Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Article in English | MEDLINE | ID: mdl-38648392

ABSTRACT

BACKGROUND: Opioid use after revision total hip arthroplasty (rTHA) has not been well characterized. The purpose of this study was to characterize preoperative, perioperative, and postoperative opioid use during rTHA. METHODS: Patients undergoing revision THA from 2010 to 2018 were screened for opioid use 3 months before revision surgery and tracked 24 months postoperatively. Patients were categorized as naïve or tolerant. Opioid prescriptions and average morphine milligram equivalents (MME) were compared between the two groups. RESULTS: One hundred twenty-four of 247 patients (50%) in the tolerant group averaged a preoperative MME of 23.7 mg/day. Postoperatively, tolerant patients received significantly higher daily MME at all time points, including at 3 months 31.4 versus 18.1 mg/day (P < 0.001), 6 months 19.9 versus 2.95 mg/day (P < 0.001), 12 months 14.3 versus 3.5 mg/day (P < 0.001), and 24 months 10.7 versus 2.17 mg/day (P < 0.001). Tolerant patients were more likely to have a prescription at 6 months (44% versus 22%), 12 months (41.4% versus 24%), and 24 months (38% versus 19.3%) (P < 0.001, P = 0.002, P < 0.001, respectively). DISCUSSION: Opioid-tolerant patients had higher postoperative MME requirements for longer recovery duration. Both groups reduced opioid use at 3 months and plateaued at 6 months. These findings can help the revision surgeon counsel patients and expectations.


Subject(s)
Analgesics, Opioid , Arthroplasty, Replacement, Hip , Pain, Postoperative , Reoperation , Humans , Analgesics, Opioid/therapeutic use , Male , Female , Pain, Postoperative/drug therapy , Middle Aged , Aged , Drug Tolerance , Retrospective Studies
2.
J Clin Med ; 10(21)2021 Oct 31.
Article in English | MEDLINE | ID: mdl-34768656

ABSTRACT

It is time to reconsider how we image the breast. Although the breast is a 3D structure, we have traditionally used 2D mammography to perform screening and diagnostic imaging. Mammography has been continuously modified and improved, most recently with tomosynthesis and contrast mammography, but it is still using modifications of compression 2D mammography. It is time to consider 3D imaging for this 3D structure. Cone-beam breast computed tomography (CBBCT) is a revolutionary modality that will assist in overcoming the limitations of current imaging for dense breast tissue and overlapping structures. It also allows easy administration of contrast material for functional imaging. With a radiation dose on par with diagnostic mammography, rapid 10 s acquisition, no breast compression, and true high-resolution isotropic imaging, CBBCT has the potential to usher in a new era in breast imaging. These advantages could translate into lower morbidity and mortality from breast cancer.

4.
J Hypertens ; 38(3): 456-466, 2020 03.
Article in English | MEDLINE | ID: mdl-31714338

ABSTRACT

BACKGROUND: Chromogranin A (CHGA) is an index granin protein critical for biogenesis and exocytotic release of catecholamine storage granules. It is elevated in plasma of patients with sympathetic over-activity and kidney dysfunction. Several CHGA polymorphisms are associated with hypertensive kidney disease. Previously, we unraveled the molecular mechanism by which CHGA expression is regulated in African Americans carrying a genetic variation associated with hypertensive chronic kidney disease (CKD). METHOD: Experimental CKD mouse model were created by 5/6th nephrectomy (Npx) using wild-type and Chga-/- knockout mouse strains to delineate the role of CHGA in CKD. RESULT: Wild-type-Npx mice expressing Chga developed exacerbated azotemia and fibrosis as compared with their knockout-Npx counterparts. Gene expression profiling revealed downregulation of mitochondrial respiratory complexes genes consistent with maladaptive mitochondria in wild-type-Npx mice, contrasted to knockout-Npx. In healthy individuals, an inverse relationship between circulating CHGA levels and glomerular function was observed. In vitro, mesangial cells treated with CHGA-triggered nitric oxide release by a signaling mechanism involving scavenger receptor SR-A. The CHGA-treated and untreated mesangial cells displayed differential expression of cytokine, chemokine, complement, acute phase inflammatory and apoptotic pathway genes. Thus, build-up of plasma CHGA because of kidney injury served as an insult to the mesangial cells resulting in expression of genes promoting inflammation, fibrosis, and progression of CKD. CONCLUSION: These findings improve understanding of the role of elevated CHGA in the progression of CKD and reveal novel pathways that could be exploited for therapeutic strategies in hypertensive kidney disease.


Subject(s)
Chromogranin A , Hypertension, Renal , Nephritis , Animals , Chromogranin A/genetics , Chromogranin A/metabolism , Hypertension, Renal/genetics , Hypertension, Renal/metabolism , Hypertension, Renal/pathology , Mice , Mice, Knockout , Nephritis/genetics , Nephritis/metabolism , Nephritis/pathology
5.
Sci Rep ; 9(1): 5055, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911067

ABSTRACT

The intra-renal dopamine (DA) system is highly expressed in the proximal tubule and contributes to Na+ and blood pressure homeostasis, as well as to the development of nephropathy. In the kidney, the enzyme DOPA Decarboxylase (DDC) originating from the circulation. We used a twin/family study design, followed by polymorphism association analysis at DDC locus to elucidate heritable influences on renal DA production. Dense single nucleotide polymorphism (SNP) genotyping across the DDC locus on chromosome 7p12 was analyzed by re-sequencing guided by trait-associated genetic markers to discover the responsible genetic variation. We also characterized kinetics of the expressed DDC mutant enzyme. Systematic polymorphism screening across the 15-Exon DDC locus revealed a single coding variant in Exon-14 that was associated with DA excretion and multiple other renal traits indicating pleiotropy. When expressed and characterized in eukaryotic cells, the 462Gln variant displayed lower Vmax (maximal rate of product formation by an enzyme) (21.3 versus 44.9 nmol/min/mg) and lower Km (substrate concentration at which half-maximal product formation is achieved by an enzyme.)(36.2 versus 46.8 µM) than the wild-type (Arg462) allele. The highly heritable DA excretion trait is substantially influenced by a previously uncharacterized common coding variant (Arg462Gln) at the DDC gene that affects multiple renal tubular and glomerular traits, and predicts accelerated functional decline in chronic kidney disease.

6.
Semin Ultrasound CT MR ; 39(1): 106-113, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29317032

ABSTRACT

Dedicated breast computed tomography (CT) is the latest in a long history of breast imaging techniques dating back to the 1960s. Breast imaging is performed both for cancer screening as well as for diagnostic evaluation of symptomatic patients. Dedicated breast CT received US Food and Drug Administration approval for diagnostic use in 2015 and is slowly gaining recognition for its value in diagnostic 3-dimensional imaging of the breast, and also for injected contrast-enhanced imaging applications. Conventional mammography has known limitations in sensitivity and specificity, especially in dense breasts. Breast tomosynthesis was US Food and Drug Administration approved in 2011 and is now widely used. Dedicated breast CT is the next technological advance, combining real 3-dimensional imaging with the ease of contrast administration. The lack of painful compression and manipulation of the breasts also makes dedicated breast CT much more acceptable for the patients.


Subject(s)
Breast Neoplasms/diagnostic imaging , Cone-Beam Computed Tomography/methods , Imaging, Three-Dimensional/methods , Mammography/methods , Breast/diagnostic imaging , Female , Humans , Sensitivity and Specificity
7.
Stem Cell Reports ; 8(4): 1086-1100, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410642

ABSTRACT

Large-scale collections of induced pluripotent stem cells (iPSCs) could serve as powerful model systems for examining how genetic variation affects biology and disease. Here we describe the iPSCORE resource: a collection of systematically derived and characterized iPSC lines from 222 ethnically diverse individuals that allows for both familial and association-based genetic studies. iPSCORE lines are pluripotent with high genomic integrity (no or low numbers of somatic copy-number variants) as determined using high-throughput RNA-sequencing and genotyping arrays, respectively. Using iPSCs from a family of individuals, we show that iPSC-derived cardiomyocytes demonstrate gene expression patterns that cluster by genetic background, and can be used to examine variants associated with physiological and disease phenotypes. The iPSCORE collection contains representative individuals for risk and non-risk alleles for 95% of SNPs associated with human phenotypes through genome-wide association studies. Our study demonstrates the utility of iPSCORE for examining how genetic variants influence molecular and physiological traits in iPSCs and derived cell lines.


Subject(s)
Arrhythmias, Cardiac/genetics , Databases, Factual , Genetic Association Studies , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Multigene Family , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Racial Groups
8.
Hum Mol Genet ; 26(1): 233-242, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28011710

ABSTRACT

Chromogranins are pro-hormone secretory proteins released from neuroendocrine cells, with effects on control of blood pressure. We conducted a genome-wide association study for plasma catestatin, the catecholamine release inhibitory peptide derived from chromogranin A (CHGA), and other CHGA- or chromogranin B (CHGB)-related peptides, in 545 US and 1252 Australian subjects. This identified loci on chromosomes 4q35 and 5q34 affecting catestatin concentration (P = 3.40 × 10-30 for rs4253311 and 1.85 × 10-19 for rs2731672, respectively). Genes in these regions include the proteolytic enzymes kallikrein (KLKB1) and Factor XII (F12). In chromaffin cells, CHGA and KLKB1 proteins co-localized in catecholamine storage granules. In vitro, kallikrein cleaved recombinant human CHGA to catestatin, verified by mass spectrometry. The peptide identified from this digestion (CHGA360-373) selectively inhibited nicotinic cholinergic stimulated catecholamine release from chromaffin cells. A proteolytic cascade involving kallikrein and Factor XII cleaves chromogranins to active compounds both in vivo and in vitro.


Subject(s)
Biomarkers/metabolism , Catecholamines/metabolism , Chromaffin Cells/metabolism , Chromogranin A/blood , Genetic Loci/genetics , Hypertension/genetics , Peptide Fragments/blood , Adolescent , Adrenal Glands/metabolism , Adult , Aged , Animals , Australia , Biomarkers/analysis , Cells, Cultured , Factor XII/genetics , Factor XII/metabolism , Female , Genome-Wide Association Study , Humans , Hypertension/blood , Kallikreins/genetics , Kallikreins/metabolism , Male , Mice , Middle Aged , Rats , United States , Young Adult
9.
BMC Med Genet ; 17: 21, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26969407

ABSTRACT

BACKGROUND: Plasma coagulation Factor XIIa (Hageman factor; encoded by F12) and kallikrein (KAL or Fletcher factor; encoded by KLKB1) are proteases of the kallikerin-kinin system involved in converting the inactive circulating prorenin to renin. Renin is a key enzyme in the formation of angiotensin II, which regulates blood pressure, fluid and electrolyte balance and is a biomarker for cardiovascular, metabolic and renal function. The renin-angiotensin system is implicated in extinction learning in posttraumatic stress disorder. METHODS & RESULTS: Active plasma renin was measured from two independent cohorts- civilian twins and siblings, as well as U.S. Marines, for a total of 1,180 subjects. Genotyping these subjects revealed that the carriers of the minor alleles at the two loci- F12 and KLKB1 had a significant association with reduced levels of active plasma renin. Meta-analyses confirmed the association across cohorts. In vitro studies verified digestion of human recombinant pro-renin by kallikrein (KAL) to generate active renin. Subsequently, the active renin was able to digest the synthetic substrate angiotensinogen to angiotensin-I. Examination of mouse juxtaglomerular cell line and mouse kidney sections showed co-localization of KAL with renin. Expression of either REN or KLKB1 was regulated in cell line and rodent models of hypertension in response to oxidative stress, interleukin or arterial blood pressure changes. CONCLUSIONS: The functional variants of KLKB1 (rs3733402) and F12 (rs1801020) disrupted the cascade of enzymatic events, resulting in diminished formation of active renin. Using genetic, cellular and molecular approaches we found that conversion of zymogen prorenin to renin was influenced by these polymorphisms. The study suggests that the variant version of protease factor XIIa due to the amino acid substitution had reduced ability to activate prekallikrein to KAL. As a result KAL has reduced efficacy in converting prorenin to renin and this step of the pathway leading to activation of renin affords a potential therapeutic target.


Subject(s)
Factor XIIa/genetics , Kallikreins/genetics , Polymorphism, Single Nucleotide , Renin-Angiotensin System/genetics , Renin/blood , Adolescent , Adult , Aged , Alleles , Angiotensin I/blood , Angiotensinogen/blood , Animals , Blood Pressure , Cell Cycle Proteins , Cell Line , Gene Expression Regulation , Genetic Loci , Genome-Wide Association Study , Genotyping Techniques , Humans , Hypertension/genetics , Juxtaglomerular Apparatus/cytology , Kallikreins/blood , Male , Mice , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prekallikrein/metabolism , Renin/genetics , Serine Endopeptidases/metabolism , Transferases , Young Adult
10.
Arterioscler Thromb Vasc Biol ; 35(7): 1704-11, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25953646

ABSTRACT

OBJECTIVE: To determine whether biomarkers of oxidized lipoproteins are genetically determined. Lipoprotein(a) (Lp[a]) is a heritable risk factor and carrier of oxidized phospholipids (OxPL). APPROACH AND RESULTS: We measured oxidized phospholipids on apolipoprotein B-containing lipoproteins (OxPL-apoB), Lp(a), IgG, and IgM autoantibodies to malondialdehyde-modified low-density lipoprotein, copper oxidized low-density lipoprotein, and apoB-immune complexes in 386 monozygotic and dizygotic twins to estimate trait heritability (h(2)) and determine specific genetic effects among traits. A genome-wide linkage study followed by genetic association was performed. The h(2) (scale: 0-1) for Lp(a) was 0.91±0.01 and for OxPL-apoB 0.87±0.02, which were higher than physiological, inflammatory, or lipid traits. h(2) of IgM malondialdehyde-modified low-density lipoprotein, copper oxidized low-density lipoprotein, and apoB-immune complexes were 0.69±0.04, 0.67±0.05, and 0.80±0.03, respectively, and for IgG malondialdehyde-modified low-density lipoprotein, copper oxidized low-density lipoprotein, and apoB-immune complexes 0.62±0.05, 0.52±0.06, and 0.53±0.06, respectively. There was an inverse correlation between the major apo(a) isoform and OxPL-apoB (R=-0.49; P<0.001) and Lp(a) (R=-0.48; P<0.001) and OxPL-apoB was modestly correlated with Lp(a) (ρ=0.57; P<0.0001). The correlation in major apo(a) isoform size was concordant (R=1.0; P<0.001) among monozygotic twins but not dizygotic twins (R=0.40; P=0.055). Lp(a) and OxPL-apoB shared genetic codetermination (genetic covariance, ρG=0.774±0.032; P=1.09×10(-38)), although not environmental determination (environmental covariance, ρE=0.081±0.15; P=0.15). In contrast, Lp(a) shared environmental but not genetic codetermination with autoantibodies to malondialdehyde-modified low-density lipoprotein and copper oxidized low-density lipoprotein, and apoB-immune complexes. Sib-pair genetic linkage of the Lp(a) trait revealed that single nucleotide polymorphism rs10455872 was significantly associated with OxPL-apoB after adjusting for Lp(a). CONCLUSIONS: OxPL-apoB and other biomarkers of oxidized lipoproteins are highly heritable cardiovascular risk factors that suggest novel genetic origins of atherothrombosis.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Lipoproteins/blood , Adolescent , Adult , Aged , Aged, 80 and over , Antigen-Antibody Complex/blood , Apolipoproteins B/blood , Apolipoproteins B/immunology , Autoantibodies/blood , Biomarkers/blood , Cholesterol, LDL/blood , Cholesterol, LDL/immunology , Female , Humans , Male , Malondialdehyde/blood , Middle Aged , Oxidation-Reduction , Peptide Fragments/blood , Peptide Fragments/immunology , Phospholipids/blood , Risk Factors , Young Adult
11.
Diabetes ; 64(1): 104-16, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25048197

