Abstract P513: Adenylosuccinate Synthase Is A Novel Methylation Target Of Smyd1

2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Magnus Creed ◽  
Marta Szulik ◽  
Ryan Bia ◽  
Chris Tracy ◽  
Aman Makaju ◽  
...  

Among the metabolic shifts in chronic heart failure is a dysregulation of purine metabolism, which has been shown to negatively impact patient outcomes, especially in individuals affected by hypertension, diabetes, and congestive heart failure, via increased serum uric acid levels and cellular oxidative stress. The underlying mechanisms which drive these changes in purine metabolism in the cardiomyocyte and ultimately reactive oxygen species and uric acid accumulation in heart failure patients remain largely unknown. We recently discovered that the methyltransferase Smyd1 interacts with the metabolic enzyme Adss (Adenylosuccinate synthetase), a key component of purine metabolism in the heart involved in AMP synthesis, via co-immunoprecipitation. We confirmed this novel interaction between Smyd1b and Adss in mouse heart and cultured primary cardiomyocytes, which is further enhanced during phenylephrine-induced hypertrophy in the latter. Our hypothesis was that Smyd1b methylates Adss to regulate its activity, therefore, we examined lysine methylation on Adss via western blotting and mass spectrometry and quantified its ability to convert IMP to sAMP in vitro in the presence and absence of Smyd1b. Using a pan-methylation antibody we initially detected di- and tri-methylation on Adss which was increased in the presence of Smyd1b. Then utilizing bottom-up proteomics, we achieved 98% sequence coverage of Adss via mass spectrometry and identified trimethylation on K373 only in the presence of Smyd1b. In addition, utilizing an enzymatic assay in vitro we have shown that Smyd1b enhances the activity of Adss as it converts IMP to s-AMP. While it has been well-established that the activities of metabolic enzymes are modulated via post-translational modifications (e.g. phosphorylation, acetylation), we believe this is the first report of a metabolic enzyme regulated by lysine methylation. These exciting results highlight a novel role for Smyd1b in regulating purine metabolism in the myocyte and begin to lay the groundwork for examining this mechanism in the setting of disease.

2015 ◽  
Vol 14 (5) ◽  
pp. 2298-2311 ◽  
Author(s):  
Anthony W. Herren ◽  
Darren M. Weber ◽  
Robert R. Rigor ◽  
Kenneth B. Margulies ◽  
Brett S. Phinney ◽  
...  

2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Jing Ren ◽  
Hanping Qi ◽  
Chao Song ◽  
Lina Ba ◽  
Renling Liu ◽  
...  

AbstractCardiac hypertrophy is a common pathological change accompanied by various cardiovascular diseases; however, its underlying mechanisms remain elusive. Mounting evidence indicates that long non-coding RNAs (lncRNAs) are novel transcripts involved in regulating multiple biological processes. However, little is known about their role in regulating cardiac hypertrophy. This study revealed a novel lncRNA4930473A02Rik (abbreviated as lncRNAA02Rik), which showed considerably increased expression in hypertrophic mouse hearts in vivo and angiotensin-II (Ang-II)-induced hypertrophic cardiomyocytes in vitro. Notably, lncRNAA02Rik knockdown partly ameliorated Ang-II induced hypertrophic cardiomyocytes in vitro and hypertrophic mouse heart function in vivo, whereas lncRNAA02Rik overexpression promoted cardiac hypertrophy in vitro. Furthermore, lncRNAA02Rik acted as a competing endogenous RNA by sponging miR-135a, while forced expression of lncRNAA02Rik could repress its activity and expression. Furthermore, forcing miR-135a overexpression exerted a significant protective effect against cardiac hypertrophy by inhibiting the activity of its downstream target TCF7, a critical member of Wnt signaling, and the protective effect could be reversed by AMO-135a. Luciferase assay showed direct interactions among lncRNAA02Rik, miR-135a, and TCF7. Altogether, our study demonstrated that lncRNAA02Rik upregulation could promote cardiac hypertrophy development via modulating miR-135a expression levels and TCF7 activity. Therefore, lncRNAA02Rik inhibition might be considered as a novel potential therapeutic strategy for cardiac hypertrophy.


2021 ◽  
Vol 2021 ◽  
pp. 1-17
Author(s):  
Yang Zhao ◽  
Huaxin Sun ◽  
Kai Li ◽  
Luxiang Shang ◽  
Xiaoyan Liang ◽  
...  

Background. Cholinergic anti-inflammatory pathway (CAP) is implicated in cardioprotection in chronic heart failure (CHF) by downregulating inflammation response. Mitochondrial injuries play an important role in ventricular remodeling of the CHF process. Herein, we aim to investigate whether CAP elicitation prevents ventricular remodeling in CHF by protecting myocardial mitochondrial injuries and its underlying mechanisms. Methods and Results. CHF models were established by ligation of anterior descending artery for 5 weeks. Postoperative survival rats were assigned into 5 groups: the sham group (sham, n = 10 ), CHF group (CHF, n = 11 ), Vag group (CHF+vagotomy, n = 10 ), PNU group (CHF+PNU-282987 for 4 weeks, n = 11 ), and Vag+PNU group (CHF+vagotomy+PNU-282987 for 4 weeks, n = 10 ). The antiventricular remodeling effect of cholinergic elicitation was evaluated in vivo, and H9C2 cells were selected for the TNF-α gradient stimulation experiment in vitro. In vivo, CAP agitated by PNU-282987 alleviated the left ventricular dysfunction and inhibited the energy metabolism remodeling. Further, cholinergic elicitation increased myocardium ATP levels and reduced systemic inflammation. CAP induction alleviates macrophage infiltration and cardiac fibrosis, of which the effect is counteracted by vagotomy. Myocardial mitochondrial injuries were ameliorated by CAP activation, including the reserved ultrastructural integrity, declining ROS overload, reduced myocardial apoptosis, and enhanced mitochondrial fusion. In vitro, TNF-α intervention significantly exacerbated the mitochondrial damage in H9C2 cells. Conclusion. CAP elicitation effectively improves ischemic ventricular remodeling by suppressing systemic and cardiac inflammatory response, attenuating cardiac fibrosis and potentially alleviating the mitochondrial dysfunction linked to hyperinflammation reaction.


2021 ◽  
Author(s):  
Mengli Chen ◽  
Hongyan Zhu ◽  
Qingqing Zhu ◽  
Xiaodong Wu ◽  
Yufei Zhou ◽  
...  

Abstract PurposeHeart failure after myocardial infarction (MI) is the leading cause of death worldwide. Citri Reticulatae Pericarpium (CRP) is a traditional Chinese herbal medicine that has been used in the clinic for centuries. In this study, we aimed to investigate the roles of CRP in cardiac remodeling and heart failure after MI, as well as the molecular mechanisms involved.MethodsMale C57BL/6 mice aged 8 weeks were subjected to coronary artery ligation to mimic the clinical situation in vivo. Echocardiography was used to assess the systolic function of the mouse heart. Masson trichrome staining and Wheat germ agglutinin (WGA) staining were utilized to determine the fibrotic area and cross-sectional area of the mouse heart, respectively. Cardiomyocytes and fibroblasts were isolated from neonatal rats aged 0–3 days in vitro using enzyme digestion. TUNEL staining and EdU staining were performed to evaluate apoptosis and proliferation, respectively. Gene expression changes were analyzed by qRT–PCR, and protein expression changes were assessed by Western blotting.ResultsOur findings revealed that CRP attenuated cardiac hypertrophy, fibrosis and apoptosis and alleviated heart failure after MI in vivo. Furthermore, CRP mitigated cardiomyocyte apoptosis and fibroblast proliferation and differentiation into myofibroblasts. In addition, the PPARγ inhibitor T0070907 completely abolished the abovementioned beneficial effects of CRP, and the PPARγ activator rosiglitazone failed to further ameliorate cardiac apoptosis and fibrosis in vitro.ConclusionCRP alleviates cardiac hypertrophy, fibrosis, and apoptosis and can ameliorate heart failure after MI via activation of PPARγ.


2014 ◽  
Vol 2014 ◽  
pp. 1-12 ◽  
Author(s):  
Yuan Cao ◽  
Yang Ruan ◽  
Tao Shen ◽  
Xiuqing Huang ◽  
Meng Li ◽  
...  

Background. Doxorubicin, a potent chemotherapeutic agent, is associated with acute and chronic cardiotoxicity, which is cumulatively dose-dependent.Astragaluspolysaccharide (APS), the extract ofAstragalus membranaceuswith strong antitumor and antiglomerulonephritis activity, can effectively alleviate inflammation. However, whether APS could ameliorate chemotherapy-induced cardiotoxicity is not understood. Here, we investigated the protective effects of APS on doxorubicin-induced cardiotoxicity and elucidated the underlying mechanisms of the protective effects of APS.Methods. We analyzed myocardial injury in cancer patients who underwent doxorubicin chemotherapy and generated a doxorubicin-induced neonatal rat cardiomyocyte injury model and a mouse heart failure model. Echocardiography, reactive oxygen species (ROS) production, TUNEL, DNA laddering, and Western blotting were performed to observe cell survival, oxidative stress, and inflammatory signal pathways in cardiomyocytes.Results. Treatment of patients with the chemotherapeutic drug doxorubicin led to heart dysfunction. Doxorubicin reduced cardiomyocyte viability and induced C57BL/6J mouse heart failure with concurrent elevated ROS generation and apoptosis, which, however, was attenuated by APS treatment. In addition, there was profound inhibition of p38MAPK and activation of Akt after APS treatment.Conclusions. These results demonstrate that APS could suppress oxidative stress and apoptosis, ameliorating doxorubicin-mediated cardiotoxicity by regulating the PI3k/Akt and p38MAPK pathways.


Antioxidants ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 358 ◽  
Author(s):  
Alessandra Altomare ◽  
Giovanna Baron ◽  
Marta Balbinot ◽  
Alessandro Pedretti ◽  
Beatrice Zoanni ◽  
...  

Advanced glycation end-products (AGEs) and advanced lipoxidation end-products (ALEs), particularly carboxymethyl-lysine (CML), have been largely proposed as factors involved in the establishment and progression of heart failure (HF). Despite this evidence, the current literature lacks the comprehensive identification and characterization of the plasma AGEs/ALEs involved in HF (untargeted approach). This work provides the first ex vivo high-resolution mass spectrometry (HR-MS) profiling of AGEs/ALEs occurring in human serum albumin (HSA), the most abundant protein in plasma, characterized by several nucleophilic sites and thus representing the main protein substrate for AGE/ALE formation. A set of AGE/ALE adducts in pooled HF-HSA samples was defined, and a semi-quantitative analysis was carried out in order to finally select those presenting in increased amounts in the HF samples with respect to the control condition. These adducts were statistically confirmed by monitoring their content in individual HF samples by applying a targeted approach. Selected AGEs/ALEs proved to be mostly CML derivatives on Lys residues (i.e., CML-Lys12, CML-Lys378, CML-Lys402), and one deoxy-fructosyl derivative on the Lys 389 (DFK-Lys 389). The nature of CML adducts was finally confirmed using immunological methods and in vitro production of such adducts further confirmed by mass spectrometry.


Pharmaceutics ◽  
2020 ◽  
Vol 13 (1) ◽  
pp. 43
Author(s):  
Young Hee Choi ◽  
Young-Won Chin

Metabolic enzyme and/or transporter-mediated pharmacokinetic (PK) changes in a drug caused by concomitant herbal products have been a primary issue of herb and drug interactions (HDIs), because PK changes of a drug may result in the alternation of efficacy and toxicity. Studies on HDIs have been carried out by predictive in vitro and in vivo preclinical studies, and clinical trials. Nevertheless, the discrepancies between predictive data and the clinical significance on HDIs still exist, and different reports of HDIs add to rather than clarify the confusion regarding the use of herbal products and drug combinations. Here, we briefly review the underlying mechanisms causing PK-based HDIs, and more importantly summarize challenging issues, such as dose and treatment period effects, to be considered in study designs and interpretations of HDI evaluations.


PeerJ ◽  
2021 ◽  
Vol 9 ◽  
pp. e11209
Author(s):  
Yi-Wei Kuo ◽  
Shih-Hung Hsieh ◽  
Jui-Fen Chen ◽  
Cheng-Ruei Liu ◽  
Ching-Wei Chen ◽  
...  

Background Uric acid (UA) is the end product of purine metabolism in the liver and is excreted by the kidneys. When purine metabolism is impaired, the serum UA level will be elevated (hyperuricemia) and eventually lead to gout. During evolution, humans and some primates have lost the gene encoding uricase, which is vital in UA metabolism. With the advances of human society, the prevalence of hyperuricemia has dramatically increased because of the refined food culture. Hyperuricemia can be controlled by drugs, such as allopurinol and probenecid. However, these drugs have no preventive effect and are associated with unpleasant side effects. An increasing number of probiotic strains, which are able to regulate host metabolism and prevent chronic diseases without harmful side effects, have been characterized. The identification of probiotic strains, which are able to exert beneficial effects on UA metabolism, will provide an alternative healthcare strategy for patients with hyperuricemia, especially for those who are allergic to anti-hyperuricemia drugs. Methods To elicit hyperuricemia, rats in the symptom control group (HP) were injected with potassium oxonate and fed a high-purine diet. Rats in the probiotic groups received the high-purine diet, oxonate injection, and supplements of probiotic strains TSR332, TSF331, or La322. Rats in the blank control group (C) received a standard diet (AIN-93G) and oxonate injection. Results Purine-utilizing strains of probiotics were screened using high-pressure liquid chromatography (HPLC) in vitro, and the lowering effect on serum UA levels was analyzed in hyperuricemia rats in vivo. We found that Lactobacillus reuteri strain TSR332 and Lactobacillus fermentum strain TSF331 displayed significantly strong assimilation of inosine (90%; p = 0.00003 and 59%; p = 0.00545, respectively) and guanosine (78%; p = 0.00012 and 51%; p = 0.00062, respectively) within 30 min in vitro. Further animal studies revealed that serum UA levels were significantly reduced by 60% (p = 0.00169) and 30% (p = 0.00912), respectively, in hyperuricemic rats treated with TSR332 and TSF331 for 8 days. Remarkably, TSR332 ameliorated the occurrence of hyperuricemia, and no evident side effects were observed. Overall, our study indicates that TSR332 and TSF331 are potential functional probiotic strains for controlling the development of hyperuricemia.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 1328.3-1329
Author(s):  
M. Chen ◽  
X. Lu ◽  
H. Wu

Background:About 20% of individuals in the USA have asymptomatic hyperuricaemia[1]. However, Urate-lowering therapy in asymptomatic hyperuricaemia condition is still controversial considering the benefit and side effects[2]. Therefore, safe and effective anti-hyperuricemia therapies are necessary.Objectives:Bergenin, the major bioactive ingredient isolated from Saxifraga stolonifera, could activate SIRT1. In this study, we identify the effect of bergenin on hyperuricemia, and explored the related mechanisms.Methods:Significant hyperuricemia was established in C57BL/6N mice treated with oxonate and yeast polysaccharide. Bergenin was administered to the mice at the same time. The serum uric acid and creatinine levels, clearance of uric acid and creatinine, the intestinal uric acid excretion, and renal pathological lesions were determined were used to evaluate the anti-hyperuricemic effects. The location and expression levels of ABCG2 in the kidney and intestine were analyzed. HK-2 and Caco-2 cell lines were exposed to soluble uric acid with or without the treatment of Bergenin. Then the expression of ABCG2 and underlying mechanisms were explored.Results:The administration of bergenin decreased serum uric acid in hyperuricemic mice by the promotion of uric acid excretion both in kidney and intestine. Bergenin recued the downregulation of ABCG2 in the kidney of hyperuricemic mice and upregulated the expression of ABCG2 in the jejunum and ileum. In vitro, Bergenin significantly increased the expression of ABCG2 as well as activated SIRT1, which was reversed by addition of PPARg antagonist GW9662 and siPPARg.Conclusion:These findings suggest bergenin increases uric acid excretion both in the kidney and intestines, which may be related to the upregulation of ABCG2 via SIRT1- PPARg pathway.References:[1]Zhu, Y., Pandya, B. J. & Choi, H. K. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007–2008. Arthritis Rheum. 63, 3136–3141 (2011).[2]Joosten LAB, Crisan TO, Bjornstad P, Johnson RJ: Asymptomatic hyperuricaemia: a silent activator of the innate immune system. Nature reviews Rheumatology 2020, 16(2):75-86.Disclosure of Interests:None declared


2018 ◽  
Vol 45 (5) ◽  
pp. 2122-2135 ◽  
Author(s):  
Meiling Yan ◽  
Kankai Chen ◽  
Li He ◽  
Shuai Li ◽  
Dong Huang ◽  
...  

Background/Aims: Hyperuricemia is associated with an increased risk for multiple cardiovascular diseases, but the underlying mechanisms remain largely elusive. Calpain-1 is a protease that is implicated in several pathological conditions that affect the heart. The aim of this current study was to test the effects of uric acid (UA) on cardiomyocyte survival and cardiac function and to investigate the role of calpain-1 in the UA-induced effects in the heart and their underlying mechanisms. Methods: In vivo, hyperuricemia was induced by oxonic acid (OA) administration in Sprague-Dawley rats for 16 weeks; TUNEL staining was used to identify apoptotic cells. Left ventricular (LV) sections were stained with Sirius Red to evaluate interstitial fibrosis. Cardiac catheterization was performed to evaluate cardiac function. In vitro, cultured H9c2 cells were incubated with different UA concentrations. MTT assays and flow cytometry were used to evaluate cell viability and apoptosis. All related gene expression levels were analyzed by quantitative real-time PCR (qRT-PCR), and all protein expression levels were analyzed by western blotting. Results: Hyperuricemia induction in vivo resulted in cellular apoptosis, interstitial fibrosis and diastolic dysfunction in the rat hearts, as well as increased activation of calpain-1 and endoplasmic reticulum (ER) stress, while allopurinol treatment mitigated the above changes. UA administration in vitro increased apoptosis and decreased H9c2 cell viability in a dose-dependent manner. Increased activation of calpain-1 and ER stress was also observed in the groups with high UA levels. Calpain-1 siRNA and the calpain inhibitor CI-III alleviated UA-induced ER stress and apoptosis, while inhibiting ER stress by tauroursodeoxycholic acid (TUDCA) mitigated UA-induced apoptosis without affecting calpain-1 expression or activity. Conclusions: These findings suggest that UA induces cardiomyocyte apoptosis through activation of calpain-1 and ER stress. These results may provide new insights into the mechanisms of hyperuricemia-associated cardiovascular risks and hopefully identify new treatment targets.


Sign in / Sign up

Export Citation Format

Share Document