Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 616
Filter
1.
Mol Biol Rep ; 51(1): 661, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758505

ABSTRACT

SCN5A mutations have been reported to cause various cardiomyopathies in humans. Most of the SCN5A mutations causes loss of function and thereby, alters the overall cellular function. Therefore, to understand the loss of SCN5A function in cardiomyocytes, we have knocked down the SCN5A gene (SCN5A-KD) in H9c2 cells and explored the cell phenotype and molecular behaviors in the presence and absence of isoproterenol (ISO), an adrenergic receptor agonist that induces cardiac hypertrophy. Expression of several genes related to hypertrophy, inflammation, fibrosis, and energy metabolism pathways were evaluated. It was found that the mRNA expression of hypertrophy-related gene, brain (B-type) natriuretic peptide (BNP) was significantly increased in SCN5A-KD cells as compared to 'control' H9c2 cells. There was a further increase in the mRNA expressions of BNP and ßMHC in SCN5A-KD cells after ISO treatment compared to their respective controls. Pro-inflammatory cytokine, tumor necrosis factor-alpha expression was significantly increased in 'SCN5A-KD' H9c2 cells. Further, metabolism-related genes like glucose transporter type 4, cluster of differentiation 36, peroxisome proliferator-activated receptor alpha, and peroxisome proliferator-activated receptor-gamma were significantly elevated in the SCN5A-KD cells as compared to the control cells. Upregulation of these metabolic genes is associated with increased ATP production. The study revealed that SCN5A knock-down causes alteration of gene expression related to cardiac hypertrophy, inflammation, and energy metabolism pathways, which may promote cardiac remodelling and cardiomyopathy.


Subject(s)
Cardiomegaly , Isoproterenol , NAV1.5 Voltage-Gated Sodium Channel , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Rats , Cell Line , Isoproterenol/pharmacology , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Animals , Gene Knockdown Techniques , Humans , Myoblasts, Cardiac/metabolism , Energy Metabolism/genetics , Gene Expression Regulation/genetics
2.
Physiol Genomics ; 56(6): 436-444, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38586874

ABSTRACT

This study aimed to investigate the relationship between pre- and postexercise cardiac biomarker release according to athletic status (trained vs. untrained) and to establish whether the I/D polymorphism in the angiotensin-converting enzyme (ACE) gene had an influence on cardiac biomarkers release with specific regard on the influence of the training state. We determined cardiac troponin I (cTnI) and N-terminal pro-brain natriuretic peptide (NT-proBNP) in 29 trained and 27 untrained male soccer players before and after moderate-intensity continuous exercise (MICE) and high-intensity interval exercise (HIIE) running tests. Trained soccer players had higher pre (trained: 0.014 ± 0.007 ng/mL; untrained: 0.010 ± 0.005 ng/mL) and post HIIE (trained: 0.031 ± 0.008 ng/mL; untrained: 0.0179 ± 0.007) and MICE (trained: 0.030 ± 0.007 ng/mL; untrained: 0.018 ± 0.007) cTnI values than untrained subjects, but the change with exercise (ΔcTnI) was similar between groups. There was no significant difference in baseline and postexercise NT-proBNP between groups. NT-proBNP levels were elevated after both HIIE and MICE. Considering three ACE genotypes, the mean pre exercise cTnI values of the trained group (DD: 0.015 ± 0.008 ng/mL, ID: 0.015 ± 0.007 ng/mL, and II: 0.014 ± 0.008 ng/mL) and their untrained counterparts (DD: 0.010 ± 0.004 ng/mL, ID: 0.011 ± 0.004 ng/mL, and II: 0.010 ± 0.006 ng/mL) did not show any significant difference. To sum up, noticeable difference in baseline cTnI was observed, which was related to athletic status but not ACE genotypes. Neither athletic status nor ACE genotypes seemed to affect the changes in cardiac biomarkers in response to HIIE and MICE, indicating that the ACE gene does not play a significant role in the release of exercise-induced cardiac biomarkers indicative of cardiac damage in Iranian soccer players.NEW & NOTEWORTHY Our study investigated the impact of athletic status and angiotensin-converting enzyme (ACE) gene I/D polymorphism on cardiac biomarkers in soccer players. Trained players showed higher baseline cardiac troponin I (cTnI) levels, whereas postexercise ΔcTnI remained consistent across groups. N-terminal pro-brain natriuretic peptide increased after exercise in both groups, staying within normal limits. ACE genotypes did not significantly affect pre-exercise cTnI. Overall, athletic status influences baseline cTnI, but neither it nor ACE genotypes significantly impact exercise-induced cardiac biomarker responses in this population.


Subject(s)
Biomarkers , Exercise , Natriuretic Peptide, Brain , Peptide Fragments , Peptidyl-Dipeptidase A , Polymorphism, Genetic , Troponin I , Male , Humans , Peptidyl-Dipeptidase A/genetics , Biomarkers/blood , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Troponin I/blood , Troponin I/genetics , Peptide Fragments/blood , Exercise/physiology , Young Adult , Adult , High-Intensity Interval Training/methods , Soccer/physiology , INDEL Mutation/genetics , Heart/physiology
3.
Cell Mol Biol (Noisy-le-grand) ; 69(13): 180-188, 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38158669

ABSTRACT

The research explored the link between Brain Natriuretic Peptides (BNP) gene promoter T-381C polymorphism, serum BNP, and lipid profiles in Kurdish people from Iraq with stable coronary artery disease (CAD). The study was conducted on 62 individuals with CAD and 31 without CAD (control group). DNA was extracted from each individual's sample using the Sanger sequencing method to study the BNP gene's polymorphism. The identified alleles were TT, TC, and CC. The frequency of the TT genotype decreased significantly among the patient group compared to the control group, while the CC genotype's frequency was higher (p<0.05). However, there was no significant increase in BNP levels in TC and CC genotypes compared to the TT genotype. Lipid profile values were not significantly different among the genotypes. The study utilized a cut-off value for BNP activity for predicting CAD and found that individuals with a BNP activity value less than the cut-off had significantly greater changes in lipid profile and renal function (p<0.05). Stepwise multivariate regression analysis showed that cholesterol was not the only primary determinant of BNP rate in subjects with stable CAD; oxidized low-density lipoprotein (Ox-LDL), a history of heart attacks, and oxidative stress malondialdehyde (MDA) had a significant effect. Homozygous C allele carriers at position 381 of the BNP precursors gene promoter were more likely to exhibit atherosclerosis lesions. We found that BNP rs198389 was not correlated with lipid profile and kidney disease.


Subject(s)
Coronary Artery Disease , Lipids , Natriuretic Peptide, Brain , Humans , Coronary Artery Disease/genetics , Genotype , Natriuretic Peptide, Brain/genetics , Polymorphism, Genetic , Lipids/blood
4.
N Engl J Med ; 389(18): 1685-1692, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37913506

ABSTRACT

Two siblings presented with cardiomyopathy, hypertension, arrhythmia, and fibrosis of the left atrium. Each had a homozygous null variant in CORIN, the gene encoding atrial natriuretic peptide (ANP)-converting enzyme. A plasma sample obtained from one of the siblings had no detectable levels of corin or N-terminal pro-ANP but had elevated levels of B-type natriuretic peptide (BNP) and one of the two protein markers of fibrosis that we tested. These and other findings support the hypothesis that BNP cannot fully compensate for a lack of activation of the ANP pathway and that corin is critical to normal ANP activity, left atrial function, and cardiovascular homeostasis.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathies , Heart Atria , Hypertension , Humans , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Atrial Fibrillation , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomyopathies/blood , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Fibrosis , Heart Atria/diagnostic imaging , Heart Atria/metabolism , Heart Atria/pathology , Hypertension/blood , Hypertension/genetics , Hypertension/metabolism , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Serine Endopeptidases/blood , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Siblings
5.
Gene ; 878: 147580, 2023 Aug 20.
Article in English | MEDLINE | ID: mdl-37339722

ABSTRACT

To investigate the correlation between NPPB gene variants and pulse pressure hypertension and the underlying regulatory mechanisms and try to confirm that NPPB may be a potential molecular target of gene therapy for pulse pressure hypertension. A total of 898 participants were recruited from the First Affiliated Hospital of Fujian Medical University and the plasmids with differential expression of NPPB were constructed. Genotype distribution of NPPB(rs3753581, rs198388, and rs198389)was analyzed and the expression of N-terminal pro-B-type natriuretic peptide(NT-proBNP) and renin-angiotensin -aldosterone system(RAAS) related indicators were identified in the groups studied. According to a genotype analysis, there was a significant difference in the genotype distribution of NPPB rs3753581 among the groups (P = 0.034). In logistic regression analysis, NPPB rs3753581 TT was associated with a 1.8-fold greater risk of pulse pressure hypertension than NPPB rs3753581 GG (odds ratio = 1.801; 95% confidence interval: 1.070-3.032; P = 0.027). The expression of NT-proBNP and RAAS related indicators in clinical and laboratory samples showed striking differences. The activity of firefly and Renilla luciferase in pGL-3-NPPB-luc (-1299G) was higher than pGL-3-NPPBmut-luc(-1299 T)(P < 0.05). The binding of NPPB gene promoter rs3753581 (-1299G) with transcription factors IRF1, PRDM1, and ZNF263 was predicted and validated by the bioinformatics software TESS and chromatin immunoprecipitation(P < 0.05). NPPB rs3753581 was correlated with genetic susceptibility to pulse pressure hypertension and the transcription factors IRF1, PRDM1, and ZNF263 may be involved in the regulation of NPPB rs3753581 promoter (-1299G) on the expression of NT-proBNP/RAAS.