ABSTRACT

Chromogranin A knockout (Chga-KO) mice exhibit enhanced insulin sensitivity despite obesity. Here, we probed the role of the chromogranin A-derived peptide pancreastatin (PST: CHGA(273-301)) by investigating the effect of diet-induced obesity (DIO) on insulin sensitivity of these mice. We found that on a high-fat diet (HFD), Chga-KO mice (KO-DIO) remain more insulin sensitive than wild-type DIO (WT-DIO) mice. Concomitant with this phenotype is enhanced Akt and AMPK signaling in muscle and white adipose tissue (WAT) as well as increased FoxO1 phosphorylation and expression of mature Srebp-1c in liver and downregulation of the hepatic gluconeogenic genes, Pepck and G6pase. KO-DIO mice also exhibited downregulation of cytokines and proinflammatory genes and upregulation of anti-inflammatory genes in WAT, and peritoneal macrophages from KO mice displayed similarly reduced proinflammatory gene expression. The insulin-sensitive, anti-inflammatory phenotype of KO-DIO mice is masked by supplementing PST. Conversely, a PST variant peptide PSTv1 (PST-NΔ3: CHGA(276-301)), lacking PST activity, simulated the KO phenotype by sensitizing WT-DIO mice to insulin. In summary, the reduced inflammation due to PST deficiency prevented the development of insulin resistance in KO-DIO mice. Thus, obesity manifests insulin resistance only in the presence of PST, and in its absence obesity is dissociated from insulin resistance.


Subject(s)
Chromogranin A/immunology , Obesity/immunology , Obesity/metabolism , Pancreatic Hormones/pharmacology , Panniculitis/immunology , Signal Transduction/immunology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Cells, Cultured , Chemotaxis/immunology , Chromogranin A/genetics , Chromogranin A/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Glucose Intolerance/drug therapy , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Insulin Resistance/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Pancreatic Hormones/immunology , Pancreatic Hormones/metabolism , Panniculitis/drug therapy , Panniculitis/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/immunology , Sterol Regulatory Element Binding Protein 1/metabolism
12.
Psychoneuroendocrinology ; 51: 459-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25456346

ABSTRACT

BACKGROUND: Research on the etiology of post-traumatic stress disorder (PTSD) has rapidly matured, moving from candidate gene studies to interrogation of the entire human genome in genome-wide association studies (GWAS). Here we present the results of a GWAS performed on samples from combat-exposed U.S. Marines and Sailors from the Marine Resiliency Study (MRS) scheduled for deployment to Iraq and/or Afghanistan. The MRS is a large, prospective study with longitudinal follow-up designed to identify risk and resiliency factors for combat-induced stress-related symptoms. Previously implicated PTSD risk loci from the literature and polygenic risk scores across psychiatric disorders were also evaluated in the MRS cohort. METHODS: Participants (N=3494) were assessed using the Clinician-Administered PTSD Scale and diagnosed using the DSM-IV diagnostic criterion. Subjects with partial and/or full PTSD diagnosis were called cases, all other subjects were designated controls, and study-wide maximum CAPS scores were used for longitudinal assessments. Genomic DNA was genotyped on the Illumina HumanOmniExpressExome array. Individual genetic ancestry was determined by supervised cluster analysis for subjects of European, African, Hispanic/Native American, and other descent. To test for association of SNPs with PTSD, logistic regressions were performed within each ancestry group and results were combined in meta-analyses. Measures of childhood and adult trauma were included to test for gene-by-environment (GxE) interactions. Polygenic risk scores from the Psychiatric Genomic Consortium were used for major depressive disorder (MDD), bipolar disorder (BPD), and schizophrenia (SCZ). RESULTS: The array produced >800K directly genotyped and >21M imputed markers in 3494 unrelated, trauma-exposed males, of which 940 were diagnosed with partial or full PTSD. The GWAS meta-analysis identified the phosphoribosyl transferase domain containing 1 gene (PRTFDC1) as a genome-wide significant PTSD locus (rs6482463; OR=1.47, SE=0.06, p=2.04×10(-9)), with a similar effect across ancestry groups. Association of PRTFDC1 with PTSD in an independent military cohort showed some evidence for replication. Loci with suggestive evidence of association (n=25 genes, p<5×10(-6)) further implicated genes related to immune response and the ubiquitin system, but these findings remain to be replicated in larger GWASs. A replication analysis of 25 putative PTSD genes from the literature found nominally significant SNPs for the majority of these genes, but associations did not remain significant after correction for multiple comparison. A cross-disorder analysis of polygenic risk scores from GWASs of BPD, MDD, and SCZ found that PTSD diagnosis was associated with risk sores of BPD, but not with MDD or SCZ. CONCLUSIONS: This first multi-ethnic/racial GWAS of PTSD highlights the potential to increase power through meta-analyses across ancestry groups. We found evidence for PRTFDC1 as a potential novel PTSD gene, a finding that awaits further replication. Our findings indicate that the genetic architecture of PTSD may be determined by many SNPs with small effects, and overlap with other neuropsychiatric disorders, consistent with current findings from large GWAS of other psychiatric disorders.


Subject(s)
Combat Disorders/genetics , Genetic Predisposition to Disease , Hypoxanthine Phosphoribosyltransferase/genetics , Resilience, Psychological , Stress Disorders, Post-Traumatic/genetics , Adolescent , Adult , Combat Disorders/psychology , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Military Personnel/psychology , Polymorphism, Single Nucleotide , Stress Disorders, Post-Traumatic/psychology , Young Adult
13.
J Am Soc Nephrol ; 26(8): 1816-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25392232

ABSTRACT

Chromogranin A (CHGA) is coreleased with catecholamines from secretory vesicles in adrenal medulla and sympathetic axons. Genetic variation in the CHGA 3'-region has been associated with autonomic control of circulation, hypertension, and hypertensive nephropathy, and the CHGA 3'-untranslated region (3'-UTR) variant C+87T (rs7610) displayed peak associations with these traits in humans. Here, we explored the molecular mechanisms underlying these associations. C+87T occurred in a microRNA-107 (miR-107) motif (match: T>C), and CHGA mRNA expression varied inversely with miR-107 abundance. In cells transfected with chimeric luciferase/CHGA 3'-UTR reporters encoding either the T allele or the C allele, changes in miR-107 expression levels had much greater effects on expression of the T allele. Cotransfection experiments with hsa-miR-107 oligonucleotides and eukaryotic CHGA plasmids produced similar results. Notably, an in vitro CHGA transcription/translation experiment revealed that changes in hsa-miR-107 expression altered expression of the T allele variant only. Mice with targeted ablation of Chga exhibited greater eGFR. Using BAC transgenesis, we created a mouse model with a humanized CHGA locus (T/T genotype at C+87T), in which treatment with a hsa-miR-107 inhibitor yielded prolonged falls in SBP/DBP compared with wild-type mice. We conclude that the CHGA 3'-UTR C+87T disrupts an miR-107 motif, with differential effects on CHGA expression, and that a cis:trans (mRNA:miR) interaction regulates the association of CHGA with BP and hypertensive nephropathy. These results indicate new strategies for probing autonomic circulatory control and ultimately, susceptibility to hypertensive renal sequelae.


Subject(s)
Chromogranin A/genetics , Hypertension, Renal/genetics , MicroRNAs/genetics , 3' Untranslated Regions , Alleles , Animals , Blood Pressure , Chromogranin A/metabolism , Glomerular Filtration Rate , HEK293 Cells , Humans , Hypertension, Renal/metabolism , Luciferases , Male , Mice , Mice, Transgenic , PC12 Cells , Polymorphism, Genetic , Rats
14.
Endocrinology ; 155(10): 3793-805, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051446

ABSTRACT

Cigarette smoking causes insulin resistance. However, nicotine induces anti-inflammation and improves glucose tolerance in insulin-resistant animal models. Here, we determined the effects of nicotine on glucose metabolism in insulin-sensitive C57BL/J6 mice. Acute nicotine administration (30 min) caused fasting hyperglycemia and lowered insulin sensitivity acutely, which depended on the activation of nicotinic-acetylcholine receptors (nAChRs) and correlated with increased catecholamine secretion, nitric oxide (NO) production, and glycogenolysis. Chlorisondamine, an inhibitor of nAChRs, reduced acute nicotine-induced hyperglycemia. qRT-PCR analysis revealed that the liver and muscle express predominantly ß4 > α10 > α3 > α7 and ß4 > α10 > ß1 > α1 mRNA for nAChR subunits respectively, whereas the adrenal gland expresses ß4 > α3 > α7 > α10 mRNA. Chronic nicotine treatment significantly suppressed expression of α3-nAChR (predominant peripheral α-subunit) in liver. Whereas acute nicotine treatment raised plasma norepinephrine (NE) and epinephrine (Epi) levels, chronic nicotine exposure raised only Epi. Acute nicotine treatment raised both basal and glucose-stimulated insulin secretion (GSIS). After chronic nicotine treatment, basal insulin level was elevated, but GSIS after acute saline or nicotine treatment was blunted. Chronic nicotine exposure caused an increased buildup of NO in plasma and liver, leading to decreased glycogen storage, along with a concomitant suppression of Pepck and G6Pase mRNA, thus preventing hyperglycemia. The insulin-sensitizing effect of chronic nicotine was independent of weight loss. Chronic nicotine treatment enhanced PI-3-kinase activities and increased Akt and glycogen synthase kinase (GSK)-3ß phosphorylation in an nAChR-dependent manner coupled with decreased cAMP response element-binding protein (CREB) phosphorylation. The latter effects caused suppression of Pepck and G6Pase gene expression. Thus, nicotine causes both insulin resistance and insulin sensitivity depending on the duration of the treatment.


Subject(s)
Blood Glucose/metabolism , Hyperglycemia/chemically induced , Insulin Resistance , Nicotine/pharmacology , Receptors, Nicotinic/physiology , Animals , Cells, Cultured , Epinephrine/blood , Homeostasis/drug effects , Homeostasis/genetics , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Norepinephrine/blood , Time Factors
15.
Hum Mol Genet ; 23(23): 6375-84, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24986918

ABSTRACT

Dopamine beta-hydroxylase (DBH) is the biosynthetic enzyme catalyzing formation of norepinephrine. Changes in DBH expression or activity have been implicated in the pathogenesis of cardiovascular and neuropsychiatric disorders. Genetic determination of DBH enzymatic activity and its secretion are only incompletely understood. We began with a genome-wide association search for loci contributing to DBH activity in human plasma. Initially, in a population sample of European ancestry, we identified the proximal DBH promoter as a region harboring three common trait-determining variants (top hit rs1611115, P = 7.2 × 10(-51)). We confirmed their effects on transcription and showed that the three variants each acted additively on gene expression. Results were replicated in a population sample of Native American descent (top hit rs1611115, P = 4.1 × 10(-15)). Jointly, DBH variants accounted for 57% of DBH trait variation. We further identified a genome-wide significant SNP at the LOC338797 locus on chromosome 12 as trans-quantitative trait locus (QTL) (rs4255618, P = 4.62 × 10(-8)). Conditional analyses on DBH identified a third genomic region contributing to DBH variation: a likely cis-QTL adjacent to DBH in SARDH (rs7040170, P = 1.31 × 10(-14)) on chromosome 9q. We conclude that three common SNPs in the DBH promoter act additively to control phenotypic variation in DBH levels, and that two additional novel loci (SARDH and LOC338797) may also contribute to the expression of this catecholamine biosynthetic trait. Identification of DBH variants with strong effects makes it possible to take advantage of Mendelian randomization approaches to test causal effects of this intermediate trait on disease.


Subject(s)
Catecholamines/biosynthesis , Dopamine beta-Hydroxylase/genetics , Protein Isoforms/genetics , American Indian or Alaska Native , Dopamine beta-Hydroxylase/blood , Genome-Wide Association Study , Humans , Male , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protein Isoforms/blood , White People
16.
Psychosom Med ; 76(4): 292-301, 2014 May.
Article in English | MEDLINE | ID: mdl-24804881

ABSTRACT

OBJECTIVE: Heart rate variability (HRV), thought to reflect autonomic nervous system function, is lowered under conditions such as posttraumatic stress disorder (PTSD). The potential confounding effects of traumatic brain injury (TBI) and depression in the relationship between HRV and PTSD have not been elucidated in a large cohort of military service members. Here we describe HRV associations with stress disorder symptoms in a large study of Marines while accounting for well-known covariates of HRV and PTSD including TBI and depression. METHODS: Four battalions of male active-duty Marines (n = 2430) were assessed 1 to 2 months before a combat deployment. HRV was measured during a 5-minute rest. Depression and PTSD were assessed using the Beck Depression Inventory and Clinician-Administered PTSD Scale, respectively. RESULTS: When adjusting for covariates, including TBI, regression analyses showed that lower levels of high-frequency HRV were associated with a diagnosis of PTSD (ß = -0.20, p = .035). Depression and PTSD severity were correlated (r = 0.49, p < .001); however, participants with PTSD but relatively low depression scores exhibited reduced high frequency compared with controls (p = .012). Marines with deployment experience (n = 1254) had lower HRV than did those with no experience (p = .033). CONCLUSIONS: This cross-sectional analysis of a large cohort supports associations between PTSD and reduced HRV when accounting for TBI and depression symptoms. Future postdeployment assessments will be used to determine whether predeployment HRV can predict vulnerability and resilience to the serious psychological and physiological consequences of combat exposure.


Subject(s)
Brain Injuries/epidemiology , Depressive Disorder/epidemiology , Heart Rate/physiology , Military Personnel/statistics & numerical data , Stress Disorders, Post-Traumatic/epidemiology , Analysis of Variance , Autonomic Nervous System/physiopathology , Brain Injuries/physiopathology , Caffeine/administration & dosage , Confounding Factors, Epidemiologic , Cross-Sectional Studies , Depressive Disorder/physiopathology , Humans , Male , Military Personnel/psychology , Nicotine/administration & dosage , Photoplethysmography/methods , Psychiatric Status Rating Scales , Regression Analysis , Severity of Illness Index , Stress Disorders, Post-Traumatic/physiopathology , Young Adult
17.
JAMA Psychiatry ; 71(4): 423-31, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24576974

ABSTRACT

IMPORTANCE: Posttraumatic stress disorder (PTSD) has been associated in cross-sectional studies with peripheral inflammation. It is not known whether this observed association is the result of PTSD predisposing to inflammation (as sometimes postulated) or to inflammation predisposing to PTSD. OBJECTIVE: To determine whether plasma concentration of the inflammatory marker C-reactive protein (CRP) helps predict PTSD symptoms. DESIGN, SETTING, AND PARTICIPANTS: The Marine Resiliency Study, a prospective study of approximately 2600 war zone-deployed Marines, evaluated PTSD symptoms and various physiological and psychological parameters before deployment and at approximately 3 and 6 months following a 7-month deployment. Participants were recruited from 4 all-male infantry battalions imminently deploying to a war zone. Participation was requested of 2978 individuals; 2610 people (87.6%) consented and 2555 (85.8%) were included in the present analysis. Postdeployment data on combat-related trauma were included for 2208 participants (86.4% of the 2555 included) and on PTSD symptoms at 3 and 6 months after deployment for 1861 (72.8%) and 1617 (63.3%) participants, respectively. MAIN OUTCOMES AND MEASURES: Severity of PTSD symptoms 3 months after deployment assessed by the Clinician-Administered PTSD Scale (CAPS). RESULTS: We determined the effects of baseline plasma CRP concentration on postdeployment CAPS using zero-inflated negative binomial regression (ZINBR), a procedure designed for distributions, such as CAPS in this study, that have an excess of zeroes in addition to being positively skewed. Adjusting for the baseline CAPS score, trauma exposure, and other relevant covariates, we found baseline plasma CRP concentration to be a highly significant overall predictor of postdeployment CAPS scores (P = .002): each 10-fold increment in CRP concentration was associated with an odds ratio of nonzero outcome (presence vs absence of any PTSD symptoms) of 1.51 (95% CI, 1.15-1.97; P = .003) and a fold increase in outcome with a nonzero value (extent of symptoms when present) of 1.06 (95% CI, 0.99-1.14; P = .09). CONCLUSIONS: AND RELEVANCE A marker of peripheral inflammation, plasma CRP may be prospectively associated with PTSD symptom emergence, suggesting that inflammation may predispose to PTSD.


Subject(s)
C-Reactive Protein/metabolism , Combat Disorders/blood , Combat Disorders/diagnosis , Military Personnel/psychology , Stress Disorders, Post-Traumatic/blood , Stress Disorders, Post-Traumatic/diagnosis , Adult , Biomarkers/blood , Combat Disorders/psychology , Humans , Inflammation/blood , Inflammation/diagnosis , Inflammation/psychology , Longitudinal Studies , Male , Neuropsychological Tests , Predictive Value of Tests , Prospective Studies , Resilience, Psychological , Risk Assessment , Stress Disorders, Post-Traumatic/psychology
18.
Am J Hypertens ; 27(6): 828-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24532572

ABSTRACT

BACKGROUND: Elevated blood pressure (BP), a heritable risk factor for many age-related disorders, is commonly investigated in population and genetic studies, but antihypertensive use can confound study results. Routine methods to adjust for antihypertensives may not sufficiently account for newer treatment protocols (i.e., combination or multiple drug therapy) found in contemporary cohorts. METHODS: We refined an existing method to impute unmedicated BP in individuals on antihypertensives by incorporating new treatment trends. We assessed BP and antihypertensive use in male twins (n = 1,237) from the Vietnam Era Twin Study of Aging: 36% reported antihypertensive use; 52% of those treated were on multiple drugs. RESULTS: Estimated heritability was 0.43 (95% confidence interval (CI) = 0.20-0.50) and 0.44 (95% CI = 0.22-0.61) for measured systolic BP (SBP) and diastolic BP (DBP), respectively. We imputed BP for antihypertensives by 3 approaches: (i) addition of a fixed value of 10/5mm Hg to measured SBP/DBP; (ii) incremented addition of mm Hg to BP based on number of medications; and (iii) a refined approach adding mm Hg based on antihypertensive drug class and ethnicity. The imputations did not significantly affect estimated heritability of BP. However, use of our most refined imputation method and other methods resulted in significantly increased phenotypic correlations between BP and body mass index, a trait known to be correlated with BP. CONCLUSIONS: This study highlights the potential usefulness of applying a representative adjustment for medication use, such as by considering drug class, ethnicity, and the combination of drugs when assessing the relationship between BP and risk factors.


Subject(s)
Algorithms , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Diseases in Twins/drug therapy , Hypertension/drug therapy , Analysis of Variance , Asian People/genetics , Blood Pressure/genetics , Body Mass Index , Confounding Factors, Epidemiologic , Diseases in Twins/diagnosis , Diseases in Twins/ethnology , Diseases in Twins/genetics , Drug Therapy, Combination , Genetic Association Studies , Genetic Predisposition to Disease , Heredity , Humans , Hypertension/diagnosis , Hypertension/ethnology , Hypertension/genetics , Linear Models , Male , Middle Aged , Models, Statistical , Nonlinear Dynamics , Phenotype , Population Surveillance , Registries , Risk Assessment , Risk Factors , Treatment Outcome , Vietnam/epidemiology
19.
J Am Coll Cardiol ; 63(15): 1542-55, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24509276

ABSTRACT

OBJECTIVES: This study coupled 2 strategies-trait extremes and genome-wide pooling-to discover a novel blood pressure (BP) locus that encodes a previously uncharacterized thiamine transporter. BACKGROUND: Hypertension is a heritable trait that remains the most potent and widespread cardiovascular risk factor, although details of its genetic determination are poorly understood. METHODS: Representative genomic deoxyribonucleic acid (DNA) pools were created from male and female subjects in the highest- and lowest-fifth percentiles of BP in a primary care population of >50,000 patients. The peak associated single-nucleotide polymorphisms were typed in individual DNA samples, as well as in twins/siblings phenotyped for cardiovascular and autonomic traits. Biochemical properties of the associated transporter were evaluated in cellular assays. RESULTS: After chip hybridization and calculation of relative allele scores, the peak associations were typed in individual samples, revealing an association between hypertension, systolic BP, and diastolic BP and the previously uncharacterized solute carrier SLC35F3. The BP genetic association at SLC35F3 was validated by meta-analysis in an independent sample from the original source population, as well as the International Consortium for Blood Pressure Genome-Wide Association Studies (across North America and western Europe). Sequence homology to a putative yeast thiamine (vitamin B1) transporter prompted us to express human SLC35F3 in Escherichia coli, which catalyzed [(3)H]-thiamine uptake. SLC35F3 risk-allele homozygotes (T/T) displayed decreased erythrocyte thiamine content on microbiological assay. In twin pairs, the SLC35F3 risk allele predicted heritable cardiovascular traits previously associated with thiamine deficiency, including elevated cardiac stroke volume with decreased vascular resistance, and elevated pressor responses to environmental (cold) stress. Allelic expression imbalance confirmed that cis variation at the human SLC35F3 locus influenced expression of that gene, and the allelic expression imbalance peak coincided with the hypertension peak. CONCLUSIONS: Novel strategies were coupled to position a new hypertension-susceptibility locus, uncovering a previously unsuspected thiamine transporter whose genetic variants predicted several disturbances in cardiac and autonomic function. The results have implications for the pathogenesis and treatment of systemic hypertension.


Subject(s)
DNA/genetics , Genetic Predisposition to Disease , Hypertension/genetics , Membrane Transport Proteins/genetics , Polymorphism, Genetic , Adult , Alleles , Blood Pressure , Female , Genome-Wide Association Study , Genotype , Humans , Hypertension/metabolism , Hypertension/physiopathology , Male , Membrane Transport Proteins/metabolism , Phenotype , Thiamine/genetics , Thiamine/metabolism
20.
PLoS One ; 9(1): e84132, 2014.
Article in English | MEDLINE | ID: mdl-24465394

ABSTRACT

RATIONALE: The chromogranin A-derived peptide pancreastatin (PST) is a dysglycemic, counter-regulatory peptide for insulin action, especially in liver. Although previous evidence for a PST binding protein has been reported, such a receptor has not been identified or sequenced. METHODS AND RESULTS: We used ligand affinity to purify the PST target, with biotinylated human PST (hCHGA273-301-amide) as "bait" and mouse liver homogenate as "prey", and identified GRP78 (a.k.a. "78 kDa Glucose Regulated Protein", HSPA5, BIP) as a major interacting partner of PST. GRP78 belongs to the family of heat shock proteins (chaperones), involved in several cellular processes including protein folding and glucose metabolism. We analyzed expression of GRP78 in the absence of PST in a mouse knockout model lacking its precursor CHGA: hepatic transcriptome data revealed global over-expression of not only GRP78 but also other heat shock transcripts (of the "adaptive UPR") in CHGA(-/-) mice compared to wild-type (+/+). By contrast, we found a global decline in expression of hepatic pro-apoptotic transcripts in CHGA(-/-) mice. GRP78's ATPase enzymatic activity was dose-dependently inhibited by PST (IC50∼5.2 µM). PST also inhibited the up-regulation of GRP78 expression during UPR activation (by tunicamycin) in hepatocytes. PST inhibited insulin-stimulated glucose uptake in adipocytes, and increased hepatic expression of G6Pase (the final step in gluconeogenesis/glycogenolysis). In hepatocytes not only PST but also other GRP78-ATPase inhibitors (VER-155008 or ADP) increased G6Pase expression. GRP78 over-expression inhibited G6Pase expression in hepatocytes, with partial restoration by GRP78-ATPase inhibitors PST, VER-155008, or ADP. CONCLUSIONS: Our results indicate that an unexpected major hepatic target of PST is the adaptive UPR chaperone GRP78. PST not only binds to GRP78 (in pH-dependent fashion), but also inhibits GRP78's ATPase enzymatic activity, and impairs its biosynthetic response to UPR activation. PST decreases insulin-stimulated cellular glucose uptake, and PST as well as other chaperone ATPase activity inhibitors augment expression of G6Pase; GRP78 over-expression antagonizes this PST action. Analysis of the novel PST/GRP78 interaction may provide a new avenue of investigation into cellular glycemic control as well as dysglycemia.


Subject(s)
Chromogranin A/chemistry , Chromogranin A/metabolism , Heat-Shock Proteins/metabolism , Liver/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adrenal Glands/metabolism , Animals , Chromogranin A/genetics , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Mice , Protein Binding , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...