Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Biophys J ; 122(18): 3690-3703, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37254483

ABSTRACT

Fetal lung fibroblasts contribute dynamic infrastructure for the developing lung. These cells undergo dynamic mechanical transitions, including cyclic stretch and spreading, which are integral to lung growth in utero. We investigated the role of the nuclear envelope protein emerin in cellular responses to these dynamic mechanical transitions. In contrast to control cells, which briskly realigned their nuclei, actin cytoskeleton, and extracellular matrices in response to cyclic stretch, fibroblasts that were acutely downregulated for emerin showed incomplete reorientation of both nuclei and actin cytoskeleton. Emerin-downregulated fibroblasts were also aberrantly circular in contrast to the spindle-shaped controls and exhibited an altered pattern of filamentous actin organization that was disconnected from the nucleus. Emerin knockdown was also associated with reduced myosin light chain phosphorylation during cell spreading. Interestingly, emerin-downregulated fibroblasts also demonstrated reduced fibronectin fibrillogenesis and production. These findings indicate that nuclear-cytoskeletal coupling serves a role in the dynamic regulation of cytoskeletal structure and function and may also impact the transmission of traction force to the extracellular matrix microenvironment.


Subject(s)
Actomyosin , Cytoskeleton , Actomyosin/metabolism , Down-Regulation , Cytoskeleton/metabolism , Actin Cytoskeleton/metabolism
2.
J Med Genet ; 59(9): 906-911, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34493544

ABSTRACT

BACKGROUND: The molecular genetic basis of pulmonary arterial hypertension (PAH) is heterogeneous, with at least 26 genes displaying putative evidence for disease causality. Heterozygous variants in the ATP13A3 gene were recently identified as a new cause of adult-onset PAH. However, the contribution of ATP13A3 risk alleles to child-onset PAH remains largely unexplored. METHODS AND RESULTS: We report three families with a novel, autosomal recessive form of childhood-onset PAH due to biallelic ATP13A3 variants. Disease onset ranged from birth to 2.5 years and was characterised by high mortality. Using genome sequencing of parent-offspring trios, we identified a homozygous missense variant in one case, which was subsequently confirmed to cosegregate with disease in an affected sibling. Independently, compound heterozygous variants in ATP13A3 were identified in two affected siblings and in an unrelated third family. The variants included three loss of function variants (two frameshift, one nonsense) and two highly conserved missense substitutions located in the catalytic phosphorylation domain. The children were largely refractory to treatment and four died in early childhood. All parents were heterozygous for the variants and asymptomatic. CONCLUSION: Our findings support biallelic predicted deleterious ATP13A3 variants in autosomal recessive, childhood-onset PAH, indicating likely semidominant dose-dependent inheritance for this gene.


Subject(s)
Pulmonary Arterial Hypertension , Adenosine Triphosphatases/genetics , Adult , Child, Preschool , Familial Primary Pulmonary Hypertension/genetics , Heterozygote , Homozygote , Humans , Membrane Transport Proteins/genetics , Morbidity
3.
Proc Natl Acad Sci U S A ; 116(8): 3278-3287, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718399

ABSTRACT

Cells express a family of three inositol hexakisphosphate kinases (IP6Ks). Although sharing the same enzymatic activity, individual IP6Ks mediate different cellular processes. Here we report that IP6K3 is enriched at the leading edge of migrating cells where it associates with dynein intermediate chain 2 (DIC2). Using immunofluorescence microscopy and total internal reflection fluorescence microscopy, we found that DIC2 and IP6K3 are recruited interdependently to the leading edge of migrating cells, where they function coordinately to enhance the turnover of focal adhesions. Deletion of IP6K3 causes defects in cell motility and neuronal dendritic growth, eventually leading to brain malformations. Our results reveal a mechanism whereby IP6K3 functions in coordination with DIC2 in a confined intracellular microenvironment to promote focal adhesion turnover.


Subject(s)
Cytoplasmic Dyneins/genetics , Dendrites/genetics , Phosphotransferases (Phosphate Group Acceptor)/genetics , Brain/metabolism , Brain/pathology , Cell Adhesion/genetics , Cell Movement/genetics , Cellular Microenvironment/genetics , Focal Adhesions/genetics , HEK293 Cells , Humans , Neurons/metabolism
4.
Pediatr Crit Care Med ; 21(2): e52-e106, 2020 02.
Article in English | MEDLINE | ID: mdl-32032273

ABSTRACT

OBJECTIVES: To develop evidence-based recommendations for clinicians caring for children (including infants, school-aged children, and adolescents) with septic shock and other sepsis-associated organ dysfunction. DESIGN: A panel of 49 international experts, representing 12 international organizations, as well as three methodologists and three public members was convened. Panel members assembled at key international meetings (for those panel members attending the conference), and a stand-alone meeting was held for all panel members in November 2018. A formal conflict-of-interest policy was developed at the onset of the process and enforced throughout. Teleconferences and electronic-based discussion among the chairs, co-chairs, methodologists, and group heads, as well as within subgroups, served as an integral part of the guideline development process. METHODS: The panel consisted of six subgroups: recognition and management of infection, hemodynamics and resuscitation, ventilation, endocrine and metabolic therapies, adjunctive therapies, and research priorities. We conducted a systematic review for each Population, Intervention, Control, and Outcomes question to identify the best available evidence, statistically summarized the evidence, and then assessed the quality of evidence using the Grading of Recommendations Assessment, Development, and Evaluation approach. We used the evidence-to-decision framework to formulate recommendations as strong or weak, or as a best practice statement. In addition, "in our practice" statements were included when evidence was inconclusive to issue a recommendation, but the panel felt that some guidance based on practice patterns may be appropriate. RESULTS: The panel provided 77 statements on the management and resuscitation of children with septic shock and other sepsis-associated organ dysfunction. Overall, six were strong recommendations, 52 were weak recommendations, and nine were best-practice statements. For 13 questions, no recommendations could be made; but, for 10 of these, "in our practice" statements were provided. In addition, 49 research priorities were identified. CONCLUSIONS: A large cohort of international experts was able to achieve consensus regarding many recommendations for the best care of children with sepsis, acknowledging that most aspects of care had relatively low quality of evidence resulting in the frequent issuance of weak recommendations. Despite this challenge, these recommendations regarding the management of children with septic shock and other sepsis-associated organ dysfunction provide a foundation for consistent care to improve outcomes and inform future research.


Subject(s)
Multiple Organ Failure/therapy , Pediatrics/standards , Sepsis/therapy , Shock, Septic/therapy , Adolescent , Anti-Bacterial Agents/therapeutic use , Child , Child, Preschool , Evidence-Based Medicine , Fluid Therapy/methods , Hemodynamics , Humans , Infant , Infant, Newborn , Lactic Acid/blood , Multiple Organ Failure/diagnosis , Multiple Organ Failure/etiology , Respiration, Artificial/methods , Resuscitation/methods , Sepsis/complications , Sepsis/diagnosis , Shock, Septic/diagnosis , Vasoconstrictor Agents/therapeutic use
5.
Arterioscler Thromb Vasc Biol ; 38(4): 913-926, 2018 04.
Article in English | MEDLINE | ID: mdl-29472234

ABSTRACT

OBJECTIVE: KLF15 (Kruppel-like factor 15) has recently been shown to suppress activation of proinflammatory processes that contribute to atherogenesis in vascular smooth muscle, however, the role of KLF15 in vascular endothelial function is unknown. Arginase mediates inflammatory vasculopathy and vascular injury in pulmonary hypertension. Here, we tested the hypothesis that KLF15 is a critical regulator of hypoxia-induced Arg2 (arginase 2) transcription in human pulmonary microvascular endothelial cells (HPMEC). APPROACH AND RESULTS: Quiescent HPMEC express ample amounts of full-length KLF15. HPMECs exposed to 24 hours of hypoxia exhibited a marked decrease in KLF15 protein levels and a reciprocal increase in Arg2 protein and mRNA. Chromatin immunoprecipitation indicated direct binding of KLF15 to the Arg2 promoter, which was relieved with HPMEC exposure to hypoxia. Furthermore, overexpression of KLF15 in HPMEC reversed hypoxia-induced augmentation of Arg2 abundance and arginase activity and rescued nitric oxide (NO) production. Ectopic KLF15 also reversed hypoxia-induced endothelium-mediated vasodilatation in isolated rat pulmonary artery rings. Mechanisms by which hypoxia regulates KLF15 abundance, stability, and compartmentalization to the nucleus in HPMEC were then investigated. Hypoxia triggered deSUMOylation of KLF15 by SENP1 (sentrin-specific protease 1), and translocation of KLF15 from nucleus to cytoplasm. CONCLUSIONS: KLF15 is a critical regulator of pulmonary endothelial homeostasis via repression of endothelial Arg2 expression. KLF15 abundance and nuclear compartmentalization are regulated by SUMOylation/deSUMOylation-a hypoxia-sensitive process that is controlled by SENP1. Strategies including overexpression of KLF15 or inhibition of SENP1 may represent novel therapeutic targets for pulmonary hypertension.


Subject(s)
Arginase/metabolism , Cysteine Endopeptidases/metabolism , Endothelial Cells/enzymology , Kruppel-Like Transcription Factors/metabolism , Lung/blood supply , Microvessels/enzymology , Nuclear Proteins/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , Arginase/genetics , Cell Hypoxia , Cells, Cultured , Cysteine Endopeptidases/genetics , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic , Humans , Kruppel-Like Transcription Factors/genetics , Microvessels/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats , Signal Transduction , Sumoylation , Vasodilation
6.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L93-L106, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28882814

ABSTRACT

We recently demonstrated that blue light induces vasorelaxation in the systemic mouse circulation, a phenomenon mediated by the nonvisual G protein-coupled receptor melanopsin (Opsin 4; Opn4). Here we tested the hypothesis that nonvisual opsins mediate photorelaxation in the pulmonary circulation. We discovered Opsin 3 (Opn3), Opn4, and G protein-coupled receptor kinase 2 (GRK2) in rat pulmonary arteries (PAs) and in pulmonary arterial smooth muscle cells (PASMCs), where the opsins interact directly with GRK2, as demonstrated with a proximity ligation assay. Light elicited an intensity-dependent relaxation of PAs preconstricted with phenylephrine (PE), with a maximum response between 400 and 460 nm (blue light). Wavelength-specific photorelaxation was attenuated in PAs from Opn4-/- mice and further reduced following shRNA-mediated knockdown of Opn3. Inhibition of GRK2 amplified the response and prevented physiological desensitization to repeated light exposure. Blue light also prevented PE-induced constriction in isolated PAs, decreased basal tone, ablated PE-induced single-cell contraction of PASMCs, and reversed PE-induced depolarization in PASMCs when GRK2 was inhibited. The photorelaxation response was modulated by soluble guanylyl cyclase but not by protein kinase G or nitric oxide. Most importantly, blue light induced significant vasorelaxation of PAs from rats with chronic pulmonary hypertension and effectively lowered pulmonary arterial pressure in isolated intact perfused rat lungs subjected to acute hypoxia. These findings show that functional Opn3 and Opn4 in PAs represent an endogenous "optogenetic system" that mediates photorelaxation in the pulmonary vasculature. Phototherapy in conjunction with GRK2 inhibition could therefore provide an alternative treatment strategy for pulmonary vasoconstrictive disorders.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Hypertension, Pulmonary/radiotherapy , Phototherapy , Pulmonary Artery/radiation effects , Rod Opsins/physiology , Vasodilation/radiation effects , Animals , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypoxia/complications , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects , Nitric Oxide/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Soluble Guanylyl Cyclase/genetics , Soluble Guanylyl Cyclase/metabolism , Vasodilation/physiology
7.
Small ; 14(27): e1702497, 2018 07.
Article in English | MEDLINE | ID: mdl-29749014

ABSTRACT

Interfacing nano/microscale elements with biological components in 3D contexts opens new possibilities for mimicry, bionics, and augmentation of organismically and anatomically inspired materials. Abiotic nanoscale elements such as plasmonic nanostructures, piezoelectric ribbons, and thin film semiconductor devices interact with electromagnetic fields to facilitate advanced capabilities such as communication at a distance, digital feedback loops, logic, and memory. Biological components such as proteins, polynucleotides, cells, and organs feature complex chemical synthetic networks that can regulate growth, change shape, adapt, and regenerate. Abiotic and biotic components can be integrated in all three dimensions in a well-ordered and programmed manner with high tunability, versatility, and resolution to produce radically new materials and hybrid devices such as sensor fabrics, anatomically mimetic microfluidic modules, artificial tissues, smart prostheses, and bionic devices. In this critical Review, applications of small scale devices in 3D hybrid integration, biomicrofluidics, advanced prostheses, and bionic organs are discussed.

8.
Am J Physiol Heart Circ Physiol ; 312(4): H711-H720, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28188215

ABSTRACT

Endothelial cystathionine γ-lyase (CSEγ) contributes to cardiovascular homeostasis, mainly through production of H2S. However, the molecular mechanisms that control CSEγ gene expression in the endothelium during cardiovascular diseases are unclear. The aim of the current study is to determine the role of specific histone deacetylases (HDACs) in the regulation of endothelial CSEγ. Reduced CSEγ mRNA expression and protein abundance were observed in human aortic endothelial cells (HAEC) exposed to oxidized LDL (OxLDL) and in aortas from atherogenic apolipoprotein E knockout (ApoE-/-) mice fed a high-fat diet compared with controls. Intact murine aortic rings exposed to OxLDL (50 µg/ml) for 24 h exhibited impaired endothelium-dependent vasorelaxation that was blocked by CSEγ overexpression or the H2S donor NaHS. CSEγ expression was upregulated by pan-HDAC inhibitors and by class II-specific HDAC inhibitors, but not by other class-specific inhibitors. The HDAC6 selective inhibitor tubacin and HDAC6-specific siRNA increased CSEγ expression and blocked OxLDL-mediated reductions in endothelial CSEγ expression and CSEγ promoter activity, indicating that HDAC6 is a specific regulator of CSEγ expression. Consistent with this finding, HDAC6 mRNA, protein expression, and activity were upregulated in OxLDL-exposed HAEC, but not in human aortic smooth muscle cells. HDAC6 protein levels in aortas from high-fat diet-fed ApoE-/- mice were comparable to those in controls, whereas HDAC6 activity was robustly upregulated. Together, our findings indicate that HDAC6 is upregulated by atherogenic stimuli via posttranslational modifications and is a critical regulator of CSEγ expression in vascular endothelium. Inhibition of HDAC6 activity may improve endothelial function and prevent or reverse the development of atherosclerosis.NEW & NOTEWORTHY Oxidative injury to endothelial cells by oxidized LDL reduced cystathionine γ-lyase (CSEγ) expression and H2S production, leading to endothelial dysfunction, which was prevented by histone deacetylase 6 (HDAC6) inhibition. Our data suggest HDAC6 as a novel therapeutic target to prevent the development of atherosclerosis.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Animals , Apolipoproteins E/genetics , Cystathionine gamma-Lyase/biosynthesis , Cystathionine gamma-Lyase/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Histone Deacetylase 6 , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Lipoproteins, LDL/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Vasodilation/drug effects , Vasodilation/genetics
10.
Circ Res ; 115(4): 450-9, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24903103

ABSTRACT

RATIONALE: Increased arginase activity contributes to endothelial dysfunction by competition for l-arginine substrate and reciprocal regulation of nitric oxide synthase (NOS). The rapid increase in arginase activity in human aortic endothelial cells exposed to oxidized low-density lipoprotein (OxLDL) is consistent with post-translational modification or subcellular trafficking. OBJECTIVE: To test the hypotheses that OxLDL triggers reverse translocation of mitochondrial arginase 2 (Arg2) to cytosol and Arg2 activation, and that this process is dependent on mitochondrial processing peptidase, lectin-like OxLDL receptor-1 receptor, and rho kinase. METHODS AND RESULTS: OxLDL-triggered translocation of Arg2 from mitochondria to cytosol in human aortic endothelial cells and in murine aortic intima with a concomitant rise in arginase activity. All of these changes were abolished by inhibition of mitochondrial processing peptidase or by its siRNA-mediated knockdown. Rho kinase inhibition and the absence of the lectin-like OxLDL receptor-1 in knockout mice also ablated translocation. Aminoterminal sequencing of Arg2 revealed 2 candidate mitochondrial targeting sequences, and deletion of either of these confined Arg2 to the cytoplasm. Inhibitors of mitochondrial processing peptidase or lectin-like OxLDL receptor-1 knockout attenuated OxLDL-mediated decrements in endothelial-specific NO production and increases in superoxide generation. Finally, Arg2(-/-) mice bred on an ApoE(-/-) background showed reduced plaque load, reduced reactive oxygen species production, enhanced NO, and improved endothelial function when compared with ApoE(-/-) controls. CONCLUSIONS: These data demonstrate dual distribution of Arg2, a protein with an unambiguous mitochondrial targeting sequence, in mammalian cells, and its reverse translocation to cytoplasm by alterations in the extracellular milieu. This novel molecular mechanism drives OxLDL-mediated arginase activation, endothelial NOS uncoupling, endothelial dysfunction, and atherogenesis.


Subject(s)
Aorta/enzymology , Arginase/metabolism , Endothelial Cells/enzymology , Lipoproteins, LDL/metabolism , Metalloendopeptidases/metabolism , Mitochondria/enzymology , rho-Associated Kinases/metabolism , Amino Acid Sequence , Animals , Aorta/drug effects , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arginase/genetics , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Cells, Cultured , Cytosol/enzymology , Disease Models, Animal , Endothelial Cells/drug effects , Enzyme Activation , Humans , Male , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/drug effects , Molecular Sequence Data , Protein Kinase Inhibitors/pharmacology , Protein Transport , RNA Interference , Scavenger Receptors, Class E/deficiency , Scavenger Receptors, Class E/genetics , Signal Transduction , Time Factors , Transfection , rho-Associated Kinases/antagonists & inhibitors , Mitochondrial Processing Peptidase
11.
J Mol Cell Cardiol ; 81: 18-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25655932

ABSTRACT

Emerging evidence strongly supports a role for HDAC2 in the transcriptional regulation of endothelial genes and vascular function. We have recently demonstrated that HDAC2 reciprocally regulates the transcription of Arginase2, which is itself a critical modulator of endothelial function via eNOS. Moreover HDAC2 levels are decreased in response to the atherogenic stimulus OxLDL via a mechanism that is apparently dependent upon proteasomal degradation. NEDDylation is a post-translational protein modification that is tightly linked to ubiquitination and thereby protein degradation. We propose that changes in NEDDylation may modulate vascular endothelial function in part through alterations in the proteasomal degradation of HDAC2. In HAEC, OxLDL exposure augmented global protein NEDDylation. Pre-incubation of mouse aortic rings with the NEDDylation activating enzyme inhibitor, MLN4924, prevented OxLDL-induced endothelial dysfunction. In HAEC, MLN enhanced HDAC2 abundance, decreased expression and activity of Arginase2, and blocked OxLDL-mediated reduction of HDAC2. Additionally, HDAC2 was shown to be a substrate for NEDD8 conjugation and this interaction was potentiated by OxLDL. Further, HDAC2 levels were reciprocally regulated by ectopic expression of NEDD8 and the de-NEDDylating enzyme SENP8. Our findings indicate that the observed improvement in endothelial dysfunction with inhibition of NEDDylation activating enzyme is likely due to an HDAC2-dependent decrease in Arginase2. NEDDylation activating enzyme may therefore be a novel target in endothelial dysfunction and atherogenesis.


Subject(s)
Atherosclerosis/genetics , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Histone Deacetylase 2/genetics , Protein Processing, Post-Translational , Ubiquitins/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Arginase/genetics , Arginase/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line , Cyclopentanes/pharmacology , Endopeptidases/genetics , Endopeptidases/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme Inhibitors/pharmacology , Histone Deacetylase 2/metabolism , Humans , Lipoproteins, LDL/pharmacology , Mice , NEDD8 Protein , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Pyrimidines/pharmacology , Signal Transduction , Tissue Culture Techniques , Ubiquitination , Ubiquitins/metabolism
12.
Arterioscler Thromb Vasc Biol ; 34(7): 1556-1566, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24833798

ABSTRACT

OBJECTIVE: Arginase 2 (Arg2) is a critical target in atherosclerosis because it controls endothelial nitric oxide, proliferation, fibrosis, and inflammation. Regulators of Arg2 transcription in the endothelium have not been characterized. The goal of the current study is to determine the role of specific histone deacetylases (HDACs) in the regulation of endothelial Arg2 transcription and endothelial function. APPROACH AND RESULTS: The HDAC inhibitor trichostatin A increased levels of Arg2 mRNA, protein, and activity in both human aortic endothelial cells and mouse aortic rings. These changes occurred in both time- and dose-dependent patterns and resulted in Arg2-dependent endothelial dysfunction. Trichostatin A and the atherogenic stimulus oxidized low-density lipoprotein enhanced the activity of common promoter regions of Arg2. HDAC inhibition with trichostatin A also decreased endothelial nitric oxide, and these effects were blunted by arginase inhibition. Nonselective class I HDAC inhibitors enhanced Arg2 expression, whereas the only selective inhibitor that increased Arg2 expression was mocetinostat, a selective inhibitor of HDACs 1 and 2. Additionally, mouse aortic rings preincubated with mocetinostat exhibited dysfunctional relaxation. Overexpression of HDAC2 (but not HDAC 1, 3, or 8) cDNA in human aortic endothelial cells suppressed Arg2 expression in a concentration-dependent manner, and siRNA knockdown of HDAC2 enhanced Arg2 expression. Chromatin immunoprecipitation indicated direct binding of HDAC2 to the Arg2 promoter, and HDAC2 overexpression in human aortic endothelial cells blocked oxidized low-density lipoprotein-mediated activation of the Arg2 promoter. Finally, overexpression of HDAC2 blocked oxidized low-density lipoprotein-mediated vascular dysfunction. CONCLUSIONS: HDAC2 is a critical regulator of Arg2 expression and thereby endothelial nitric oxide and endothelial function. Overexpression or activation of HDAC2 represents a novel therapy for endothelial dysfunction and atherosclerosis.


Subject(s)
Arginase/metabolism , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Histone Deacetylase 2/metabolism , Transcription, Genetic , Animals , Arginase/antagonists & inhibitors , Arginase/genetics , Binding Sites , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Gene Expression Regulation, Enzymologic , HEK293 Cells , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic/drug effects , Transfection , Vasodilation , Vasodilator Agents/pharmacology
13.
Pediatr Crit Care Med ; 21(2): 186-195, 2020 02.
Article in English | MEDLINE | ID: mdl-32032264
14.
Pulm Circ ; 13(4): e12305, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37915400

ABSTRACT

Pulmonary hypertension (PH) is a significant health problem that contributes to high morbidity and mortality in diverse cardiac, pulmonary, and systemic diseases in children. Evidence-based advances in PH care have been challenged by a paucity of quality endpoints for assessing clinical course and the lack of robust clinical trial data to guide pharmacologic therapies in children. While the landmark adult AMBITION trial demonstrated the benefit of up-front combination PH therapy with ambrisentan and tadalafil, it remains unknown whether upfront combination therapy leads to more rapid and sustained clinical benefits in children with various categories of PH. In this article, we describe the inception of the Kids Mod PAH Trial, a multicenter Phase III trial, to address whether upfront combination therapy (sildenafil and bosentan vs. sildenafil alone) improves PH outcomes in children, recognizing that marked differences between the etiology and therapeutic response between adults and children exist. The primary endpoint of this study is WHO functional class (FC) 12 months after initiation of study drug therapy. In addition to the primary outcome, secondary endpoints are being assessed, including a composite measure of time to clinical worsening, WHO FC at 24 months, echocardiographic assessment of PH and quantitative assessment of right ventricular function, 6-min walk distance, and NT-proBNP levels. Exploratory endpoints include selected biomarkers, actigraphy, and assessments of quality of life. This study is designed to pave the way for additional clinical trials by establishing a robust infrastructure through the development of a PPHNet Clinical Trials Network.

15.
Adv Sci (Weinh) ; 9(17): e2104649, 2022 06.
Article in English | MEDLINE | ID: mdl-35434926

ABSTRACT

Multicellular organization with precise spatial definition is essential to various biological processes, including morphogenesis, development, and healing in vascular and other tissues. Gradients and patterns of chemoattractants are well-described guides of multicellular organization, but the influences of 3D geometry of soft hydrogels are less well defined. Here, the discovery of a new mode of endothelial cell self-organization guided by combinatorial effects of stiffness and geometry, independent of protein or chemical patterning, is described. Endothelial cells in 2 kPa microwells are found to be ≈30 times more likely to migrate to the edge to organize in ring-like patterns than in stiff 35 kPa microwells. This organization is independent of curvature and significantly more pronounced in 2 kPa microwells with aspect ratio (perimeter/depth) < 25. Physical factors of cells and substrates that drive this behavior are systematically investigated and a mathematical model that explains the organization by balancing the dynamic interaction between tangential cytoskeletal tension, cell-cell, and cell-substrate adhesion is presented. These findings demonstrate the importance of combinatorial effects of geometry and stiffness in complex cellular organization that can be leveraged to facilitate the engineering of bionics and integrated model organoid systems with customized nutrient vascular networks.


Subject(s)
Endothelial Cells , Hydrogels , Cell Adhesion , Endothelial Cells/metabolism , Hydrogels/pharmacology
16.
Nat Cell Biol ; 4(4): 286-93, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11912490

ABSTRACT

Chromophore-assisted laser inactivation (CALI) is a light-mediated technique used to selectively inactivate proteins within cells. Here, we demonstrate that GFP can be used as a CALI reagent to locally inactivate proteins in living cells. We show that focused laser irradiation of EGFP-alpha-actinin expressed in Swiss 3T3 fibroblasts results in the detachment of stress fibres from focal adhesions (FAs), whereas the integrity of FAs, as determined by interference reflection microscopy (IRM), is preserved. Moreover, consistent with a function for focal adhesion kinase (FAK) in FA signalling and not FA structure, laser irradiation of EGFP-FAK did not cause either visible FA damage or stress fibre detachment, although in vitro CALI of isolated EGFP-FAK decreased its kinase activity, but not its binding to paxillin. These data indicate that CALI of specific FA components may be used to precisely dissect the functional significance of individual proteins required for the maintenance of this cytoskeletal structure. In vitro CALI experiments also demonstrated a reduction of EGFP-alpha-actinin binding to the cytoplasmic domain of the beta(1) integrin subunit, but not to actin. Thus, alpha-actinin is essential for the binding of microfilaments to integrins in the FA. CALI-induced changes in alpha-actinin result in the breakage of that link and the subsequent retraction of the stress fibre.


Subject(s)
Luminescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , 3T3 Cells , Actinin/metabolism , Animals , Blotting, Western , Cell Adhesion , Cell Line , Cytological Techniques , Cytoskeleton/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Green Fluorescent Proteins , Integrin beta1/metabolism , Integrins/metabolism , Lasers , Mice , Models, Biological , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Stress Fibers/metabolism , Time Factors
17.
J Cell Biol ; 174(2): 277-88, 2006 Jul 17.
Article in English | MEDLINE | ID: mdl-16847103

ABSTRACT

Focal adhesion kinase (FAK) transduces cell adhesion to the extracellular matrix into proliferative signals. We show that FAK overexpression induced proliferation in endothelial cells, which are normally growth arrested by limited adhesion. Interestingly, displacement of FAK from adhesions by using a FAK-/- cell line or by expressing the C-terminal fragment FRNK also caused an escape of adhesion-regulated growth arrest, suggesting dual positive and negative roles for FAK in growth regulation. Expressing kinase-dead FAK-Y397F in FAK-/- cells prevented uncontrolled growth, demonstrating the antiproliferative function of inactive FAK. Unlike FAK overexpression-induced growth, loss of growth control in FAK-/- or FRNK-expressing cells increased RhoA activity, cytoskeletal tension, and focal adhesion formation. ROCK inhibition rescued adhesion-dependent growth control in these cells, and expression of constitutively active RhoA or ROCK dysregulated growth. These findings demonstrate the ability of FAK to suppress and promote growth, and underscore the importance of multiple mechanisms, even from one molecule, to control cell proliferation.


Subject(s)
Cell Proliferation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Shape , Cells, Cultured , Cytoskeleton/metabolism , Endothelial Cells/cytology , Endothelial Cells/enzymology , Focal Adhesions/metabolism , Growth Inhibitors/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mice , Models, Biological , Protein-Tyrosine Kinases/metabolism , rho-Associated Kinases
18.
Biophys J ; 99(9): L78-80, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21044567

ABSTRACT

Recent work has indicated that the shape and size of a cell can influence how a cell spreads, develops focal adhesions, and exerts forces on the substrate. However, it is unclear how cell shape regulates these events. Here we present a computational model that uses cell shape to predict the magnitude and direction of forces generated by cells. The predicted results are compared to experimentally measured traction forces, and show that the model can predict traction force direction, relative magnitude, and force distribution within the cell using only cell shape as an input. Analysis of the model shows that the magnitude and direction of the traction force at a given point is proportional to the first moment of area about that point in the cell, suggesting that contractile forces within the cell act on the entire cytoskeletal network as a single cohesive unit. Through this model, we demonstrate that intrinsic properties of cell shape can facilitate changes in traction force patterns, independently of heterogeneous mechanical properties or signaling events within the cell.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Cell Shape/physiology , Models, Biological , Animals , Biomechanical Phenomena , Biophysical Phenomena , Cells, Cultured , Cytoskeleton/physiology , Mice , Signal Transduction
19.
Radiat Environ Biophys ; 49(3): 397-404, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20401726

ABSTRACT

Radiation exposure can increase the risk for many non-malignant physiological complications, including cardiovascular disease. We have previously demonstrated that ionizing radiation can induce endothelial dysfunction, which contributes to increased vascular stiffness. In this study, we demonstrate that gamma radiation exposure reduced endothelial cell viability or proliferative capacity using an in vitro aortic angiogenesis assay. Segments of mouse aorta were embedded in a Matrigel-media matrix 1 day after mice received whole-body gamma irradiation between 0 and 20 Gy. Using three-dimensional phase contrast microscopy, we quantified cellular outgrowth from the aorta. Through fluorescent imaging of embedded aortas from Tie2GFP transgenic mice, we determined that the cellular outgrowth is primarily of endothelial cell origin. Significantly less endothelial cell outgrowth was observed in aortas of mice receiving radiation of 5, 10, and 20 Gy radiation, suggesting radiation-induced endothelial injury. Following 0.5 and 1 Gy doses of whole-body irradiation, reduced outgrowth was still detected. Furthermore, outgrowth was not affected by the location of the aortic segments excised along the descending aorta. In conclusion, a single exposure to gamma radiation significantly reduces endothelial cell outgrowth in a dose-dependent manner. Consequently, radiation exposure may inhibit re-endothelialization or angiogenesis after a vascular injury, which would impede vascular recovery.


Subject(s)
Aorta/physiology , Aorta/radiation effects , Neovascularization, Physiologic/radiation effects , Animals , Aorta/cytology , Aorta, Thoracic/cytology , Aorta, Thoracic/physiology , Aorta, Thoracic/radiation effects , Cell Proliferation/radiation effects , Dose-Response Relationship, Radiation , Endothelial Cells/cytology , Endothelial Cells/radiation effects , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Whole-Body Irradiation
20.
Mol Biol Cell ; 18(1): 253-64, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17093062

ABSTRACT

FAK, a cytoplasmic protein tyrosine kinase, is activated and localized to focal adhesions upon cell attachment to extracellular matrix. FAK null cells spread poorly and exhibit altered focal adhesion turnover. Rac1 is a member of the Rho-family GTPases that promotes membrane ruffling, leading edge extension, and cell spreading. We investigated the activation and subcellular location of Rac1 in FAK null and FAK reexpressing fibroblasts. FAK reexpressers had a more robust pattern of Rac1 activation after cell adhesion to fibronectin than the FAK null cells. Translocation of Rac1 to focal adhesions was observed in FAK reexpressers, but seldom in FAK null cells. Experiments with constitutively active L61Rac1 and dominant negative N17Rac1 indicated that the activation state of Rac1 regulated its localization to focal adhesions. We demonstrated that FAK tyrosine-phosphorylated betaPIX and thereby increased its binding to Rac1. In addition, betaPIX facilitated the targeting of activated Rac1 to focal adhesions and the efficiency of cell spreading. These data indicate that FAK has a role in the activation and focal adhesion translocation of Rac1 through the tyrosine phosphorylation of betaPIX.


Subject(s)
Cell Cycle Proteins/metabolism , Extracellular Matrix/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Guanine Nucleotide Exchange Factors/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Movement/drug effects , Extracellular Matrix/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibronectins/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/deficiency , Focal Adhesions/drug effects , Humans , Mice , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Rho Guanine Nucleotide Exchange Factors
SELECTION OF CITATIONS
SEARCH DETAIL