation/genetics , Pregnancy , Pregnancy Complications, Cardiovascular/pathology , Transcription Factors/genetics , Trophoblasts/metabolism , Trophoblasts/pathology
9.
Kidney Int ; 98(4): 958-969, 2020 10.
Article in English | MEDLINE | ID: mdl-32505465

ABSTRACT

Biallelic mutations in MAPKBP1 were recently associated with late-onset cilia-independent nephronophthisis. MAPKBP1 was found at mitotic spindle poles but could not be detected at primary cilia or centrosomes. Here, by identification and characterization of novel MAPKBP1 variants, we aimed at further investigating its role in health and disease. Genetic analysis was done by exome sequencing, homozygosity mapping, and a targeted kidney gene panel while coimmunoprecipitation was used to explore wild-type and mutant protein-protein interactions. Expression of MAPKBP1 in non-ciliated HeLa and ciliated inner medullary collecting duct cells enabled co-localization studies by fluorescence microscopy. By next generation sequencing, we identified two novel homozygous MAPKBP1 splice-site variants in patients with nephronophthisis-related chronic kidney disease. Splice-site analyses revealed truncation of C-terminal coiled-coil domains and patient-derived deletion constructs lost their ability to homodimerize and heterodimerize with paralogous WDR62. While wild-type MAPKBP1 exhibited centrosomal, basal body, and microtubule association, mutant proteins lost the latter and showed reduced recruitment to cell cycle dependent centriolar structures. Wild-type and mutant proteins had no reciprocal influence upon co-expression excluding dominant negative effects. Thus, MAPKBP1 appears to be a novel microtubule-binding protein with cell cycle dependent centriolar localization. Truncation of its coiled-coil domain is enough to abrogate its dimerization and results in severely disturbed intracellular localizations. Delineating the impact of impaired dimerization on cell cycle regulation and intracellular kidney signaling may provide new insights into common mechanisms of kidney degeneration. Thus, due to milder clinical presentation, MAPKBP1-associated nephronophthisis should be considered in adult patients with otherwise unexplained chronic kidney disease.


Subject(s)
Centrosome , Polycystic Kidney Diseases , Adult , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Cilia/metabolism , Dimerization , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/metabolism , Polycystic Kidney Diseases/metabolism
10.
Curr Opin Hematol ; 26(1): 41-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30451721

ABSTRACT

PURPOSE OF REVIEW: The serine protease activated protein C (aPC) was initially characterized as an endogenous anticoagulant, but in addition conveys anti-inflammatory, barrier-protective, and pro cell-survival functions. Its endogenous anticoagulant function hampered the successful and continuous implantation of aPC as a therapeutic agent in septic patients. However, it became increasingly apparent that aPC controls cellular function largely independent of its anticoagulant effects through cell-specific and context-specific receptor complexes and intracellular signaling pathways. The purpose of this review is to outline the mechanisms of aPC-dependent cell signaling and its intracellular molecular targets. RECENT FINDINGS: With the advent of new therapeutic agents either modulating directly and specifically the activity of coagulation proteases or interfering with protease-activated receptor signaling a better understanding not only of the receptor mechanisms but also of the intracellular signaling mechanisms controlled by aPC in a disease-specific and context-specific fashion, is required to tailor new therapeutic approaches based on aPC's anti-inflammatory, barrier-protective, and pro cell-survival functions. SUMMARY: This review summarizes recent insights into the intracellular signaling pathways controlled by aPC in a cell-specific and context-specific fashion. We focus on aPC-mediated barrier protection, inhibition of inflammation, and cytoprotecting within this review.