Subject(s)
Hypertension , Transcription Factors , Humans , Blood Pressure/genetics , Transcription Factors/genetics , Hypertension/genetics , Natriuretic Peptide, Brain/genetics , Genotype , Peptide Fragments/genetics , DNA-Binding Proteins/genetics , Interferon Regulatory Factor-1/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics
6.
Redox Biol ; 62: 102702, 2023 06.
Article in English | MEDLINE | ID: mdl-37116257

ABSTRACT

Brain natriuretic peptide (BNP) belongs to the family of natriuretic peptides, which are responsible for a wide range of actions. Diabetic cardiomyopathy (DCM) is often associated with increased BNP levels. This present research intends to explore the role of BNP in the development of DCM and the underlying mechanisms. Diabetes was induced in mice using streptozotocin (STZ). Primary neonatal cardiomyocytes were treated with high glucose. It was found that the levels of plasma BNP started to increase at 8 weeks after diabetes, which preceded the development of DCM. Addition of exogenous BNP promoted Opa1-mediated mitochondrial fusion, inhibited mitochondrial oxidative stress, preserved mitochondrial respiratory capacity and prevented the development of DCM, while knockdown of endogenous BNP exacerbated mitochondrial dysfunction and accelerated DCM. Opa1 knockdown attenuated the aforementioned protective action of BNP both in vivo and in vitro. BNP-induced mitochondrial fusion requires the activation of STAT3, which facilitated Opa1 transcription by binding to its promoter regions. PKG, a crucial signaling biomolecule in the BNP signaling pathway, interacted with STAT3 and induced its activation. Knockdown of NPRA (the receptor of BNP) or PKG blunted the promoting effect of BNP on STAT3 phosphorylation and Opa1-mediated mitochondrial fusion. The results of this study demonstrate for the first time that there is a rise in BNP during the early stages of DCM as a compensatory protection mechanism. BNP is a novel mitochondrial fusion activator in protecting against hyperglycemia-induced mitochondrial oxidative injury and DCM through the activation of NPRA-PKG-STAT3-Opa1 signaling pathway.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Animals , Mice , Diabetes Mellitus/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Mitochondrial Dynamics , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Signal Transduction , Cyclic GMP-Dependent Protein Kinases
7.
Circ Res ; 132(5): 586-600, 2023 03 03.
Article in English | MEDLINE | ID: mdl-36756875

ABSTRACT

BACKGROUND: Myocardial infarction (MI) elicits cardiac fibroblast activation and extracellular matrix (ECM) deposition to maintain the structural integrity of the heart. Recent studies demonstrate that Fap (fibroblast activation protein)-a prolyl-specific serine protease-is an important marker of activated cardiac fibroblasts after MI. METHODS: Left ventricle and plasma samples from patients and healthy donors were used to analyze the expression level of FAP and its prognostic value. Echocardiography and histological analysis of heart sections were used to analyze cardiac functions, scar formation, ECM deposition and angiogenesis after MI. RNA-Sequencing, biochemical analysis, cardiac fibroblasts (CFs) and endothelial cells co-culture were used to reveal the molecular and cellular mechanisms by which Fap regulates angiogenesis. RESULTS: We found that Fap is upregulated in patient cardiac fibroblasts after cardiac injuries, while plasma Fap is downregulated and functions as a prognostic marker for cardiac repair. Genetic or pharmacological inhibition of Fap in mice significantly improved cardiac function after MI. Histological and transcriptomic analyses showed that Fap inhibition leads to increased angiogenesis in the peri-infarct zone, which promotes ECM deposition and alignment by cardiac fibroblasts and prevents their overactivation, thereby limiting scar expansion. Mechanistically, we found that BNP (brain natriuretic peptide) is a novel substrate of Fap that mediates postischemic angiogenesis. Fap degrades BNP to inhibit vascular endothelial cell migration and tube formation. Pharmacological inhibition of Fap in Nppb (encoding pre-proBNP) or Npr1 (encoding the BNP receptor)-deficient mice showed no cardioprotective effects, suggesting that BNP is a physiological substrate of Fap. CONCLUSIONS: This study identifies Fap as a negative regulator of cardiac repair and a potential drug target to treat MI. Inhibition of Fap stabilizes BNP to promote angiogenesis and cardiac repair.


Subject(s)
Myocardial Infarction , Natriuretic Peptide, Brain , Animals , Mice , Cicatrix , Endopeptidases/genetics , Endothelial Cells/pathology , Myocardial Infarction/pathology , Natriuretic Peptide, Brain/genetics
8.
J Physiol Pharmacol ; 74(6)2023 Dec.
Article in English | MEDLINE | ID: mdl-38345442

ABSTRACT

Dyxin is a LIM-domain containing transcriptional regulator protein shown to play a role in a hypertrophic response in the heart. Here, the effect of adenoviral dyxin overexpression was studied on cardiac function and gene expression in the normal heart and in angiotensin II (Ang II)-induced hypertension in rats. The adenovirus-mediated intramyocardial gene transfer of dyxin (1.5x109 infectious units/animal) was performed into the left ventricle (LV) of Sprague-Dawley rats with and without the Ang II (33 µg/kg/h) infusion, administered via osmotic minipumps for 1 and 2 weeks. Echocardiography was used to assess the structural and functional changes. Dyxin expression and localization in the heart was analyzed with quantitative RT-PCR and immunohistochemistry, respectively. In the normal rat heart, the adenoviral overexpression of dyxin did not alter LV function in normal hearts as assessed by echocardiography. Dyxin was found to be localized in the cardiomyocytes as shown by the immunohistochemical staining. In Ang II-induced hypertrophy, echocardiographic data revealed a significant increase in the posterior wall diameter both in systole (21%, P<0.05) and diastole (21%, P<0.01) as well as in the diameter of the interventricular septum in systole (19%, P<0.05) in the dyxin-injected group compared with the LacZ-injected animals after two weeks of Ang II infusion. Interestingly, a significant decrease in the levels of both atrial natriuretic peptide (ANP) mRNA (55%, P<0.01) and B-type natriuretic peptide (BNP) mRNA (68%, P<0.05) was observed in the dyxin-injected group compared with the LacZ control group after one week of Ang II infusion. These results indicate that dyxin overexpression was deteriorative against pressure overload by inducing structural changes in the LV in rats. Interestingly, simultaneous adenoviral overexpression of dyxin suppressed the Ang II-induced changes of ANP and BNP genes suggesting that dyxin might have a role as a regulator of the cardiac hypertrophic gene program.


Subject(s)
Angiotensin II , Hypertension , Rats , Animals , Angiotensin II/pharmacology , Angiotensin II/metabolism , Rats, Sprague-Dawley , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Myocytes, Cardiac , Natriuretic Peptide, Brain/genetics , RNA, Messenger/metabolism , Atrial Natriuretic Factor/genetics
9.
Sci Rep ; 12(1): 12740, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35882940

ABSTRACT

There is growing interest in 3-iodothyronamine (T1AM), an active thyroid hormone metabolite, that induces negative inotropic and chronotropic actions in the heart and exerts systemic hypothermic action. We explored the direct impact of T1AM on cardiomyocytes with a focus on the regulation of the intracellular temperature and natriuretic peptide (NP) expression. A thermoprobe was successfully introduced into neonatal rat cardiomyocytes, and the temperature-dependent changes in the fluorescence intensity ratio were measured using a fluorescence microscope. After one-hour incubation with T1AM, the degree of change in the fluorescence intensity ratio was significantly lower in T1AM-treated cardiomyocytes than in equivalent solvent-treated controls (P < 0.01), indicating the direct hypothermic action of T1AM on cardiomyocytes. Furthermore, T1AM treatment upregulated B-type NP (BNP) gene expression comparable to treatment with endothelin-1 or phenylephrine. Of note, ERK phosphorylation was markedly increased after T1AM treatment, and inhibition of ERK phosphorylation by an MEK inhibitor completely cancelled both T1AM-induced decrease in thermoprobe-measured temperature and the increase in BNP expression. In summary, T1AM decreases fluorescent thermoprobe-measured temperatures (estimated intracellular temperatures) and increases BNP expression in cardiomyocytes by activating the MEK/ERK pathway. The present findings provide new insight into the direct myocardial cellular actions of T1AM in patients with severe heart failure.


Subject(s)
Myocytes, Cardiac , Natriuretic Peptides , Animals , Mitogen-Activated Protein Kinase Kinases , Natriuretic Peptide, Brain/genetics , Rats , Temperature , Thyronines
10.
Cell Biochem Biophys ; 80(3): 547-554, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35776316

ABSTRACT

Oxidized low-density lipoprotein (ox-LDL) is a type of modified cholesterol that promotes apoptosis and inflammation and advances the progression of heart failure. Leucine-zipper and sterile-α motif kinase (ZAK) is a kinase of the MAP3K family which is highly expressed in the heart and encodes two variants, ZAKα and ZAKß. Our previous study serendipitously found opposite effects of ZAKα and ZAKß in which ZAKß antagonizes ZAKα-induced apoptosis and hypertrophy of the heart. This study aims to test the hypothesis of whether ZAKα and ZAKß are involved in the damaging effects of ox-LDL in the cardiomyoblast. Cardiomyoblast cells H9c2 were treated with different concentrations of ox-LDL. Cell viability and apoptosis were measured by MTT and TUNEL assay, respectively. Western blot was used to detect apoptosis, hypertrophy, and pro-survival signaling proteins. Plasmid transfection, pharmacological inhibition with D2825, and siRNA transfection were utilized to upregulate or downregulate ZAKß, respectively. Ox-LDL concentration-dependently reduces the viability and expression of several pro-survival proteins, such as phospho-PI3K, phospho-Akt, and Bcl-xL. Furthermore, ox-LDL increases cleaved caspase-3, cleaved caspase-9 as indicators of apoptosis and increases B-type natriuretic peptide (BNP) as an indicator of hypertrophy. Overexpression of ZAKß by plasmid transfection attenuates apoptosis and prevents upregulation of BNP. Importantly, these effects were abolished by inhibiting ZAKß either by D2825 or siZAKß application. Our results suggest that ZAKß upregulation in response to ox-LDL treatment confers protective effects on cardiomyoblast.


Subject(s)
Lipoproteins, LDL , Natriuretic Peptide, Brain , Animals , Apoptosis , Hypertrophy , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Natriuretic Peptide, Brain/genetics , Protein Kinases , Rats , Up-Regulation
11.
J Interv Card Electrophysiol ; 65(1): 153-163, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35553305

ABSTRACT

BACKGROUND: Cardiac hypertrophy is an adaptive and compensatory mechanism preserving cardiac output during detrimental stimuli. Circular RNAs (circRNAs) have been illustrated to exert important implications in the pathogenesis of multiple cardiovascular diseases (CVD) including demonstrated cardiac hypertrophy. Toll-like receptor 4 (TLR4) has been previously reported to be a crucial regulator in inflammatory response and cardiac hypertrophy. However, the role of circular isoforms derived from TLR4 in cardiac hypertrophy remains unclear. METHODS: Expression of circ-TLR4 and TLR4 in cardiomyocytes was detected by RT-qPCR. The indicators of cardiac hypertrophy responses, including cell surface area, atrial natriuretic factor (ANF), B-type natriuretic peptide (BNP) and ß-myosin heavy chain (ß-MHC) were measured by immunofluorescence staining and western blot. RIP assay was used to validate the interaction between circ-TLR4 and TLR4. RESULTS: Circ-TLR4 and TLR4 was up-regulated in cellular models of cardiac hypertrophy. Circ-TLR4 knockdown attenuated angiotensin II (Ang II)-induced hypertrophy responses in cardiomyocytes. Moreover, circ-TLR4 positively regulated TLR4 expression through recruiting FUS to stabilize TLR4 mRNA. Furthermore, TLR4 overexpression rescued the cardiac responses mediated by circ-TLR4 silencing. CONCLUSION: Circ-TLR4 promotes cardiac hypertrophy through recruiting FUS to stabilize TLR4 mRNA.


Subject(s)
Atrial Natriuretic Factor , Toll-Like Receptor 4 , Angiotensin II/adverse effects , Angiotensin II/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Humans , Myocytes, Cardiac , Myosin Heavy Chains/adverse effects , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/genetics , RNA, Circular/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein FUS/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
Cells ; 11(5)2022 02 22.
Article in English | MEDLINE | ID: mdl-35269388

ABSTRACT

Plasma concentrations of natriuretic peptides (NP) contribute to risk stratification and management of patients undergoing non-cardiac surgery. However, genetically determined variability in the levels of these biomarkers has been described previously. In the perioperative setting, genetic contribution to NP plasma level variability has not yet been determined. A cohort of 427 patients presenting for non-cardiac surgery was genotyped for single-nucleotide polymorphisms (SNPs) from the NPPA/NPPB locus. Haplotype population frequencies were estimated and adjusted haplotype trait associations for brain natriuretic peptide (BNP) and amino-terminal pro natriuretic peptide (NT-proBNP) were calculated. Five SNPs were included in the analysis. Compared to the reference haplotype TATAT (rs198358, rs5068, rs632793, rs198389, rs6676300), haplotype CACGC, with an estimated frequency of 4%, showed elevated BNP and NT-proBNP plasma concentrations by 44% and 94%, respectively. Haplotype CGCGC, with an estimated frequency of 9%, lowered NT-proBNP concentrations by 28%. ASA classification status III and IV, as well as coronary artery disease, were the strongest predictors of increased NP plasma levels. Inclusion of genetic information might improve perioperative risk stratification of patients based on adjusted thresholds of NP plasma levels.


Subject(s)
Coronary Artery Disease , Natriuretic Peptide, Brain , Atrial Natriuretic Factor/genetics , Coronary Artery Disease/genetics , Haplotypes/genetics , Humans , Natriuretic Peptide, Brain/genetics , Natriuretic Peptides , Nitrobenzoates , Peptide Fragments , Procainamide/analogs & derivatives
13.
Cell Tissue Res ; 388(2): 225-238, 2022 May.
Article in English | MEDLINE | ID: mdl-35171324

ABSTRACT

The diversified natriuretic peptide (NP) family, consisting of four CNPs (CNP1-4), ANP, BNP, and VNP, has been identified in the eel. Here, we successfully cloned additional cnp genes from the brain of eel (a basal teleost) and zebrafish (a later branching teleost). The genes were identified as paralogues of cnp4 generated by the third round of whole genome duplication (3R) in the teleost lineage, thereby being named eel cnp4b and zebrafish cnp4-like, respectively. To examine the histological patterns of their expressions, we employed a newly developed in situ hybridization (ISH) chain reaction using short hairpin DNAs, in addition to conventional ISH. Eel cnp4b was expressed in the medulla oblongata, while mRNAs of eel cnp4a (former cnp4) were localized in the preoptic area. In the zebrafish brain, cnp4-like mRNA was undetectable, while the known cnp4 was expressed in both the preoptic area and medulla oblongata. Together with the different mRNA distribution of cnp4a and cnp4b in eel peripheral tissues determined by RT-PCR and ISH, it is suggested that subfunctionalization by duplicated cnp4s in ancestral teleosts has been retained only in basal teleosts. Intriguingly, cnp4b-expressing neurons in the glossopharyngeal-vagal motor complex of the medulla oblongata were co-localized with choline acetyltransferase, suggesting an involvement of Cnp4b in swallowing and respiration functions that are modulated by the vagus. Since teleost Cnp4 is an ortholog of mammalian CNP, the identified localization of teleost Cnp4 will contribute to future studies aimed at deciphering the physiological functions of CNP.


Subject(s)
Gene Duplication , Natriuretic Peptide, C-Type , Animals , Atrial Natriuretic Factor/genetics , Mammals/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, C-Type/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/genetics , Zebrafish/metabolism
14.
Circ Heart Fail ; 15(3): e008910, 2022 03.
Article in English | MEDLINE | ID: mdl-34865514

ABSTRACT

BACKGROUND: Extrinsic control of cardiomyocyte metabolism is poorly understood in heart failure (HF). FGF21 (Fibroblast growth factor 21), a hormonal regulator of metabolism produced mainly in the liver and adipose tissue, is a prime candidate for such signaling. METHODS: To investigate this further, we examined blood and tissue obtained from human subjects with end-stage HF with reduced ejection fraction at the time of left ventricular assist device implantation and correlated serum FGF21 levels with cardiac gene expression, immunohistochemistry, and clinical parameters. RESULTS: Circulating FGF21 levels were substantially elevated in HF with reduced ejection fraction, compared with healthy subjects (HF with reduced ejection fraction: 834.4 [95% CI, 628.4-1040.3] pg/mL, n=40; controls: 146.0 [86.3-205.7] pg/mL, n=20, P=1.9×10-5). There was clear FGF21 staining in diseased cardiomyocytes, and circulating FGF21 levels negatively correlated with the expression of cardiac genes involved in ketone metabolism, consistent with cardiac FGF21 signaling. FGF21 gene expression was very low in failing and nonfailing hearts, suggesting extracardiac production of the circulating hormone. Circulating FGF21 levels were correlated with BNP (B-type natriuretic peptide) and total bilirubin, markers of chronic cardiac and hepatic congestion. CONCLUSIONS: Circulating FGF21 levels are elevated in HF with reduced ejection fraction and appear to bind to the heart. The liver is likely the main extracardiac source. This supports a model of hepatic FGF21 communication to diseased cardiomyocytes, defining a potential cardiohepatic signaling circuit in human HF.


Subject(s)
Fibroblast Growth Factors , Heart Failure , Ventricular Dysfunction, Left , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Heart Failure/genetics , Humans , Natriuretic Peptide, Brain/genetics
15.
Cardiovasc Res ; 118(9): 2124-2138, 2022 07 20.
Article in English | MEDLINE | ID: mdl-34329394

ABSTRACT

AIMS: The cardiac natriuretic peptides [atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP)] are important regulators of cardiovascular physiology, with reduced natriuretic peptide (NP) activity linked to multiple human cardiovascular diseases. We hypothesized that deficiency of either ANP or BNP would lead to similar changes in left ventricular structure and function given their shared receptor affinities. METHODS AND RESULTS: We directly compared murine models deficient of ANP or BNP in the same genetic backgrounds (C57BL6/J) and environments. We evaluated control, ANP-deficient (Nppa-/-) or BNP-deficient (Nppb-/-) mice under unstressed conditions and multiple forms of pathological myocardial stress. Survival, myocardial structure, function and electrophysiology, tissue histology, and biochemical analyses were evaluated in the groups. In vitro validation of our findings was performed using human-derived induced pluripotent stem cell cardiomyocytes (iPS-CMs). In the unstressed state, both ANP- and BNP-deficient mice displayed mild ventricular hypertrophy which did not increase up to 1 year of life. NP-deficient mice exposed to acute myocardial stress secondary to thoracic aortic constriction (TAC) had similar pathological myocardial remodelling but a significant increase in sudden death. We discovered that the NP-deficient mice are more susceptible to stress-induced ventricular arrhythmias using both in vivo and ex vivo models. Mechanistically, deficiency of either ANP or BNP led to reduced myocardial cGMP levels and reduced phosphorylation of the cAMP response element-binding protein (CREBS133) transcriptional regulator. Selective CREB inhibition sensitized wild-type hearts to stress-induced ventricular arrhythmias. ANP and BNP regulate cardiomyocyte CREBS133 phosphorylation through a cGMP-dependent protein kinase 1 (PKG1) and p38 mitogen-activated protein kinase (p38 MAPK) signalling cascade. CONCLUSIONS: Our data show that ANP and BNP act in a non-redundant fashion to maintain myocardial cGMP levels to regulate cardiomyocyte p38 MAPK and CREB activity. Cardiac natriuretic peptide deficiency leads to a reduction in CREB signalling which sensitizes the heart to stress-induced ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac , Atrial Natriuretic Factor , Natriuretic Peptide, Brain , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cyclic GMP , Mice , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptides/metabolism , Vasodilator Agents , p38 Mitogen-Activated Protein Kinases
16.
ESC Heart Fail ; 9(1): 729-739, 2022 02.
Article in English | MEDLINE | ID: mdl-34734498

ABSTRACT

AIMS: Elevated brain natriuretic peptide (BNP) and the N-terminal fragment of its pro-hormone (NT-proBNP) have become established biomarkers for heart failure and are associated with cardiovascular morbidity and mortality. Investigating sources of inter-individual heterogeneity, particularly genetic factors, could help better identify patients at risk of future cardiovascular disease. The aim of this study was to estimate the heritability of circulating NT-proBNP levels, to perform a genome-wide association study (GWAS) and gene-candidate analysis focused on NPPB-NPPA genes on these levels, and to examine their association with cardiovascular or metabolic outcomes. METHODS AND RESULTS: A total of 1555 individuals from the STANISLAS study were included. The heritability of circulating NT-proBNP levels was estimated at 15%, with seven single nucleotide polymorphisms (SNPs) reaching the significant threshold in the GWAS. All above SNPs were located on the same gene cluster constituted of MTHFR, CLCN6, NPPA, NPPB, and C1orf167. NPPA gene expression was also associated with NT-proBNP levels. Moreover, six other SNPs from NPPA-NPPB genes were associated with diastolic function (lateral e' on echocardiography) and metabolic features (glycated haemoglobin). CONCLUSIONS: The heritability of natriuretic peptides appears relatively low (15%) and mainly based on the same gene cluster constituted of MTHFR, CLCN6, NPPA, NPPB, and C1orf167. Natriuretic peptide polymorphisms are associated with natriuretic peptide levels and diastolic function. These results suggest that natriuretic peptide polymorphisms may have an impact in the early stages of cardiovascular and metabolic disease.


Subject(s)
Atrial Natriuretic Factor , Genome-Wide Association Study , Atrial Natriuretic Factor/metabolism , Cohort Studies , Humans , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptides , Polymorphism, Single Nucleotide
17.
Elife ; 102021 12 17.
Article in English | MEDLINE | ID: mdl-34919054

ABSTRACT

Histamine-dependent and -independent itch is conveyed by parallel peripheral neural pathways that express gastrin-releasing peptide (GRP) and neuromedin B (NMB), respectively, to the spinal cord of mice. B-type natriuretic peptide (BNP) has been proposed to transmit both types of itch via its receptor NPRA encoded by Npr1. However, BNP also binds to its cognate receptor, NPRC encoded by Npr3 with equal potency. Moreover, natriuretic peptides (NP) signal through the Gi-couped inhibitory cGMP pathway that is supposed to inhibit neuronal activity, raising the question of how BNP may transmit itch information. Here, we report that Npr3 expression in laminae I-II of the dorsal horn partially overlaps with NMB receptor (NMBR) that transmits histaminergic itch via Gq-couped PLCß-Ca2+ signaling pathway. Functional studies indicate that NPRC is required for itch evoked by histamine but not chloroquine (CQ), a nonhistaminergic pruritogen. Importantly, BNP significantly facilitates scratching behaviors mediated by NMB, but not GRP. Consistently, BNP evoked Ca2+ responses in NMBR/NPRC HEK 293 cells and NMBR/NPRC dorsal horn neurons. These results reveal a previously unknown mechanism by which BNP facilitates NMB-encoded itch through a novel NPRC-NMBR cross-signaling in mice. Our studies uncover distinct modes of action for neuropeptides in transmission and modulation of itch in mice.


An itch is a common sensation that makes us want to scratch. Most short-term itches are caused by histamine, a chemical that is released by immune cells following an infection or in response to an allergic reaction. Chronic itching, on the other hand, is not usually triggered by histamine, and is typically the result of neurological or skin disorders, such as atopic dermatitis. The sensation of itching is generated by signals that travel from the skin to nerve cells in the spinal cord. Studies in mice have shown that the neuropeptides responsible for delivering these signals differ depending on whether or not the itch involves histamine: GRPs (short for gastrin-releasing proteins) convey histamine-independent itches, while NMBs (short for neuromedin B) convey histamine-dependent itches. It has been proposed that another neuropeptide called BNP (short for B-type natriuretic peptide) is able to transmit both types of itch signals to the spinal cord. But it remains unclear how this signaling molecule is able to do this. To investigate, Meng, Liu, Liu, Liu et al. carried out a combination of behavioral, molecular and pharmacological experiments in mice and nerve cells cultured in a laboratory. The experiments showed that BNP alone cannot transmit the sensation of itching, but it can boost itching signals that are triggered by histamine. It is widely believed that BNP activates a receptor protein called NPRA. However, Meng et al. found that the BNP actually binds to another protein which alters the function of the receptor activated by NMBs. These findings suggest that BNP modulates rather than initiates histamine-dependent itching by enhancing the interaction between NMBs and their receptor. Understanding how itch signals travel from the skin to neurons in the spinal cord is crucial for designing new treatments for chronic itching. The work by Meng et al. suggests that treatments targeting NPRA, which was thought to be a key itch receptor, may not be effective against chronic itching, and that other drug targets need to be explored.


Subject(s)
Natriuretic Peptide, Brain/genetics , Neurokinin B/analogs & derivatives , Pruritus/genetics , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Animals , Ganglia, Spinal/metabolism , HEK293 Cells , Histamine/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Natriuretic Peptide, Brain/metabolism , Neurokinin B/genetics , Neurokinin B/metabolism , Pruritus/physiopathology , Receptors, Atrial Natriuretic Factor/metabolism , Spinal Cord/metabolism
18.
J Cardiovasc Pharmacol ; 78(6): 792-801, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34882111

ABSTRACT

ABSTRACT: Left-ventricular hypertrophy, characterized by cardiomyocyte hypertrophy, interstitial cell proliferation, and immune cell infiltration, is a high risk factor for heart failure and death. Chemokines interacting with G protein-coupled chemokine receptors probably play a role in left-ventricular hypertrophy development by promoting recruitment of activated leukocytes and modulating left-ventricular remodeling. Using the minimally invasive model of transverse aortic constriction in mice, we demonstrated that a variety of chemokine and chemokine receptor messenger Ribonucleic Acid are overexpressed in the early and late phase of hypertrophy progression. Among the chemokine receptors, Cx3cr1 and Ccr2 were most strongly overexpressed and were significantly upregulated at 3, 7, and 14 days after transverse aortic constriction. Ligands of CX3CR1 (Cx3cl1) and CCR2 (Ccl2, Ccl7, Ccl12) were significantly overexpressed in the left ventricle at the early stages after mechanical pressure overload. Pharmacological inhibition of CX3CR1 signaling using the antagonist AZD8797 led to a significant reduction of hypertrophy, whereas inhibition of CCR2 with the RS504393 antagonist did not show any effect. Furthermore, AZD8797 treatment reduced the expression of the hypertrophic marker genes Nppa and Nppb as well as the profibrotic genes Tgfb1 and Col1a1 at 14 days after transverse aortic constriction. These findings strongly suggest the involvement of the CX3CR1/CX3CL1 pathway in the pathogenesis of left-ventricular hypertrophy.


Subject(s)
CX3C Chemokine Receptor 1/antagonists & inhibitors , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/drug effects , Pyrimidines/pharmacology , Thiazoles/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Aorta/physiopathology , Aorta/surgery , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Constriction , Disease Models, Animal , Fibrosis , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Signal Transduction , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
19.
Int Heart J ; 62(6): 1379-1386, 2021.
Article in English | MEDLINE | ID: mdl-34853228

ABSTRACT

Clinical studies have indicated that 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, also known as statins, can potentially inhibit chronic heart failure. In the Stat-LVDF study, a difference was noted in terms of the effect of lipophilic pitavastatin (PTV) and hydrophilic rosuvastatin (RSV) on plasma BNP, suggesting that statin lipophilicity and pharmacokinetics change the pleiotropic effect on heart failure in humans. Therefore, we assessed the beneficial effects of PTV on hypertrophy in cardiac myocytes compared with RSV at clinically used doses. Cultured cardiomyocytes were stimulated with 30 µM phenylephrine (PE) in the presence of PTV (250 nM) or RSV (50 nM). These doses were calculated based on the maximum blood concentration of statins used in clinical situations in Japan. The results showed that PTV, but not RSV, significantly inhibits the PE-induced increase in cell size and leucine incorporation without causing cell toxicity. In addition, PTV significantly suppressed PE-induced mRNA expression of hypertrophic response genes. PE-induced ERK phosphorylation was inhibited by PTV, but not by RSV. Furthermore, PTV significantly suppressed the angiotensin-II-induced proline incorporation in primary cultured cardiac fibroblasts. In conclusion, a clinical dose of PTV was noted to directly inhibit cardiomyocyte hypertrophy and cardiac fibrosis, suggesting that lipophilic PTV can be a potential drug candidate against chronic heart failure.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Myocytes, Cardiac/drug effects , Quinolines/administration & dosage , Rosuvastatin Calcium/administration & dosage , Actins/genetics , Actins/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Hypertrophy , Leucine/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Phosphorylation/drug effects , RNA, Messenger/metabolism , Rats, Sprague-Dawley
20.
Biomed Pharmacother ; 143: 112178, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34649308

ABSTRACT

Modified citrus pectin (MCP) is a specific inhibitor of galectin-3 (Gal-3) that is regarded as a new biomarker of cardiac hypertrophy, but its effect is unclear. The aim of this study is to investigate the role and mechanism of MCP in isoproterenol (ISO)-induced cardiac hypertrophy. Rats were injected with ISO to induce cardiac hypertrophy and treated with MCP. Cardiac function was detected by ECG and echocardiography. Pathomorphological changes were evaluated by the haematoxylin eosin (H&E) and wheat germ agglutinin (WGA) staining. The hypertrophy-related genes for atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and ß-myosin heavy chain (ß-MHC), and the associated signal molecules were analysed by qRT-PCR and western blotting. The results show that MCP prevented cardiac hypertrophy and ameliorated cardiac dysfunction and structural disorder. MCP also decreased the levels of ANP, BNP, and ß-MHC and inhibited the expression of Gal-3 and Toll-like receptor 4 (TLR4). Additionally, MCP blocked the phosphorylation of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3), but it promoted the phosphorylation of p38. Thus, MCP prevented ISO-induced cardiac hypertrophy by activating p38 signalling and inhibiting the Gal-3/TLR4/JAK2/STAT3 pathway.


Subject(s)
Cardiomegaly/drug therapy , Cardiovascular Agents/pharmacology , Janus Kinase 2/metabolism , Myocytes, Cardiac/drug effects , Pectins/pharmacology , STAT3 Transcription Factor/metabolism , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Disease Models, Animal , Galectin 3/metabolism , Isoproterenol , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Phosphorylation , Rats, Wistar , Signal Transduction , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...