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
1.
Clin Pract Cases Emerg Med ; 5(4): 515-518, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34813461

ABSTRACT

INTRODUCTION: Cranial nerve (CN) VI palsy is a common complaint seen in the emergency department (ED) and has a wide range of causes. Bilateral CN VI palsies are uncommon and appear to be associated with more severe complications. CASE REPORT: A 29-year-old male presented to the ED from an ophthalmology office for diplopia, headache, and strabismus. He was found to have bilateral CN VI palsies and new-onset seizure in the ED. A lumbar puncture revealed cryptococcal meningitis. Additional tests revealed a new diagnosis of human immunodeficiency virus (HIV), acquired immunodeficiency syndrome (AIDS), and syphilis. CONCLUSION: Cryptococcal meningitis remains a life-threatening complication of HIV/AIDS. Coinfections with HIV, particularly syphilis, further complicate a patient's prognosis as both can lead to devastating neurological sequelae. In cryptococcal meningitis, elevated intracranial pressure is a complication that can manifest as seizures, altered mental status, and cranial nerve palsies.

2.
PLoS One ; 7(7): e40048, 2012.
Article in English | MEDLINE | ID: mdl-22802950

ABSTRACT

Cardiac hypertrophy is a well-established risk factor for cardiovascular morbidity and mortality. Activation of G(q/11)-mediated signaling is required for pressure overload-induced cardiomyocyte (CM) hypertrophy to develop. We previously showed that among Regulators of G protein Signaling, RGS2 selectively inhibits G(q/11) signaling and its hypertrophic effects in isolated CM. In this study, we generated transgenic mice with CM-specific, conditional RGS2 expression (dTG) to investigate whether RGS2 overexpression can be used to attenuate G(q/11)-mediated signaling and hypertrophy in vivo. Transverse aortic constriction (TAC) induced a comparable rise in ventricular mass and ANF expression and corresponding hemodynamic changes in dTG compared to wild types (WT), regardless of the TAC duration (1-8 wks) and timing of RGS2 expression (from birth or adulthood). Inhibition of endothelin-1-induced G(q/11)-mediated phospholipase C ß activity in ventricles and atrial appendages indicated functionality of transgenic RGS2. However, the inhibitory effect of transgenic RGS2 on G(q/11)-mediated PLCß activation differed between ventricles and atria: (i) in sham-operated dTG mice the magnitude of the inhibitory effect was less pronounced in ventricles than in atria, and (ii) after TAC, negative regulation of G(q/11) signaling was absent in ventricles but fully preserved in atria. Neither difference could be explained by differences in expression levels, including marked RGS2 downregulation after TAC in left ventricle and atrium. Counter-regulatory changes in other G(q/11)-regulating RGS proteins (RGS4, RGS5, RGS6) and random insertion were also excluded as potential causes. Taken together, despite ample evidence for a role of RGS2 in negatively regulating G(q/11) signaling and hypertrophy in CM, CM-specific RGS2 overexpression in transgenic mice in vivo did not lead to attenuate ventricular G(q/11)-mediated signaling and hypertrophy in response to pressure overload. Furthermore, our study suggests chamber-specific differences in the regulation of RGS2 functionality and potential future utility of the new transgenic model in mitigating G(q/11) signaling in the atria in vivo.


Subject(s)
Cardiomegaly/physiopathology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Myocytes, Cardiac/physiology , RGS Proteins/physiology , Signal Transduction/physiology , Animals , Aorta, Thoracic/surgery , Aortic Diseases/physiopathology , Constriction, Pathologic/physiopathology , Mice , Mice, Transgenic , Phospholipase C beta/metabolism
3.
OMICS ; 13(6): 501-11, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20001863

ABSTRACT

We have previously found that CHF1/Hey2 prevents the development of phenylephrine-induced cardiac hypertrophy. To determine the role of CHF1/Hey2 in pressure overload hypertrophy, we performed ascending aortic banding on wild-type and transgenic mice overexpressing CHF1/Hey2 in the myocardium. We found that both wild-type and transgenic mice developed increased ventricular weight to body weight ratios 1 week after aortic banding. Wild-type mice also developed decreased fractional shortening after 1 week when compared to preoperative echocardiograms and sham-operated controls. Transgenic mice, in comparison, demonstrated preserved fractional shortening. Histological examination of explanted heart tissue demonstrated extensive fibrosis in wild-type hearts, but minimal fibrosis in transgenic hearts. TUNEL staining demonstrated increased apoptosis in the wild-type hearts but not in the transgenic hearts. Exposure of cultured neonatal myocytes from wild-type and transgenic animals to hydrogen peroxide, a potent inducer of apoptosis, demonstrated increased apoptosis in the wild-type cells. Gene Set Analysis of microarray data from wild-type and transgenic hearts 1 week after banding revealed suppression and activation of multiple pathways involving apoptosis, cell signaling, and biosynthesis. These findings demonstrate that CHF1/Hey2 promotes physiological over pathological hypertrophy through suppression of apoptosis and regulation of multiple transcriptional pathways. These findings also suggest that CHF1/Hey2 and its downstream pathways provide a variety of targets for novel heart failure drug discovery, and that genetic polymorphisms in CHF1/Hey2 may affect susceptibility to hypertrophy and heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiomegaly/metabolism , Gene Expression Regulation , Myocardium , Repressor Proteins/metabolism , Signal Transduction/physiology , Transcription, Genetic , Animals , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Blood Pressure/physiology , Cardiomegaly/pathology , Cells, Cultured , Fibrosis/pathology , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Microarray Analysis , Molecular Sequence Data , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Repressor Proteins/genetics
4.
Circ Res ; 103(8): 825-35, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18787193

ABSTRACT

Recently, the side population (SP) phenotype has been introduced as a reliable marker to identify subpopulations of cells with stem/progenitor cell properties in various tissues. We and others have identified SP cells from postmitotic tissues, including adult myocardium, in which they have been suggested to contribute to cellular regeneration following injury. SP cells are identified and characterized by a unique efflux of Hoechst 33342 dye. Abcg2 belongs to the ATP-binding cassette (ABC) transporter superfamily and constitutes the molecular basis for the dye efflux, hence the SP phenotype, in hematopoietic stem cells. Although Abcg2 is also expressed in cardiac SP (cSP) cells, its role in regulating the SP phenotype and function of cSP cells is unknown. Herein, we demonstrate that regulation of the SP phenotype in cSP cells occurs in a dynamic, age-dependent fashion, with Abcg2 as the molecular determinant of the cSP phenotype in the neonatal heart and another ABC transporter, Mdr1, as the main contributor to the SP phenotype in the adult heart. Using loss- and gain-of-function experiments, we find that Abcg2 tightly regulates cell fate and function. Adult cSP cells isolated from mice with genetic ablation of Abcg2 exhibit blunted proliferation capacity and augmented cell death. Conversely, overexpression of Abcg2 is sufficient to enhance cell proliferation, although with a limitation of cardiomyogenic differentiation. In summary, for the first time, we reveal a functional role for Abcg2 in modulating the proliferation, differentiation, and survival of adult cSP cells that goes beyond its distinct role in Hoechst dye efflux.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Myocardium/metabolism , Stem Cells/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Age Factors , Aging/metabolism , Animals , Animals, Newborn , Benzimidazoles/metabolism , Cell Death , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Fluorescent Dyes/metabolism , Male , Mice , Mice, Knockout , Myocardium/cytology , Phenotype , Transduction, Genetic , ATP-Binding Cassette Sub-Family B Member 4
5.
Circulation ; 117(11): 1423-35, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18316486

ABSTRACT

BACKGROUND: Heart failure is the leading cause of death in the United States. By delineating the pathways that regulate cardiomyocyte function, we can better understand the pathogenesis of cardiac disease. Many cardiomyocyte signaling pathways activate protein tyrosine kinases. However, the role of specific protein tyrosine phosphatases (PTPs) in these pathways is unknown. METHODS AND RESULTS: Here, we show that mice with muscle-specific deletion of Ptpn11, the gene encoding the SH2 domain-containing PTP Shp2, rapidly develop a compensated dilated cardiomyopathy without an intervening hypertrophic phase, with signs of cardiac dysfunction appearing by the second postnatal month. Shp2-deficient primary cardiomyocytes are defective in extracellular signal-regulated kinase/mitogen-activated protein kinase (Erk/MAPK) activation in response to a variety of soluble agonists and pressure overload but show hyperactivation of the RhoA signaling pathway. Treatment of primary cardiomyocytes with Erk1/2- and RhoA pathway-specific inhibitors suggests that both abnormal Erk/MAPK and RhoA activities contribute to the dilated phenotype of Shp2-deficient hearts. CONCLUSIONS: Our results identify Shp2 as the first PTP with a critical role in adult cardiac function, indicate that in the absence of Shp2 cardiac hypertrophy does not occur in response to pressure overload, and demonstrate that the cardioprotective role of Shp2 is mediated via control of both the Erk/MAPK and RhoA signaling pathways.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Myocytes, Cardiac/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , LEOPARD Syndrome/enzymology , LEOPARD Syndrome/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Models, Animal , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Organ Specificity , Phenotype , Pressure , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein
6.
Blood ; 111(6): 3236-44, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18096761

ABSTRACT

Pharmacologic activation of the heterodimeric HIF transcription factor appears promising as a strategy to treat diseases, such as anemia, myocardial infarction, and stroke, in which tissue hypoxia is a prominent feature. HIF accumulation is normally linked to oxygen availability because an oxygen-dependent posttranslational modification (prolyl hydroxylation) marks the HIFalpha subunit for polyubiquitination and destruction. Three enzymes (PHD1, PHD2, and PHD3) capable of catalyzing this reaction have been identified, although PHD2 (also called Egln1) appears to be the primary HIF prolyl hydroxylase in cell culture experiments. We found that conditional inactivation of PHD2 in mice is sufficient to activate a subset of HIF target genes, including erythropoietin, leading to striking increases in red blood cell production. Mice lacking PHD2 exhibit premature mortality associated with marked venous congestion and dilated cardiomyopathy. The latter is likely the result of hyperviscosity syndrome and volume overload, although a direct effect of chronic, high-level HIF stimulation on cardiac myocytes cannot be excluded.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heart Failure/enzymology , Heart Failure/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Polycythemia/enzymology , Polycythemia/genetics , Alleles , Animals , Cells, Cultured , Echocardiography , Enzyme Activation , Erythropoiesis/genetics , Gene Expression Regulation, Enzymologic , Heart Failure/diagnostic imaging , Hypoxia-Inducible Factor-Proline Dioxygenases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Polycythemia/pathology , Procollagen-Proline Dioxygenase , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...