Subject(s)
Anticoagulants , Protein C , Sepsis , Signal Transduction/drug effects , Animals , Anticoagulants/metabolism , Anticoagulants/therapeutic use , Humans , Protein C/metabolism , Protein C/therapeutic use , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/pathology
11.
Blood ; 130(24): 2664-2677, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28882883

ABSTRACT

Cytoprotection by activated protein C (aPC) after ischemia-reperfusion injury (IRI) is associated with apoptosis inhibition. However, IRI is hallmarked by inflammation, and hence, cell-death forms disjunct from immunologically silent apoptosis are, in theory, more likely to be relevant. Because pyroptosis (ie, cell death resulting from inflammasome activation) is typically observed in IRI, we speculated that aPC ameliorates IRI by inhibiting inflammasome activation. Here we analyzed the impact of aPC on inflammasome activity in myocardial and renal IRIs. aPC treatment before or after myocardial IRI reduced infarct size and Nlrp3 inflammasome activation in mice. Kinetic in vivo analyses revealed that Nlrp3 inflammasome activation preceded myocardial injury and apoptosis, corroborating a pathogenic role of the Nlrp3 inflammasome. The constitutively active Nlrp3A350V mutation abolished the protective effect of aPC, demonstrating that Nlrp3 suppression is required for aPC-mediated protection from IRI. In vitro aPC inhibited inflammasome activation in macrophages, cardiomyocytes, and cardiac fibroblasts via proteinase-activated receptor 1 (PAR-1) and mammalian target of rapamycin complex 1 (mTORC1) signaling. Accordingly, inhibiting PAR-1 signaling, but not the anticoagulant properties of aPC, abolished the ability of aPC to restrict Nlrp3 inflammasome activity and tissue damage in myocardial IRI. Targeting biased PAR-1 signaling via parmodulin-2 restricted mTORC1 and Nlrp3 inflammasome activation and limited myocardial IRI as efficiently as aPC. The relevance of aPC-mediated Nlrp3 inflammasome suppression after IRI was corroborated in renal IRI, where the tissue protective effect of aPC was likewise dependent on Nlrp3 inflammasome suppression. These studies reveal that aPC protects from IRI by restricting mTORC1-dependent inflammasome activation and that mimicking biased aPC PAR-1 signaling using parmodulins may be a feasible therapeutic approach to combat IRI.


Subject(s)
Inflammasomes/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein C/pharmacology , Reperfusion Injury/prevention & control , Animals , Animals, Newborn , Anticoagulants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/genetics , Immunoblotting , Inflammasomes/metabolism , Kidney/blood supply , Kidney/drug effects , Kidney/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Protective Agents/pharmacology , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Reperfusion Injury/metabolism
12.
Blood ; 130(12): 1445-1455, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28687614

ABSTRACT

Coagulation proteases have increasingly recognized functions beyond hemostasis and thrombosis. Disruption of activated protein C (aPC) or insulin signaling impair function of podocytes and ultimately cause dysfunction of the glomerular filtration barrier and diabetic kidney disease (DKD). We here show that insulin and aPC converge on a common spliced-X-box binding protein-1 (sXBP1) signaling pathway to maintain endoplasmic reticulum (ER) homeostasis. Analogous to insulin, physiological levels of aPC maintain ER proteostasis in DKD. Accordingly, genetically impaired protein C activation exacerbates maladaptive ER response, whereas genetic or pharmacological restoration of aPC maintains ER proteostasis in DKD models. Importantly, in mice with podocyte-specific deficiency of insulin receptor (INSR), aPC selectively restores the activity of the cytoprotective ER-transcription factor sXBP1 by temporally targeting INSR downstream signaling intermediates, the regulatory subunits of PI3Kinase, p85α and p85ß. Genome-wide mapping of condition-specific XBP1-transcriptional regulatory patterns confirmed that concordant unfolded protein response target genes are involved in maintenance of ER proteostasis by both insulin and aPC. Thus, aPC efficiently employs disengaged insulin signaling components to reconfigure ER signaling and restore proteostasis. These results identify ER reprogramming as a novel hormonelike function of coagulation proteases and demonstrate that targeting insulin signaling intermediates may be a feasible therapeutic approach ameliorating defective insulin signaling.


