Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 14(660): eabj7465, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36044595

ABSTRACT

Arterial and venous thrombosis constitutes a major source of morbidity and mortality worldwide. Long considered as distinct entities, accumulating evidence indicates that arterial and venous thrombosis can occur in the same populations, suggesting that common mechanisms are likely operative. Although hyperactivation of the immune system is a common forerunner to the genesis of thrombotic events in both vascular systems, the key molecular control points remain poorly understood. Consequently, antithrombotic therapies targeting the immune system for therapeutics gain are lacking. Here, we show that neutrophils are key effectors of both arterial and venous thrombosis and can be targeted through immunoregulatory nanoparticles. Using antiphospholipid antibody syndrome (APS) as a model for arterial and venous thrombosis, we identified the transcription factor Krüppel-like factor 2 (KLF2) as a key regulator of neutrophil activation. Upon activation through genetic loss of KLF2 or administration of antiphospholipid antibodies, neutrophils clustered P-selectin glycoprotein ligand 1 (PSGL-1) by cortical actin remodeling, thereby increasing adhesion potential at sites of thrombosis. Targeting clustered PSGL-1 using nanoparticles attenuated neutrophil-mediated thrombosis in APS and KLF2 knockout models, illustrating the importance and feasibility of targeting activated neutrophils to prevent pathological thrombosis. Together, our results demonstrate a role for activated neutrophils in both arterial and venous thrombosis and identify key molecular events that serve as potential targets for therapeutics against diverse causes of immunothrombosis.


Subject(s)
Antiphospholipid Syndrome , Thrombosis , Venous Thrombosis , Antibodies, Antiphospholipid , Humans , Neutrophils/metabolism , Thrombosis/etiology
3.
Nat Commun ; 11(1): 5872, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208733

ABSTRACT

Substantial evidence implicates crosstalk between metabolic tissues and the immune system in the inception and progression of obesity. However, molecular regulators that orchestrate metaflammation both centrally and peripherally remains incompletely understood. Here, we identify myeloid Krüppel-like factor 2 (KLF2) as an essential regulator of obesity and its sequelae. In mice and humans, consumption of a fatty diet downregulates myeloid KLF2 levels. Under basal conditions, myeloid-specific KLF2 knockout mice (K2KO) exhibit increased feeding and weight gain. High-fat diet (HFD) feeding further exacerbates the K2KO metabolic disease phenotype. Mechanistically, loss of myeloid KLF2 increases metaflammation in peripheral and central tissues. A combination of pair-feeding, bone marrow-transplant, and microglial ablation implicate central and peripheral contributions to K2KO-induced metabolic dysfunction observed. Finally, overexpression of myeloid KLF2 protects mice from HFD-induced obesity and insulin resistance. Together, these data establish myeloid KLF2 as a nodal regulator of central and peripheral metabolic inflammation in homeostasis and disease.


Subject(s)
Kruppel-Like Transcription Factors/immunology , Metabolic Diseases/immunology , Myeloid Cells/immunology , Obesity/immunology , Animals , Central Nervous System/immunology , Diet, High-Fat/adverse effects , Eating , Humans , Inflammation , Insulin Resistance , Kruppel-Like Transcription Factors/genetics , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Mice , Mice, Knockout , Obesity/etiology , Obesity/genetics , Obesity/physiopathology , Peripheral Nervous System/immunology
4.
Curr Top Med Chem ; 19(16): 1436-1444, 2019.
Article in English | MEDLINE | ID: mdl-31512997

ABSTRACT

Type 2 diabetes is a major health issue worldwide with complex metabolic and endocrine abnormalities. Hyperglycemia, defects in insulin secretion and insulin resistance are classic features of type 2 diabetes. Insulin signaling regulates metabolic homeostasis by regulating glucose and lipid turnover in the liver, skeletal muscle and adipose tissue. Major treatment modalities for diabetes include the drugs from the class of sulfonyl urea, Insulin, GLP-1 agonists, SGLT2 inhibitors, DPP-IV inhibitors and Thiazolidinediones. Emerging antidiabetic therapeutics also include classes of drugs targeting GPCRs in the liver, adipose tissue and skeletal muscle. Interestingly, recent research highlights several shared intermediates between insulin and GPCR signaling cascades opening potential novel avenues for diabetic drug discovery.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Discovery , Hypoglycemic Agents/pharmacology , Receptor, Insulin/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Hypoglycemic Agents/chemistry , Receptor, Insulin/metabolism , Receptors, G-Protein-Coupled/metabolism
5.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29712858

ABSTRACT

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Subject(s)
Cardiomegaly/pathology , Cardiomyopathies/pathology , Heart Failure/pathology , Kruppel-Like Transcription Factors/metabolism , Macrophages/pathology , Myocardium/pathology , Animals , Cardiomegaly/immunology , Cardiomegaly/metabolism , Cardiomyopathies/immunology , Cardiomyopathies/metabolism , Cells, Cultured , Heart Failure/immunology , Heart Failure/metabolism , Kruppel-Like Factor 4 , Macrophages/immunology , Macrophages/metabolism , Mice , Myocardium/immunology , Myocardium/metabolism , Pressure
6.
Methods Cell Biol ; 142: 51-57, 2017.
Article in English | MEDLINE | ID: mdl-28964339

ABSTRACT

GPCRs are a major family of cell surface receptors and the most druggable protein targets in modern medicine. Recent elucidation of crystal structures of a vast number of GPCRs eludes to the finer details of biased agonism or functional selectivity of many of these receptors and warrants a better understanding of their biological effects as well as therapeutic potential. Receptor function is measured in terms desensitization/resensitization, which provides insights on receptor activation and differential coupling to various G proteins. This review article presents thoughts on the potential of GPCR desensitization assay/cAMP assay, a highly sensitive and versatile platform for high-throughput assays for elucidating novel functions of many orphan GPCRs. Besides, these assays also are very sensitive to screen for agonists, partial agonists, or antagonists and provide a simplified system for novel drug discovery for Gs-coupled GPCRs and understanding their physiological implications.


Subject(s)
Biological Assay/methods , Cyclic AMP/analysis , Drug Discovery/methods , Receptors, G-Protein-Coupled/metabolism , Fluorescence , HEK293 Cells , Humans , Ligands , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors
7.
J Cardiovasc Pharmacol ; 70(2): 61-73, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28763371

ABSTRACT

Proinflammatory reaction by the body occurs acutely in response to injury that is considered primarily beneficial. However, sustained proinflammatory cytokines observed with chronic pathologies such as metabolic syndrome, cancer, and arthritis are detrimental and in many cases is a major cardiovascular risk factor. Proinflammatory cytokines such as interleukin-1, interleukin-6, and tumor necrosis factor α (TNFα) have long been implicated in cardiovascular risk and considered to be a major underlying cause for heart failure (HF). The failure of the anti-TNFα therapy for HF indicates our elusive understanding on the dichotomous role of proinflammatory cytokines on acutely beneficial effects versus long-term deleterious effects. Despite these well-described observations, less is known about the mechanistic underpinnings of proinflammatory cytokines especially TNFα in pathogenesis of HF. Increasing evidence suggests the existence of an active cross-talk between the TNFα receptor signaling and G-protein-coupled receptors such as ß-adrenergic receptor (ßAR). Given that ßARs are the key regulators of cardiac function, the review will discuss the current state of understanding on the role of proinflammatory cytokine TNFα in regulating ßAR function.


Subject(s)
Cytokines/physiology , Inflammation Mediators/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Humans , Inflammation/metabolism , Inflammation/physiopathology , Signal Transduction/physiology
8.
mBio ; 7(3)2016 06 14.
Article in English | MEDLINE | ID: mdl-27302755

ABSTRACT

UNLABELLED: The adaptive immune response is tightly regulated by complex signals in dendritic cells (DCs). Although Th2 polarization is dictated by defined functional DC subsets, the molecular factors that govern the amplitude of these responses are not well understood. Krüppel-like factor 2 (KLF2) is a transcription factor that negatively regulates the activation of numerous immune cells in response to stimuli. Here, we demonstrate that suppression of KLF2 in conditioned DCs preferentially amplifies Th2 responses in two model systems, one of which is a prototypical intracellular pathogen and the other an allergen. This elevation in Th2 responses was dependent on contact-mediated Notch signaling in vitro and in vivo A deficiency of KLF2 increased the expression of Notch ligand Jagged2 via hypoxia-inducible factor 1α (HIF-1α), which led to Th2 amplification. Our results revealed a novel circuit in DCs for Th2 polarization that is governed by KLF2. IMPORTANCE: Dendritic cells are the key element that bridges innate and adaptive immunity. A complex and not-well-understood area in dendritic cell biology is the regulatory network that predetermines or moderates their function to shape the adaptive immune response. Our study for the first time demonstrates that KLF2, a transcription factor, conditions dendritic cells to regulate Th2 responses via a Jagged2/Notch axis. Downregulation of KLF2 expression in dendritic cells may provide a beneficial effect for treatment of diseases such as obesity or parasitic infections but may be deleterious in the case of invasion by intracellular pathogens. Strategies to tune KLF2 may be useful for future therapeutic approaches to particular diseases of mankind.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Jagged-2 Protein/metabolism , Kruppel-Like Transcription Factors/metabolism , Receptors, Notch/metabolism , Th2 Cells/immunology , Animals , Mice, Inbred C57BL , Transcription, Genetic
9.
Circulation ; 128(4): 377-87, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23785004

ABSTRACT

BACKGROUND: Proinflammatory cytokine tumor necrosis factor-α (TNFα) induces ß-adrenergic receptor (ßAR) desensitization, but mechanisms proximal to the receptor in contributing to cardiac dysfunction are not known. METHODS AND RESULTS: Two different proinflammatory transgenic mouse models with cardiac overexpression of myotrophin (a prohypertrophic molecule) or TNFα showed that TNFα alone is sufficient to mediate ßAR desensitization as measured by cardiac adenylyl cyclase activity. M-mode echocardiography in these mouse models showed cardiac dysfunction paralleling ßAR desensitization independent of sympathetic overdrive. TNFα-mediated ßAR desensitization that precedes cardiac dysfunction is associated with selective upregulation of G-protein coupled receptor kinase 2 (GRK2) in both mouse models. In vitro studies in ß2AR-overexpressing human embryonic kidney 293 cells showed significant ßAR desensitization, GRK2 upregulation, and recruitment to the ßAR complex following TNFα. Interestingly, inhibition of phosphoinositide 3-kinase abolished GRK2-mediated ßAR phosphorylation and GRK2 recruitment on TNFα. Furthermore, TNFα-mediated ßAR phosphorylation was not blocked with ßAR antagonist propranolol. Additionally, TNFα administration in transgenic mice with cardiac overexpression of Gßγ-sequestering peptide ßARK-ct could not prevent ßAR desensitization or cardiac dysfunction showing that GRK2 recruitment to the ßAR is Gßγ independent. Small interfering RNA knockdown of GRK2 resulted in the loss of TNFα-mediated ßAR phosphorylation. Consistently, cardiomyocytes from mice with cardiac-specific GRK2 ablation normalized the TNFα-mediated loss in contractility, showing that TNFα-induced ßAR desensitization is GRK2 dependent. CONCLUSIONS: TNFα-induced ßAR desensitization is mediated by GRK2 and is independent of Gßγ, uncovering a hitherto unknown cross-talk between TNFα and ßAR function, providing the underpinnings of inflammation-mediated cardiac dysfunction.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Heart Failure/metabolism , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Disease Models, Animal , HEK293 Cells , Heart Failure/pathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/physiology , Propranolol/pharmacology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Sympathetic Nervous System/physiology , Tumor Necrosis Factor-alpha/genetics
10.
J Mol Cell Cardiol ; 62: 131-41, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23735785

ABSTRACT

High fidelity genome-wide expression analysis has strengthened the idea that microRNA (miRNA) signatures in peripheral blood mononuclear cells (PBMCs) can be potentially used to predict the pathology when anatomical samples are inaccessible like the heart. PBMCs from 48 non-failing controls and 44 patients with relatively stable chronic heart failure (ejection fraction of ≤ 40%) associated with dilated cardiomyopathy (DCM) were used for miRNA analysis. Genome-wide miRNA-microarray on PBMCs from chronic heart failure patients identified miRNA signature uniquely characterized by the downregulation of miRNA-548 family members. We have also independently validated downregulation of miRNA-548 family members (miRNA-548c & 548i) using real time-PCR in a large cohort of independent patient samples. Independent in silico Ingenuity Pathway Analysis (IPA) of miRNA-548 targets shows unique enrichment of signaling molecules and pathways associated with cardiovascular disease and hypertrophy. Consistent with specificity of miRNA changes with pathology, PBMCs from breast cancer patients showed no alterations in miRNA-548c expression compared to healthy controls. These studies suggest that miRNA-548 family signature in PBMCs can therefore be used to detect early heart failure. Our studies show that cognate networking of predicted miRNA-548 targets in heart failure can be used as a powerful ancillary tool to predict the ongoing pathology.


Subject(s)
Cardiomyopathy, Dilated/genetics , Leukocytes, Mononuclear/metabolism , MicroRNAs/genetics , Breast Neoplasms/genetics , Cells, Cultured , Female , Gene Expression Profiling , Heart Failure/genetics , Humans , Male , Middle Aged
11.
Sci Signal ; 6(259): ra4, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23354687

ABSTRACT

Activation of cardiac phosphoinositide 3-kinase α (PI3Kα) by growth factors, such as insulin, or activation of PI3Kγ downstream of heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors stimulates the activity of the kinase Akt, which phosphorylates and inhibits glycogen synthase kinase-3 (GSK-3). We found that PI3Kγ inhibited GSK-3 independently of the insulin-PI3Kα-Akt axis. Although insulin treatment activated Akt in PI3Kγ knockout mice, phosphorylation of GSK-3 was decreased compared to control mice. GSK-3 is activated when dephosphorylated by the protein phosphatase 2A (PP2A), which is activated when methylated by the PP2A methyltransferase PPMT-1. PI3Kγ knockout mice showed increased activity of PPMT-1 and PP2A and enhanced nuclear export of the GSK-3 substrate NFATc3. GSK-3 inhibits cardiac hypertrophy, and the hearts of PI3Kγ knockout mice were smaller compared to those of wild-type mice. Cardiac overexpression of a catalytically inactive PI3Kγ (PI3Kγ(inact)) transgene in PI3Kγ knockout mice reduced the activities of PPMT-1 and PP2A and increased phosphorylation of GSK-3. Furthermore, PI3Kγ knockout mice expressing the PI3Kγ(inact) transgene had larger hearts than wild-type or PI3Kγ knockout mice. Our studies show that a kinase-independent function of PI3Kγ could directly inhibit GSK-3 function by preventing the PP2A-PPMT-1 interaction and that this inhibition of GSK-3 was independent of Akt.


Subject(s)
Cardiomegaly/enzymology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Glycogen Synthase Kinase 3/metabolism , Muscle Proteins/metabolism , Myocardium/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cardiomegaly/genetics , Cardiomegaly/pathology , Class Ib Phosphatidylinositol 3-Kinase/genetics , Enzyme Activation/genetics , Glycogen Synthase Kinase 3/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardium/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-akt/genetics
12.
Curr Mol Pharmacol ; 2012 May 30.
Article in English | MEDLINE | ID: mdl-22697395

ABSTRACT

G-protein coupled receptors (GPCRs) are seven transmembrane receptors that are pivotal regulators of cellular responses including vision, cardiac contractility, olfaction, and platelet activation. GPCRs have been a major target for drug discovery due to their role in regulating a broad range of physiological and pathological responses. GPCRs mediate these responses through a cyclical process of receptor activation (initiation of downstream signals), desensitization (inactivation that results in diminution of downstream signals), and resensitization (receptor reactivation for next wave of activation). Although these steps may be of equal importance in regulating receptor function, significant advances have been made in understanding activation and desensitization with limited effort towards resensitization. Inadequate importance has been given to resensitization due to the understanding that resensitization is a homeostasis maintaining process and is not acutely regulated. Evidence indicates that resensitization is a critical step in regulating GPCR function and may contribute towards receptor signaling and cellular responses. In light of these observations, it is imperative to discuss resensitization as a dynamic and mechanistic regulator of GPCR function. In this review we discuss components regulating GPCR function like activation, desensitization, and internalization with special emphasis on resensitization. Although we have used ß-adrenergic receptor as a proto-type GPCR to discuss mechanisms regulating receptor function, other GPCRs are also described to put forth a view point on the universality of such mechanisms.

13.
Cell Cycle ; 10(21): 3684-91, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22041711

ABSTRACT

G protein-coupled receptors are the largest family of cell surface receptors regulating multiple cellular processes. ß-adrenergic receptor (ßAR) is a prototypical member of GPCR family and has been one of the most well studied receptors in determining regulation of receptor function. Agonist activation of ßAR leads to conformational change resulting in coupling to G protein generating cAMP as secondary messenger. The activated ßAR is phosphorylated resulting in binding of ß-arrestin that physically interdicts further G protein coupling leading to receptor desensitization. The phosphorylated ßAR is internalized and undergoes resensitization by dephosphorylation mediated by protein phosphatase 2A in the early endosomes. Although desensitization and resensitization are two sides of the same coin maintaining the homeostatic functioning of the receptor, significant interest has revolved around understanding mechanisms of receptor desensitization while little is known about resensitization. In our current review we provide an overview on regulation of ßAR function with a special emphasis on receptor resensitization and its functional relevance in the context of fine tuning receptor signaling.


Subject(s)
Receptors, Adrenergic, beta/physiology , Arrestins/metabolism , Cyclic AMP/metabolism , Endosomes/metabolism , Endosomes/physiology , GTP-Binding Proteins/metabolism , Models, Biological , Phosphorylation , Protein Interaction Domains and Motifs , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/physiology , Receptors, Adrenergic, beta/chemistry , Signal Transduction , beta-Arrestins
14.
Mol Cell ; 41(6): 636-48, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21419339

ABSTRACT

Phosphoinositide 3-kinase γ (PI3Kγ) is activated by G protein-coupled receptors (GPCRs). We show here that PI3Kγ inhibits protein phosphatase 2A (PP2A) at the ß-adrenergic receptor (ßAR, a GPCR) complex altering G protein coupling. PI3Kγ inhibition results in significant increase of ßAR-associated phosphatase activity leading to receptor dephosphorylation and resensitization preserving cardiac function. Mechanistically, PI3Kγ inhibits PP2A activity at the ßAR complex by phosphorylating an intracellular inhibitor of PP2A (I2PP2A) on serine residues 9 and 93, resulting in enhanced binding to PP2A. Indeed, enhanced phosphorylation of ß2ARs is observed with a phosphomimetic I2PP2A mutant that was completely reversed with a mutant mimicking dephosphorylated state. siRNA depletion of endogenous I2PP2A augments PP2A activity despite active PI3K resulting in ß2AR dephosphorylation and sustained signaling. Our study provides the underpinnings of a PI3Kγ-mediated regulation of PP2A activity that has significant consequences on receptor function with broad implications in cellular signaling.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Receptors, Adrenergic, beta-2/physiology , Signal Transduction/physiology , Animals , Cell Membrane/metabolism , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , DNA-Binding Proteins , Endosomes/metabolism , Histone Chaperones/genetics , Histone Chaperones/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL