Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
EMBO J ; 42(16): e113258, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37409632

ABSTRACT

Mitochondrial biogenesis is the process of generating new mitochondria to maintain cellular homeostasis. Here, we report that viruses exploit mitochondrial biogenesis to antagonize innate antiviral immunity. We found that nuclear respiratory factor-1 (NRF1), a vital transcriptional factor involved in nuclear-mitochondrial interactions, is essential for RNA (VSV) or DNA (HSV-1) virus-induced mitochondrial biogenesis. NRF1 deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in mice. Mechanistically, the inhibition of NRF1-mediated mitochondrial biogenesis aggravated virus-induced mitochondrial damage, promoted the release of mitochondrial DNA (mtDNA), increased the production of mitochondrial reactive oxygen species (mtROS), and activated the innate immune response. Notably, virus-activated kinase TBK1 phosphorylated NRF1 at Ser318 and thereby triggered the inactivation of the NRF1-TFAM axis during HSV-1 infection. A knock-in (KI) strategy that mimicked TBK1-NRF1 signaling revealed that interrupting the TBK1-NRF1 connection ablated mtDNA release and thereby attenuated the HSV-1-induced innate antiviral response. Our study reveals a previously unidentified antiviral mechanism that utilizes a NRF1-mediated negative feedback loop to modulate mitochondrial biogenesis and antagonize innate immune response.


Subject(s)
Antiviral Agents , Organelle Biogenesis , Animals , Mice , DNA, Mitochondrial/genetics , Immunity, Innate , Nuclear Respiratory Factor 1/genetics
2.
Nat Chem Biol ; 20(7): 835-846, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38287154

ABSTRACT

Synchronized ferroptosis contributes to nephron loss in acute kidney injury (AKI). However, the propagation signals and the underlying mechanisms of the synchronized ferroptosis for renal tubular injury remain unresolved. Here we report that platelet-activating factor (PAF) and PAF-like phospholipids (PAF-LPLs) mediated synchronized ferroptosis and contributed to AKI. The emergence of PAF and PAF-LPLs in ferroptosis caused the instability of biomembranes and signaled the cell death of neighboring cells. This cascade could be suppressed by PAF-acetylhydrolase (II) (PAFAH2) or by addition of antibodies against PAF. Genetic knockout or pharmacological inhibition of PAFAH2 increased PAF production, augmented synchronized ferroptosis and exacerbated ischemia/reperfusion (I/R)-induced AKI. Notably, intravenous administration of wild-type PAFAH2 protein, but not its enzymatically inactive mutants, prevented synchronized tubular cell death, nephron loss and AKI. Our findings offer an insight into the mechanisms of synchronized ferroptosis and suggest a possibility for the preventive intervention of AKI.


Subject(s)
Acute Kidney Injury , Ferroptosis , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/drug therapy , Ferroptosis/drug effects , Animals , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Platelet Activating Factor/metabolism , Mice, Knockout , Humans , Male
3.
Cytokine ; 176: 156514, 2024 04.
Article in English | MEDLINE | ID: mdl-38277928

ABSTRACT

BACKGROUND: Inflammation is linked to coronavirus disease 2019 (COVID-19)-related heart failure (HF), but the specific mechanisms are unclear. This study aimed to assess the relationship between specific inflammatory factors, such as interleukin (IL)-1ß, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, interferon (IFN)-α, and IFN-γ, and COVID-19-related HF. METHODS: We retrospectively identified 212 adult patients with COVID-19 who were hospitalized at Shanghai Public Health Center from March 1 to May 30, 2022 (including 80 patients with HF and 132 without HF). High-sensitivity C-reactive protein (hs-CRP), procalcitonin (PCT), and inflammatory factors, including IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-17, IFN-α, and IFN-γ, were compared between patients with COVID-19 with and without HF. RESULTS: Patients with COVID-19 having and not having HF differed with regard to sex, age, hs-CRP, PCT, and IL-6 levels (p < 0.05). Logistic regression analysis indicated a significant positive association between IL and 6 and HF (odds ratio = 1.055; 95 % confidence interval: 1.019-1.093, p < 0.005). Sex, age, and hs-CRP were also associated with HF. Women had a greater risk of HF than men. Older age, higher levels of hs-CRP, and IL-6 were associated with a greater risk of HF. CONCLUSIONS: In patients with COVID-19, increased IL-6 levels are significantly associated with COVID-19-related HF.


Subject(s)
COVID-19 , Heart Failure , Adult , Female , Humans , Male , C-Reactive Protein/metabolism , China , COVID-19/complications , Interleukin-10 , Interleukin-12 , Interleukin-17 , Interleukin-2 , Interleukin-4 , Interleukin-5 , Interleukin-6 , Interleukin-8 , Retrospective Studies
4.
EMBO Rep ; 22(3): e50629, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33554448

ABSTRACT

Mitophagy is an essential cellular autophagic process that selectively removes superfluous and damaged mitochondria, and it is coordinated with mitochondrial biogenesis to fine tune the quantity and quality of mitochondria. Coordination between these two opposing processes to maintain the functional mitochondrial network is of paramount importance for normal cellular and organismal metabolism. However, the underlying mechanism is not completely understood. Here we report that PGC-1α and nuclear respiratory factor 1 (NRF1), master regulators of mitochondrial biogenesis and metabolic adaptation, also transcriptionally upregulate the gene encoding FUNDC1, a previously characterized mitophagy receptor, in response to cold stress in brown fat tissue. NRF1 binds to the classic consensus site in the promoter of Fundc1 to upregulate its expression and to enhance mitophagy through its interaction with LC3. Specific knockout of Fundc1 in BAT results in reduced mitochondrial turnover and accumulation of functionally compromised mitochondria, leading to impaired adaptive thermogenesis. Our results demonstrate that FUNDC1-dependent mitophagy is directly coupled with mitochondrial biogenesis through the PGC-1α/NRF1 pathway, which dictates mitochondrial quantity, quality, and turnover and contributes to adaptive thermogenesis.


Subject(s)
Mitophagy , Nuclear Respiratory Factor 1 , Adipose Tissue, Brown/metabolism , Homeostasis , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
5.
Proc Natl Acad Sci U S A ; 117(44): 27667-27675, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33087571

ABSTRACT

Chronic neurodegeneration in survivors of traumatic brain injury (TBI) is a major cause of morbidity, with no effective therapies to mitigate this progressive and debilitating form of nerve cell death. Here, we report that pharmacologic restoration of the blood-brain barrier (BBB), 12 mo after murine TBI, is associated with arrested axonal neurodegeneration and cognitive recovery, benefits that persisted for months after treatment cessation. Recovery was achieved by 30 d of once-daily administration of P7C3-A20, a compound that stabilizes cellular energy levels. Four months after P7C3-A20, electron microscopy revealed full repair of TBI-induced breaks in cortical and hippocampal BBB endothelium. Immunohistochemical staining identified additional benefits of P7C3-A20, including restoration of normal BBB endothelium length, increased brain capillary pericyte density, increased expression of BBB tight junction proteins, reduced brain infiltration of immunoglobulin, and attenuated neuroinflammation. These changes were accompanied by cessation of TBI-induced chronic axonal degeneration. Specificity for P7C3-A20 action on the endothelium was confirmed by protection of cultured human brain microvascular endothelial cells from hydrogen peroxide-induced cell death, as well as preservation of BBB integrity in mice after exposure to toxic levels of lipopolysaccharide. P7C3-A20 also protected mice from BBB degradation after acute TBI. Collectively, our results provide insights into the pathophysiologic mechanisms behind chronic neurodegeneration after TBI, along with a putative treatment strategy. Because TBI increases the risks of other forms of neurodegeneration involving BBB deterioration (e.g., Alzheimer's disease, Parkinson's disease, vascular dementia, chronic traumatic encephalopathy), P7C3-A20 may have widespread clinical utility in the setting of neurodegenerative conditions.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/drug therapy , Carbazoles/pharmacology , Cognition/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Blood-Brain Barrier/cytology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Carbazoles/therapeutic use , Cells, Cultured , Chronic Disease/drug therapy , Cognition/physiology , Disease Models, Animal , Endothelial Cells , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Male , Mice , Microscopy, Electron , Microvessels/cytology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Survivors
7.
FASEB J ; 34(1): 95-106, 2020 01.
Article in English | MEDLINE | ID: mdl-31914697

ABSTRACT

Diabetic nephropathy (DN) is one of the leading causes of mortality in diabetic patients, but its pathogenesis is unclear. We aimed to study the role of the pro-ANP convertase Corin in the pathogenesis of DN. Corin and ANP expression in DN rat kidneys and high-glucose-treated HK-2 cells was analyzed by real-time PCR, western blotting, and immunohistochemical staining. The effect of Corin-siRNA or ANP-siRNA HK-2 cells on EA.hy926 cell migration was determined by scratch-wound healing assay. The expression of mitogen-activated protein kinase (MAPK) and endothelial NO synthase (eNOS) in EA.hy926 cells treated with conditioned medium from Corin-siRNA- or ANP-siRNA-transfected HK-2 cells was determined by western blotting. We found a significant reduction in Corin and ANP expression in DN rat kidneys. These results were recapitulated in HK-2 cells treated with high glucose. EA.hy926 cells treated with conditioned medium from Corin-deficient HK-2 cells had inhibited migration, increased MAPK activity, and decreased eNOS activity. Similar effects were observed with ANP-siRNA transfection. Finally, adding ANP to the Corin-deficient HK-2 conditioned medium rescued the above defects, indicating that Corin mediates its effects through ANP. In conclusion, Corin plays a renoprotective role through pro-ANP processing, and defects in Corin cause endothelial dysfunction through MAPK and eNOS signaling in DN.


Subject(s)
Diabetic Nephropathies/metabolism , Endothelium/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitric Oxide Synthase Type III/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Line , Cell Proliferation , Cell Survival , Diabetes Mellitus, Experimental , Endothelium/metabolism , Gene Expression Regulation/drug effects , Glucose/toxicity , Human Umbilical Vein Endothelial Cells , Humans , Kidney Tubules, Proximal/cytology , Male , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Nitric Oxide Synthase Type III/genetics , RNA Interference , RNA, Small Interfering , Rats, Sprague-Dawley , Serine Endopeptidases/genetics , Serine Endopeptidases/urine
8.
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
9.
Acta Neuropathol ; 137(6): 939-960, 2019 06.
Article in English | MEDLINE | ID: mdl-30877431

ABSTRACT

Both α-Synuclein (αSyn) accumulation and mitochondrial dysfunction have been implicated in the pathology of Parkinson's disease (PD). Although studies suggest that αSyn and its missense mutant, A53T, preferentially accumulate in the mitochondria, the mechanisms by which αSyn and mitochondrial proteins regulate each other to trigger mitochondrial and neuronal toxicity are poorly understood. ATP-dependent Clp protease (ClpP), a mitochondrial matrix protease, plays an important role in regulating mitochondrial protein turnover and bioenergetics activity. Here, we show that the protein level of ClpP is selectively decreased in αSyn-expressing cell culture and neurons derived from iPS cells of PD patient carrying αSyn A53T mutant, and in dopaminergic (DA) neurons of αSyn A53T mice and PD patient postmortem brains. Deficiency in ClpP induces an overload of mitochondrial misfolded/unfolded proteins, suppresses mitochondrial respiratory activity, increases mitochondrial oxidative damage and causes cell death. Overexpression of ClpP reduces αSyn-induced mitochondrial oxidative stress through enhancing the level of Superoxide Dismutase-2 (SOD2), and suppresses the accumulation of αSyn S129 phosphorylation and promotes neuronal morphology in neurons derived from PD patient iPS cells carrying αSyn A53T mutant. Moreover, we find that αSyn WT and A53T mutant interact with ClpP and suppress its peptidase activity. The binding of αSyn to ClpP further promotes a distribution of ClpP from soluble to insoluble cellular fraction in vitro and in vivo, leading to reduced solubility of ClpP. Compensating for the loss of ClpP in the substantia nigra of αSyn A53T mice by viral expression of ClpP suppresses mitochondrial oxidative damage, and reduces αSyn pathology and behavioral deficits of mice. Our findings provide novel insights into the mechanism underlying αSyn-induced neuronal pathology, and they suggest that ClpP might be a useful therapeutic target for PD and other synucleinopathies.


Subject(s)
Endopeptidase Clp/physiology , Mitochondria/enzymology , Mutation, Missense , Nerve Tissue Proteins/physiology , Parkinson Disease/genetics , alpha-Synuclein/physiology , Animals , Cell Respiration , Cells, Cultured , Dopaminergic Neurons/metabolism , Endopeptidase Clp/antagonists & inhibitors , Endopeptidase Clp/deficiency , Gain of Function Mutation , Genes, Reporter , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Oxidative Stress , Protein Folding , Protein Interaction Mapping , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species , Recombinant Proteins/metabolism , Solubility , Substantia Nigra/metabolism , Superoxide Dismutase/metabolism , alpha-Synuclein/genetics
10.
Circulation ; 135(7): 683-699, 2017 02 14.
Article in English | MEDLINE | ID: mdl-27899394

ABSTRACT

BACKGROUND: Ventricular arrhythmia is a leading cause of cardiac mortality. Most antiarrhythmics present paradoxical proarrhythmic side effects, culminating in a greater risk of sudden death. METHODS: We describe a new regulatory mechanism linking mitogen-activated kinase kinase-7 deficiency with increased arrhythmia vulnerability in hypertrophied and failing hearts using mouse models harboring mitogen-activated kinase kinase-7 knockout or overexpression. The human relevance of this arrhythmogenic mechanism is evaluated in human-induced pluripotent stem cell-derived cardiomyocytes. Therapeutic potentials by targeting this mechanism are explored in the mouse models and human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS: Mechanistically, hypertrophic stress dampens expression and phosphorylation of mitogen-activated kinase kinase-7. Such mitogen-activated kinase kinase-7 deficiency leaves histone deacetylase-2 unphosphorylated and filamin-A accumulated in the nucleus to form a complex with Krüppel-like factor-4. This complex leads to Krüppel-like factor-4 disassociation from the promoter regions of multiple key potassium channel genes (Kv4.2, KChIP2, Kv1.5, ERG1, and Kir6.2) and reduction of their transcript levels. Consequent repolarization delays result in ventricular arrhythmias. Therapeutically, targeting the repressive function of the Krüppel-like factor-4/histone deacetylase-2/filamin-A complex with the histone deacetylase-2 inhibitor valproic acid restores K+ channel expression and alleviates ventricular arrhythmias in pathologically remodeled hearts. CONCLUSIONS: Our findings unveil this new gene regulatory avenue as a new antiarrhythmic target where repurposing of the antiepileptic drug valproic acid as an antiarrhythmic is supported.


Subject(s)
Arrhythmias, Cardiac/prevention & control , MAP Kinase Kinase 7/metabolism , Animals , Arrhythmias, Cardiac/physiopathology , Epigenesis, Genetic , Humans , Kruppel-Like Factor 4 , Mice , Myocytes, Cardiac/metabolism , Rats
11.
J Mol Cell Cardiol ; 112: 64-73, 2017 11.
Article in English | MEDLINE | ID: mdl-28882480

ABSTRACT

Heart failure is associated with mitochondrial dysfunction so that restoring or improving mitochondrial health is of therapeutic importance. Recently, reduction in NAD+ levels and NAD+-mediated deacetylase activity has been recognized as negative regulators of mitochondrial function. Using a cardiac specific KLF4 deficient mouse line that is sensitive to stress, we found mitochondrial protein hyperacetylation coupled with reduced Sirt3 and NAD+ levels in the heart before stress, suggesting that the KLF4-deficient heart is predisposed to NAD+-associated defects. Further, we demonstrated that short-term administration of Nicotinamide Mononucleotide (NMN) successfully protected the mutant mice from pressure overload-induced heart failure. Mechanically, we showed that NMN preserved mitochondrial ultrastructure, reduced ROS and prevented cell death in the heart. In cultured cardiomyocytes, NMN treatment significantly increased long-chain fatty acid oxidation despite no direct effect on pyruvate oxidation. Collectively, these results provide cogent evidence that hyperacetylation of mitochondrial proteins is critical in the pathogenesis of cardiac disease and that administration of NMN may serve as a promising therapy.


Subject(s)
Heart Failure/metabolism , Heart Failure/prevention & control , Homeostasis , Nicotinamide Mononucleotide/administration & dosage , Nicotinamide Mononucleotide/therapeutic use , Acetylation , Animals , Cell Death , Fatty Acids/metabolism , Heart Failure/pathology , Homeostasis/drug effects , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , NAD/metabolism , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidation-Reduction , Pressure , Rats , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism
12.
Vasc Med ; 22(5): 363-369, 2017 10.
Article in English | MEDLINE | ID: mdl-28825355

ABSTRACT

Hemoglobin subunit alpha (HBA) expression in endothelial cells (ECs) has recently been shown to control vascular tone and function. We sought to elucidate the transcriptional regulation of HBA expression in the EC. Gain of KLF2 or KLF4 function studies led to significant induction of HBA in ECs. An opposite effect was observed in ECs isolated from animals with endothelial-specific ablation of Klf2, Klf4 or both. Promoter reporter assays demonstrated that KLF2/KLF4 transactivated the hemoglobin alpha promoter, an effect that was abrogated following mutation of all four putative KLF-binding sites. Fine promoter mutational studies localized three out of four KLF-binding sites (sites 2, 3, and 4) as critical for the transactivation of the HBA promoter by KLF2/KLF4. Chromatin immunoprecipitation studies showed that KLF4 bound to the HBA promoter in ECs. Thus, KLF2 and KLF4 serve as important regulators that promote HBA expression in the endothelium.


Subject(s)
Endothelial Cells/metabolism , Hemoglobins/metabolism , Kruppel-Like Transcription Factors/metabolism , Peptide Fragments/metabolism , Animals , Binding Sites , Cattle , Cells, Cultured , Genotype , Hemoglobins/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Mutation , Peptide Fragments/genetics , Phenotype , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic , Transcriptional Activation , Transfection , Up-Regulation
13.
J Biol Chem ; 289(17): 12016-12028, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24599951

ABSTRACT

Regulation of endothelial cell biology by the Notch signaling pathway (Notch) is essential to vascular development, homeostasis, and sprouting angiogenesis. Although Notch determines cell fate and differentiation in a wide variety of cells, the molecular basis of upstream regulation of Notch remains poorly understood. Our group and others have implicated the Krüppel-like factor family of transcription factors as critical regulators of endothelial function. Here, we show that Krüppel-like factor 4 (KLF4) is a central regulator of sprouting angiogenesis via regulating Notch. Using a murine model in which KLF4 is overexpressed exclusively in the endothelium, we found that sustained expression of KLF4 promotes ineffective angiogenesis leading to diminished tumor growth independent of endothelial cell proliferation or cell cycling effects. These tumors feature increased vessel density yet are hypoperfused, leading to tumor hypoxia. Mechanistically, we show that KLF4 differentially regulates expression of Notch receptors, ligands, and target genes. We also demonstrate that KLF4 limits cleavage-mediated activation of Notch1. Finally, we rescue Notch target gene expression and the KLF4 sprouting angiogenesis phenotype by supplementation of DLL4 recombinant protein. Identification of this hitherto undiscovered role of KLF4 implicates this transcription factor as a critical regulator of Notch, tumor angiogenesis, and sprouting angiogenesis.


Subject(s)
Endothelium, Vascular/metabolism , Kruppel-Like Transcription Factors/physiology , Neovascularization, Pathologic , Receptors, Notch/metabolism , Signal Transduction , Animals , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Gene Knockdown Techniques , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Transgenic
14.
J Biol Chem ; 289(9): 5914-24, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24407292

ABSTRACT

The mammalian heart, the body's largest energy consumer, has evolved robust mechanisms to tightly couple fuel supply with energy demand across a wide range of physiologic and pathophysiologic states, yet, when compared with other organs, relatively little is known about the molecular machinery that directly governs metabolic plasticity in the heart. Although previous studies have defined Kruppel-like factor 15 (KLF15) as a transcriptional repressor of pathologic cardiac hypertrophy, a direct role for the KLF family in cardiac metabolism has not been previously established. We show in human heart samples that KLF15 is induced after birth and reduced in heart failure, a myocardial expression pattern that parallels reliance on lipid oxidation. Isolated working heart studies and unbiased transcriptomic profiling in Klf15-deficient hearts demonstrate that KLF15 is an essential regulator of lipid flux and metabolic homeostasis in the adult myocardium. An important mechanism by which KLF15 regulates its direct transcriptional targets is via interaction with p300 and recruitment of this critical co-activator to promoters. This study establishes KLF15 as a key regulator of myocardial lipid utilization and is the first to implicate the KLF transcription factor family in cardiac metabolism.


Subject(s)
DNA-Binding Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Line , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardium/pathology , Nuclear Proteins/genetics , Oxidation-Reduction , Transcription Factors/genetics
15.
Arterioscler Thromb Vasc Biol ; 33(6): 1135-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23640482

ABSTRACT

Macrophages are key regulators of many organ systems, including innate and adaptive immunity, systemic metabolism, hematopoiesis, vasculogenesis, malignancy, and reproduction. The pleiotropic roles of macrophages are mirrored by similarly diverse cellular phenotypes. A simplified schema classifies macrophages as M1, classically activated macrophages, or M2, alternatively activated macrophages. These cells are characterized by their expression of cell surface markers, secreted cytokines and chemokines, and transcription and epigenetic pathways. Transcriptional regulation is central to the differential speciation of macrophages, and several major pathways have been described as essential for subset differentiation. In this review, we discuss the transcriptional regulation of macrophages.


Subject(s)
Adaptive Immunity/genetics , Immunity, Innate/genetics , Macrophage Activation/immunology , Macrophages/immunology , Transcriptional Activation/genetics , Adaptive Immunity/immunology , Animals , Antigens, Surface/genetics , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Immunity, Innate/immunology , Macrophage Activation/genetics , Macrophages/cytology , Mice , Sensitivity and Specificity , Signal Transduction
16.
Nat Cancer ; 5(4): 572-589, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38291304

ABSTRACT

Acquired drug resistance is a major challenge for cancer therapy and is the leading cause of cancer mortality; however, the mechanisms of drug resistance are diverse and the strategy to specifically target drug-resistant cancer cells remains an unmet clinical issue. Here, we established a colorectal cancer-derived organoid biobank and induced acquired drug resistance by repeated low-level exposures of chemo-agents. Chemosensitivity profiling and transcriptomic analysis studies revealed that chemoresistant cancer-derived organoids exhibited elevated expression of LGR4 and activation of the Wnt signaling pathway. Further, we generated a monoclonal antibody (LGR4-mAb) that potently inhibited LGR4-Wnt signaling and found that treatment with LGR4-mAb notably sensitized drug-induced ferroptosis. Mechanistically, LGR4-dependent Wnt signaling transcriptionally upregulated SLC7A11, a key inhibitor of ferroptosis, to confer acquired drug resistance. Our findings reveal that targeting of Wnt signaling by LGR4-mAb augments ferroptosis when co-administrated with chemotherapeutic agents, demonstrating a potential opportunity to fight refractory and recurrent cancers.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Ferroptosis , Receptors, G-Protein-Coupled , Animals , Humans , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Ferroptosis/drug effects , Organoids/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Wnt Signaling Pathway/drug effects
17.
Arterioscler Thromb Vasc Biol ; 32(12): 2836-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23065827

ABSTRACT

OBJECTIVE: To investigate the role of Krüppel-like factor 4 (KLF4), an essential transcriptional regulator of macrophage polarization (M1/M2), in the pathogenesis of atherosclerosis. METHODS AND RESULTS: Despite the acknowledged importance of macrophages in atherosclerosis, the role of M1 (classically activated or proinflammatory) versus M2 (alternatively activated or anti-inflammatory) macrophages in this process remains incompletely understood. We recently identified KLF4 as a regulator of macrophage subset specification; that is, KLF4 promotes M2 and inhibits M1 phenotype. Here, we provide evidence that KLF4-deficient macrophages exhibit enhanced proinflammatory activation and foam cell formation in response to oxidized lipids. In vivo, myeloid KLF4-deficient mice (ApoE(-/-) background) develop significantly more vascular inflammation and atherosclerotic lesion formation. CONCLUSIONS: Our findings identify myeloid KLF4 as an essential regulator of vascular inflammation and experimental atherogenesis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/physiopathology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Atherosclerosis/pathology , Disease Models, Animal , Foam Cells/pathology , Foam Cells/physiology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Macrophages/pathology , Macrophages/physiology , Mice , Mice, Knockout , Phenotype
18.
J Am Heart Assoc ; 12(4): e024303, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36789992

ABSTRACT

Background Proper function of endothelial cells is critical for vascular integrity and organismal survival. Studies over the past 2 decades have identified 2 members of the KLF (Krüppel-like factor) family of proteins, KLF2 and KLF4, as nodal regulators of endothelial function. Strikingly, inducible postnatal deletion of both KLF2 and KLF4 resulted in widespread vascular leak, coagulopathy, and rapid death. Importantly, while transcriptomic studies revealed profound alterations in gene expression, the molecular mechanisms underlying these changes remain poorly understood. Here, we seek to determine mechanisms of KLF2 and KLF4 transcriptional control in multiple vascular beds to further understand their roles as critical endothelial regulators. Methods and Results We integrate chromatin occupancy and transcription studies from multiple transgenic mouse models to demonstrate that KLF2 and KLF4 have overlapping yet distinct binding patterns and transcriptional targets in heart and lung endothelium. Mechanistically, KLFs use open chromatin regions in promoters and enhancers and bind in context-specific patterns that govern transcription in microvasculature. Importantly, this occurs during homeostasis in vivo without additional exogenous stimuli. Conclusions Together, this work provides mechanistic insight behind the well-described transcriptional and functional heterogeneity seen in vascular populations, while also establishing tools into exploring microvascular endothelial dynamics in vivo.


Subject(s)
Endothelium , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Animals , Mice , Chromatin/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Gene Expression , Kruppel-Like Factor 4/genetics , Kruppel-Like Factor 4/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
19.
Sci Total Environ ; 904: 166667, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37652374

ABSTRACT

Lead (Pb) is widely used in industrial and daily-use consumer products. Early-life exposure may increase the risk of lead-related heart problems in childhood. However, the effects of early-life lead exposure on fetal heart development and long-term cardiac outcomes are unknown. In this study, pregnant ICR mice were exposed to lead acetate trihydrate (50 mg/kg/d) via oral gavage from gestation day 1.5 until offspring weaning. Thereafter, the second hit model was established, two groups of offspring (4 weeks old) were either administered sterile saline or Angiotensin II (Ang II) for 4 weeks until euthanasia. We investigated lead-induced offspring heart damage from embryonic period to adulthood by echocardiographic analysis, pathological H&E staining, and ultrastructural examination, as well as mitochondrial function detection. The results showed early-life lead exposure predisposed offspring mice to decreased ejection fraction, increased left ventricular volume, accompanied by hypertrophy and dilation, cardiomyocyte sarcomere dysplasia, abnormal mitochondrial structure, mitochondrial dysfunction, and decreased expression of key sarcomeric and mitochondrial genes, rendering them more susceptible to cardiac hypertrophy, vascular wall thickening, cardiac fibrosis, apoptosis, and heart failure induced by Ang II infusion. This study elucidates early-life low dose lead exposure compromises cardiac development and exacerbates second hit-induced cardiac pathological responses in adulthood, which furnishes crucial scientific evidence pertaining to the cardiac toxicity and risk evaluation associated with early-life exposure to lead.


Subject(s)
Cardiomegaly , Lead , Humans , Pregnancy , Female , Mice , Animals , Lead/toxicity , Lead/metabolism , Mice, Inbred ICR , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Myocytes, Cardiac , Blood Pressure , Angiotensin II/pharmacology , Angiotensin II/toxicity
20.
J Cardiovasc Dev Dis ; 10(11)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37998519

ABSTRACT

Integrin ß plays an important role in the pathogenesis of thrombosis and inflammation, and it may be a shared pathogenic mechanism between arterial and venous thromboses. With the goal of identifying new treatment targets for thrombotic diseases and specific diagnostic markers for venous thromboembolism (VTE), this prospective clinical study was performed to clarify the relationship between integrin and thrombosis. The levels of integrin ß1-3, interleukin-6 (IL-6), and C-reactive protein were significantly higher in patients with acute myocardial infarction (AMI; n = 44) and acute VTE (n = 43) compared to healthy controls (n = 33). The IL-6 and integrin ß1-3 levels were also significantly higher in the AMI group compared to the VTE and control groups. Logistic regression analysis identified IL-6 and integrin ß1-3 levels as independent risk factors for thrombotic disease. Based on the receiver-operating characteristic curve, Youden index, sensitivity, and specificity, the diagnostic accuracy value for VTE was greater than 0.8 when integrins ß1, ß2, and ß3 were combined. Overall, these results suggest that integrin ß levels can contribute to improving the diagnosis and treatment of arteriovenous thrombosis.

SELECTION OF CITATIONS
SEARCH DETAIL