Subject(s)
Blood Coagulation , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Insulin/metabolism , Peptide Hydrolases/metabolism , Protein C/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , Animals , Diabetic Nephropathies/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Homeostasis , Humans , Mice, Inbred C57BL , Models, Biological , Thrombomodulin/metabolism , Unfolded Protein Response/genetics
13.
Blood ; 128(17): 2153-2164, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27589872

ABSTRACT

Preeclampsia (PE) is a placenta-induced inflammatory disease associated with maternal and fetal morbidity and mortality. The mechanisms underlying PE remain enigmatic and delivery of the placenta is the only known remedy. PE is associated with coagulation and platelet activation and increased extracellular vesicle (EV) formation. However, thrombotic occlusion of the placental vascular bed is rarely observed and the mechanistic relevance of EV and platelet activation remains unknown. Here we show that EVs induce a thromboinflammatory response specifically in the placenta. Following EV injection, activated platelets accumulate particularly within the placental vascular bed. EVs cause adenosine triphosphate (ATP) release from platelets and inflammasome activation within trophoblast cells through purinergic signaling. Inflammasome activation in trophoblast cells triggers a PE-like phenotype, characterized by pregnancy failure, elevated blood pressure, increased plasma soluble fms-like tyrosine kinase 1, and renal dysfunction. Intriguingly, genetic inhibition of inflammasome activation specifically in the placenta, pharmacological inhibition of inflammasome or purinergic signaling, or genetic inhibition of maternal platelet activation abolishes the PE-like phenotype. Inflammasome activation in trophoblast cells of women with preeclampsia corroborates the translational relevance of these findings. These results strongly suggest that EVs cause placental sterile inflammation and PE through activation of maternal platelets and purinergic inflammasome activation in trophoblast cells, uncovering a novel thromboinflammatory mechanism at the maternal-embryonic interface.


Subject(s)
Extracellular Vesicles/pathology , Inflammasomes/immunology , Platelet Activation/physiology , Pre-Eclampsia/physiopathology , Trophoblasts/pathology , Animals , Blood Platelets/immunology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Extracellular Vesicles/immunology , Female , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Pre-Eclampsia/immunology , Pre-Eclampsia/pathology , Pregnancy , Trophoblasts/immunology
14.
J Am Soc Nephrol ; 28(9): 2618-2630, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28424276

ABSTRACT

Nephrotic syndrome is characterized by massive proteinuria and injury of specialized glomerular epithelial cells called podocytes. Studies have shown that, whereas low-concentration thrombin may be cytoprotective, higher thrombin concentrations may contribute to podocyte injury. We and others have demonstrated that ex vivo plasma thrombin generation is enhanced during nephrosis, suggesting that thrombin may contribute to nephrotic progression. Moreover, nonspecific thrombin inhibition has been shown to decrease proteinuria in nephrotic animal models. We thus hypothesized that thrombin contributes to podocyte injury in a protease-activated receptor-specific manner during nephrosis. Here, we show that specific inhibition of thrombin with hirudin reduced proteinuria in two rat nephrosis models, and thrombin colocalized with a podocyte-specific marker in rat glomeruli. Furthermore, flow cytometry immunophenotyping revealed that rat podocytes express the protease-activated receptor family of coagulation receptors in vivo High-concentration thrombin directly injured conditionally immortalized human and rat podocytes. Using receptor-blocking antibodies and activation peptides, we determined that thrombin-mediated injury depended upon interactions between protease-activated receptor 3 and protease-activated receptor 4 in human podocytes, and between protease-activated receptor 1 and protease-activated receptor 4 in rat podocytes. Proximity ligation and coimmunoprecipitation assays confirmed thrombin-dependent interactions between human protease-activated receptor 3 and protease-activated receptor 4, and between rat protease-activated receptor 1 and protease-activated receptor 4 in cultured podocytes. Collectively, these data implicate thrombinuria as a contributor to podocyte injury during nephrosis, and suggest that thrombin and/or podocyte-expressed thrombin receptors may be novel therapeutic targets for nephrotic syndrome.


Subject(s)
Kidney Glomerulus/metabolism , Nephrosis/metabolism , Podocytes/pathology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Animals , Antithrombins/pharmacology , Cell Survival , Cells, Cultured , Disease Models, Animal , Gene Expression , Hirudins/pharmacology , Humans , Immunophenotyping , Nephrosis/complications , Nephrosis/pathology , Nephrosis/urine , Podocytes/metabolism , Proteinuria/etiology , Rats , Receptor, PAR-1/genetics , Receptors, Thrombin/genetics , Signal Transduction , Thrombin/antagonists & inhibitors , Thrombin/pharmacology , Thrombin/urine
15.
J Am Soc Nephrol ; 28(11): 3182-3189, 2017 11.
Article in English | MEDLINE | ID: mdl-28696246

ABSTRACT

Established therapies for diabetic nephropathy (dNP) delay but do not prevent its progression. The shortage of established therapies may reflect the inability to target the tubular compartment. The chemical chaperone tauroursodeoxycholic acid (TUDCA) ameliorates maladaptive endoplasmic reticulum (ER) stress signaling and experimental dNP. Additionally, TUDCA activates the farnesoid X receptor (FXR), which is highly expressed in tubular cells. We hypothesized that TUDCA ameliorates maladaptive ER signaling via FXR agonism specifically in tubular cells. Indeed, TUDCA induced expression of FXR-dependent genes (SOCS3 and DDAH1) in tubular cells but not in other renal cells. In vivo, TUDCA reduced glomerular and tubular injury in db/db and diabetic endothelial nitric oxide synthase-deficient mice. FXR inhibition with Z-guggulsterone or vivo-morpholino targeting of FXR diminished the ER-stabilizing and renoprotective effects of TUDCA. Notably, these in vivo approaches abolished tubular but not glomerular protection by TUDCA. Combined intervention with TUDCA and the angiotensin-converting enzyme inhibitor enalapril in 16-week-old db/db mice reduced albuminuria more efficiently than did either treatment alone. Although both therapies reduced glomerular damage, only TUDCA ameliorated tubular damage. Thus, interventions that specifically protect the tubular compartment in dNP, such as FXR agonism, may provide renoprotective effects on top of those achieved by inhibiting angiotensin-converting enzyme.


Subject(s)
Diabetic Nephropathies/prevention & control , Kidney Tubules , Receptors, Cytoplasmic and Nuclear/agonists , Taurochenodeoxycholic Acid/therapeutic use , Animals , Humans , Male , Mice , Mice, Inbred C57BL
16.
J Am Soc Nephrol ; 27(8): 2270-5, 2016 08.
Article in English | MEDLINE | ID: mdl-26832955

ABSTRACT

Glomerular apoptosis may contribute to diabetic nephropathy (dNP), but the pathophysiologic relevance of this process remains obscure. Here, we administered two partially disjunct polycaspase inhibitors in 8-week-old diabetic (db/db) mice: M-920 (inhibiting caspase-1, -3, -4, -5, -6, -7, and -8) and CIX (inhibiting caspase-3, -6, -7, -8, and -10). Notably, despite reduction in glomerular cell death and caspase-3 activity by both inhibitors, only M-920 ameliorated dNP. Nephroprotection by M-920 was associated with reduced renal caspase-1 and inflammasome activity. Accordingly, analysis of gene expression data in the Nephromine database revealed persistently elevated glomerular expression of inflammasome markers (NLRP3, CASP1, PYCARD, IL-18, IL-1ß), but not of apoptosis markers (CASP3, CASP7, PARP1), in patients with and murine models of dNP. In vitro, increased levels of markers of inflammasome activation (Nlrp3, caspase-1 cleavage) preceded those of markers of apoptosis activation (caspase-3 and -7, PARP1 cleavage) in glucose-stressed podocytes. Finally, caspase-3 deficiency did not protect mice from dNP, whereas both homozygous and hemizygous caspase-1 deficiency did. Hence, these results suggest caspase-3-dependent cell death has a negligible effect, whereas caspase-1-dependent inflammasome activation has a crucial function in the establishment of dNP. Furthermore, small molecules targeting caspase-1 or inflammasome activation may be a feasible therapeutic approach in dNP.


Subject(s)
Caspase 1/physiology , Caspase 3/physiology , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/etiology , Animals , Inflammasomes , Mice
17.
Proc Natl Acad Sci U S A ; 110(2): 648-53, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23267072

ABSTRACT

The coagulation protease activated protein C (aPC) confers cytoprotective effects in various in vitro and in vivo disease models, including diabetic nephropathy. The nephroprotective effect may be related to antioxidant effects of aPC. However, the mechanism through which aPC may convey these antioxidant effects and the functional relevance of these properties remain unknown. Here, we show that endogenous and exogenous aPC prevents glomerular accumulation of oxidative stress markers and of the redox-regulating protein p66(Shc) in experimental diabetic nephropathy. These effects were predominately observed in podocytes. In vitro, aPC inhibited glucose-induced expression of p66(Shc) mRNA and protein in podocytes (via PAR-1 and PAR-3) and various endothelial cell lines, but not in glomerular endothelial cells. Treatment with aPC reversed glucose-induced hypomethylation and hyperacetylation of the p66(Shc) promoter in podocytes. The hyperacetylating agent sodium butyrate abolished the suppressive effect of aPC on p66(Shc) expression both in vitro and in vivo. Moreover, sodium butyrate abolished the beneficial effects of aPC in experimental diabetic nephropathy. Inhibition of p66(Shc) expression and mitochondrial translocation by aPC normalized mitochondrial ROS production and the mitochondrial membrane potential in glucose-treated podocytes. Genetic ablation of p66(Shc) compensated for the loss of protein C activation in vivo, normalizing markers of diabetic nephropathy and oxidative stress. These studies identify a unique mechanism underlying the cytoprotective effect of aPC. Activated PC epigenetically controls expression of the redox-regulating protein p66(Shc), thus linking the extracellular protease aPC to mitochondrial function in diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/drug therapy , Epigenetic Repression/drug effects , Mitochondria/drug effects , Protein C/pharmacology , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Analysis of Variance , Animals , Butyrates/pharmacology , Chromatin Immunoprecipitation , DNA Methylation/drug effects , DNA Primers/genetics , Diabetic Nephropathies/etiology , Gene Knockdown Techniques , Immunoblotting , Immunohistochemistry , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Podocytes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Statistics, Nonparametric , Subcellular Fractions
18.
J Am Soc Nephrol ; 26(11): 2789-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26015455

ABSTRACT

Ischemia-reperfusion injury (IRI) is the leading cause of ARF. A pathophysiologic role of the coagulation system in renal IRI has been established, but the functional relevance of thrombomodulin (TM)-dependent activated protein C (aPC) generation and the intracellular targets of aPC remain undefined. Here, we investigated the role of TM-dependent aPC generation and therapeutic aPC application in a murine renal IRI model and in an in vitro hypoxia and reoxygenation (HR) model using proximal tubular cells. In renal IRI, endogenous aPC levels were reduced. Genetic or therapeutic reconstitution of aPC efficiently ameliorated renal IRI independently of its anticoagulant properties. In tubular cells, cytoprotective aPC signaling was mediated through protease activated receptor-1- and endothelial protein C receptor-dependent regulation of the cold-shock protein Y-box binding protein-1 (YB-1). The mature 50 kD form of YB-1 was required for the nephro- and cytoprotective effects of aPC in vivo and in vitro, respectively. Reduction of mature YB-1 and K48-linked ubiquitination of YB-1 was prevented by aPC after renal IRI or tubular HR injury. aPC preserved the interaction of YB-1 with the deubiquitinating enzyme otubain-1 and maintained expression of otubain-1, which was required to reduce K48-linked YB-1 ubiquitination and to stabilize the 50 kD form of YB-1 after renal IRI and tubular HR injury. These data link the cyto- and nephroprotective effects of aPC with the ubiquitin-proteasome system and identify YB-1 as a novel intracellular target of aPC. These insights may provide new impetus for translational efforts aiming to restrict renal IRI.


Subject(s)
Kidney/pathology , Protein C/metabolism , Reperfusion Injury/pathology , Transcription Factors/metabolism , Ubiquitination , Alleles , Animals , Anticoagulants/chemistry , Crosses, Genetic , Cysteine Endopeptidases/genetics , Disease Models, Animal , Exons , Hypoxia/pathology , Kidney Tubules/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/chemistry , Signal Transduction , Thrombosis/metabolism
19.
Kidney Int ; 87(1): 74-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25075770

ABSTRACT

Diabetic nephropathy is a growing health concern with characteristic sterile inflammation. As the underlying mechanisms of this inflammation remain poorly defined, specific therapies targeting sterile inflammation in diabetic nephropathy are lacking. Intriguingly, an association of diabetic nephropathy with inflammasome activation has recently been shown, but the pathophysiological relevance of this finding remains unknown. Within glomeruli, inflammasome activation was detected in endothelial cells and podocytes in diabetic humans and mice and in glucose-stressed glomerular endothelial cells and podocytes in vitro. Abolishing Nlrp3 or caspase-1 expression in bone marrow-derived cells fails to protect mice against diabetic nephropathy. Conversely, Nlrp3-deficient mice are protected against diabetic nephropathy despite transplantation of wild-type bone marrow. Pharmacological IL-1R antagonism prevented or even reversed diabetic nephropathy in mice. Mitochondrial reactive oxygen species (ROS) activate the Nlrp3 inflammasome in glucose or advanced glycation end product stressed podocytes. Inhibition of mitochondrial ROS prevents glomerular inflammasome activation and nephropathy in diabetic mice. Thus, mitochondrial ROS and Nlrp3-inflammasome activation in non-myeloid-derived cells aggravate diabetic nephropathy. Targeting the inflammasome may be a potential therapeutic approach to diabetic nephropathy.


Subject(s)
Carrier Proteins/immunology , Diabetic Nephropathies/immunology , Inflammasomes/immunology , Kidney Glomerulus/cytology , Animals , Endothelial Cells/immunology , Humans , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Podocytes/immunology , Severity of Illness Index
20.
Development ; 138(11): 2235-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558372

ABSTRACT

Absence of the leucine zipper transcription factor p45NF-E2 results in thrombocytopenia, impaired placental vascularization and intrauterine growth restriction (IUGR) in mice. The mechanism underlying the p45NF-E2-dependent placental defect and IUGR remains unknown. Here, we show that the placental defect and IUGR of p45NF-E2 (Nfe2) null mouse embryos is unrelated to thrombocytopenia, establishing that embryonic platelets and platelet-released mediators are dispensable for placentation. Rather, p45NF-E2, which was hitherto thought to be specific to hematopoietic cells, is expressed in trophoblast cells, where it is required for normal syncytiotrophoblast formation, placental vascularization and embryonic growth. Expression of p45NF-E2 in labyrinthine trophoblast cells colocalizes with that of Gcm1, a transcription factor crucial for syncytiotrophoblast formation. In the absence of p45NF-E2, the width of syncytiotrophoblast layer 2 and the expression of Gcm1 and Gcm1-dependent genes (Synb and Cebpa) are increased. In vitro, p45NF-E2 deficiency results in spontaneous syncytiotrophoblast formation, which can be reversed by Gcm1 knockdown. Increased Gcm1 expression in the absence of p45NF-E2 is dependent on enhanced protein acetylation, including post-translational modification of Gcm1. Increasing and inhibiting acetylation in the placenta of wild-type control embryos phenocopies and corrects, respectively, the changes observed in p45NF-E2-deficient embryos. These studies identify a novel function of p45NF-E2 during placental development: in trophoblast cells, p45NF-E2 represses Gcm1 and syncytiotrophoblast formation via acetylation.


Subject(s)
Embryonic Development , NF-E2 Transcription Factor, p45 Subunit/metabolism , Neovascularization, Physiologic , Neuropeptides/metabolism , Placenta/blood supply , Trophoblasts/metabolism , Acetylation , Animals , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Fetal Growth Retardation , Gene Knock-In Techniques , Giant Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , NF-E2 Transcription Factor, p45 Subunit/genetics , Neuropeptides/genetics , Placenta/metabolism , Placentation , Polymerase Chain Reaction , Pregnancy , Protein Processing, Post-Translational , Thrombocytopenia , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL