Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Nat Genet ; 49(2): 238-248, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067909

ABSTRACT

Arhinia, or absence of the nose, is a rare malformation of unknown etiology that is often accompanied by ocular and reproductive defects. Sequencing of 40 people with arhinia revealed that 84% of probands harbor a missense mutation localized to a constrained region of SMCHD1 encompassing the ATPase domain. SMCHD1 mutations cause facioscapulohumeral muscular dystrophy type 2 (FSHD2) via a trans-acting loss-of-function epigenetic mechanism. We discovered shared mutations and comparable DNA hypomethylation patterning between these distinct disorders. CRISPR/Cas9-mediated alteration of smchd1 in zebrafish yielded arhinia-relevant phenotypes. Transcriptome and protein analyses in arhinia probands and controls showed no differences in SMCHD1 mRNA or protein abundance but revealed regulatory changes in genes and pathways associated with craniofacial patterning. Mutations in SMCHD1 thus contribute to distinct phenotypic spectra, from craniofacial malformation and reproductive disorders to muscular dystrophy, which we speculate to be consistent with oligogenic mechanisms resulting in pleiotropic outcomes.


Subject(s)
Choanal Atresia/genetics , Chromosomal Proteins, Non-Histone/genetics , Genetic Predisposition to Disease/genetics , Microphthalmos/genetics , Muscular Dystrophies/genetics , Mutation/genetics , Nose/abnormalities , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Phenotype
3.
Biomaterials ; 53: 679-87, 2015.
Article in English | MEDLINE | ID: mdl-25890763

ABSTRACT

Therapeutic angiogenesis has achieved promising results for ischemic diseases or peripheral artery disease in preclinical and early-phase clinical studies. We examined the therapeutic angiogenic effects of HPOX, which is biodegradable polymer composing the antioxidant p-hydroxybenzyl alcohol (HBA), in a mouse model of hindlimb ischemia. HPOX effectively stimulated blood flow recovery, compared with its degraded compounds HBA and 1,4-cyclohexendimethanol, via promotion of capillary vessel density in the ischemic hindlimb. These effects were highly correlated with levels of angiogenic inducers, vascular endothelial cell growth factor (VEGF), heme oxygenase-1 (HO-1), and Akt/AMPK/endothelial nitric oxide synthase (eNOS) in ischemic mouse hindlimb muscle. Blood perfusion and neovascularization induced by HPOX were reduced in eNOS(-/-) and HO-1(+/-) mice. HPOX also elevated the endothelial cell markers VEGF receptor-2, CD31, and eNOS mRNAs in the ischemic hindlimb, indicating that HPOX increases endothelial cell population and angiogenesis in the ischemic muscle. However, this nanoparticle suppressed expression levels of several inflammatory genes in ischemic tissues. These results suggest that HPOX significantly promotes angiogenesis and blood flow perfusion in the ischemic mouse hindlimb via increased angiogenic inducers, along with suppression of inflammatory gene expression. Thus, HPOX can be used potentially as a noninvasive drug intervention to facilitate therapeutic angiogenesis.


Subject(s)
Benzyl Alcohols/administration & dosage , Hindlimb/blood supply , Ischemia/therapy , Nanoparticles , Neovascularization, Physiologic , Animals , Benzyl Alcohols/pharmacology , Blood Circulation , Disease Models, Animal , Heme Oxygenase-1/genetics , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics
4.
PLoS One ; 9(9): e108382, 2014.
Article in English | MEDLINE | ID: mdl-25268137

ABSTRACT

AIM: Altered vitamin D signaling is associated with cardiac dysfunction, but the pathogenic mechanism is not clearly understood. We examine the mechanism and the role of vitamin D signaling in the development of cardiac dysfunction. METHODS AND RESULTS: We analyzed 1α-hydroxylase (1α-OHase) knockout (1α-OHase-/-) mice, which lack 1α-OH enzymes that convert the inactive form to hormonally active form of vitamin D. 1α-OHase-/- mice showed modest cardiac hypertrophy at baseline. Induction of pressure overload by transverse aortic constriction (TAC) demonstrated exaggerated cardiac dysfunction in 1α-OHase-/- mice compared to their WT littermates with a significant increase in fibrosis and expression of inflammatory cytokines. Analysis of calcium (Ca2+) transient demonstrated profound Ca2+ handling abnormalities in 1α-OHase-/- mouse cardiomyocytes (CMs), and treatment with paricalcitol (PC), an activated vitamin D3 analog, significantly attenuated defective Ca2+ handling in 1α-OHase-/- CMs. We further delineated the effect of vitamin D deficiency condition to TAC by first correcting the vitamin D deficiency in 1α-OHase-/- mice, followed then by either a daily maintenance dose of vitamin D or vehicle (to achieve vitamin D deficiency) at the time of sham or TAC. In mice treated with vitamin D, there was a significant attenuation of TAC-induced cardiac hypertrophy, interstitial fibrosis, inflammatory markers, Ca2+ handling abnormalities and cardiac function compared to the vehicle treated animals. CONCLUSIONS: Our results provide insight into the mechanism of cardiac dysfunction, which is associated with severely defective Ca2+ handling and defective vitamin D signaling in 1α-OHase-/- mice.


Subject(s)
Aortic Valve Stenosis/metabolism , Calcium/metabolism , Cardiomegaly/metabolism , Signal Transduction , Steroid Hydroxylases/genetics , Vitamin D/metabolism , Animals , Aortic Valve Stenosis/diet therapy , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Cardiomegaly/diet therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Ergocalciferols/pharmacology , Fibrosis , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture , Steroid Hydroxylases/deficiency , Vitamin D/administration & dosage
5.
J Appl Physiol (1985) ; 114(8): 979-87, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23429874

ABSTRACT

Accumulating evidence suggests that vitamin D deficiency plays a crucial role in heart failure. However, whether vitamin D signaling itself plays an important role in cardioprotection is poorly understood. In this study, we examined the mechanism of modulating vitamin D signaling on progression to heart failure after myocardial infarction (MI) in mice. Vitamin D signaling was activated by administration of paricalcitol (PC), an activated vitamin D analog. Wild-type (WT) mice underwent sham or MI surgery and then were treated with either vehicle or PC. Compared with vehicle group, PC attenuated development of heart failure after MI associated with decreases in biomarkers, apoptosis, inflammation, and fibrosis. There was also improvement of cardiac function with PC treatment after MI. Furthermore, vitamin D receptor (VDR) mRNA and protein levels were restored by PC treatment. Next, to explore whether defective vitamin D signaling exhibited deleterious responses after MI, WT and VDR knockout (KO) mice underwent sham or MI surgery and were analyzed 4 wk after MI. VDR KO mice displayed a significant decline in survival rate and cardiac function compared with WT mice after MI. VDR KO mice also demonstrated a significant increase in heart failure biomarkers, apoptosis, inflammation, and fibrosis. Vitamin D signaling promotes cardioprotection after MI through anti-inflammatory, antifibrotic and antiapoptotic mechanisms.


Subject(s)
Ergocalciferols/pharmacology , Heart Failure/prevention & control , Myocardial Infarction/drug therapy , Myocardium/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Vitamins/pharmacology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cells, Cultured , Chemokine CCL2/metabolism , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/metabolism , Disease Models, Animal , Fibrosis , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/complications , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Calcitriol/deficiency , Receptors, Calcitriol/genetics , Recovery of Function , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Ventricular Function, Left/drug effects
6.
Curr Pharm Des ; 17(18): 1783-93, 2011.
Article in English | MEDLINE | ID: mdl-21631422

ABSTRACT

Apoptosis or progress of programmed cell death is a tightly regulated process which plays an important role in various cardiovascular diseases particularly in myocardial infarction, reperfusion injury, and heart failure. Over the past two decades, investigations of several pathways have broadened our understanding of programmed cell death. Many anti-apoptotic interventions have targeted ischemia-reperfusion, however only a limited number have been considered at the chronic stage of heart failure. Endogenous inhibitors, caspase inhibitors, PARP-1 inhibitors, as well as various other agents have been implicated as anti-apoptotic interventions. This review summarizes the apoptotic pathways associated with heart failure, discusses the current anti-apoptotic interventions available and reviews the clinical implications.


Subject(s)
Apoptosis/drug effects , Cardiovascular Agents/therapeutic use , Heart Diseases/drug therapy , Heart Diseases/metabolism , Gene Expression Regulation , Heart Diseases/pathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...