1
|
Kamel R, Kassouf S, Nasser SM, Mcheik A, Hayeck N, Abi-Ramia E, ElKazzaz H, Khalil C, Abi-Gerges A. Pulmonary effects of waterpipe generated smoke in adult diabetic rats. Toxicol Appl Pharmacol 2025; 499:117319. [PMID: 40185287 DOI: 10.1016/j.taap.2025.117319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Waterpipe smoking (WPS) is associated with pulmonary inflammation and DNA damage. Tobacco use among diabetic patients adds substantial clinical and public health burden. This study aims to investigate the combined pulmonary effects of diabetes and smoking. To achieve this goal, type 1 diabetes (T1D) was induced in adult male rats by Streptozotocin (65 mg/kg) injection. Rats were then exposed either to fresh air or WPS for one hour daily over five weeks (five days/week). Lung remodeling was evaluated by histology. Changes affecting inflammation, oxidative stress, apoptosis and survival pathways were characterized by real-time quantitative PCR and Western blot. Our findings showed that T1D was associated with pulmonary remodeling characterized by increases in lung weight/BW ratio, exacerbated by WPS, and elevated alveolar count. Both T1D and WPS exposure led to an accumulation of alveolar foamy macrophages and decreased alveolar septal thickness. Upregulation in the transcript levels of pro-inflammatory cytokine, TNF-α and anti-inflammatory marker, IL-10, were reported in diabetic lungs irrespective of WPS exposure. Moreover, diabetic lungs also displayed significant changes in the expression of mitochondrial complexes III and IV and antioxidant enzyme, SOD2, irrespective of the exposure condition. We also noted significant downregulation in the expression of caspases 3 and 9, p-P53/P53 ratio and JNK protein in diabetic lungs compared to control irrespective of the exposure condition. Lastly, diabetes and WPS exposure triggered significant decreases in EGFR expression. In conclusion, we show for the first time pulmonary remodeling and damages caused by the combined effects of T1D and smoking. Our findings highlight the pressing need for future better management of waterpipe consumption among patients with diabetes.
Collapse
Affiliation(s)
- Rima Kamel
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Sibelle Kassouf
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Selim M Nasser
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Amale Mcheik
- Department of Physical Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon
| | - Nathalie Hayeck
- Department of Physical Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon
| | - Elias Abi-Ramia
- Animal Facility, Institutional Review Board Office, Graduate Studies and Research, Lebanese American University, Lebanon
| | - Hanan ElKazzaz
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Christian Khalil
- School of Arts and Sciences, Department of Biological Sciences, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Aniella Abi-Gerges
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| |
Collapse
|
2
|
Bakhos JJ, Saliba Y, Hajal J, Achkouty G, Oskaridjian H, Albuquerque M, Azevedo C, Semaan A, Suffee N, Balse E, Hatem SN, Fares N. Inhibiting atrial natriuretic peptide clearance reduces myocardial fibrosis and improves cardiac function in diabetic rats. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf031. [PMID: 40201591 PMCID: PMC11977460 DOI: 10.1093/ehjopen/oeaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025]
Abstract
Aims Natriuretic peptides (NPs) exert pleiotropic effects through the recruitment of cyclic guanosine monophosphate (cGMP) signalling pathways depending on their bioavailability, which is regulated by clearance receptors and peptidases. Here, we tested the hypothesis that increasing myocardial bioavailability of NP has a beneficial effect on heart failure. We studied the effects of a mutated NP, M-atrial natriuretic peptide (MANP), resistant to neprilysin in a model of diabetic cardiomyopathy characterized by marked myocardial fibrosis. Methods and results Natriuretic peptides as well as sacubitril were delivered via osmotic mini-pumps to high-fat/streptozotocin-induced Type 2 diabetic (T2D) rats. Cardiac function was evaluated by echocardiography. Myocardial remodelling was studied by histological approaches, collagen phenotype, and atrial natriuretic peptide (ANP)/cGMP concentrations. Live-cell cGMP biosensing was conducted on cultured rat cardiac fibroblasts to investigate the biological effects of NP. Cyclic guanosine monophosphate signalling pathway was studied using multiple antibody arrays and biochemical assays in cardiac tissue and cultured fibroblasts. M-atrial natriuretic peptide exhibits superior efficacy than ANP in reducing left ventricular dysfunction and myocardial fibrosis with less extracellular matrix deposition. In vitro, MANP and ANP similarly generated cGMP and activated the protein kinase G (PKG) signalling pathway in cardiac fibroblasts, attenuating Mothers against decapentaplegic homolog 2 (SMAD) activation, collagen secretion, and cell proliferation. Nevertheless, in vivo, MANP specifically enhanced cardiac cGMP accumulation and was more potent than ANP in activating myocardial cGMP/PKG signalling and inhibiting the profibrotic SMAD, extracellular signal-regulated kinases 1/2, and nuclear factor of activated T cells 3 pathways. Endopeptidase inhibition using sacubitril also led to cardiac ANP/cGMP accumulation and reduced myocardial fibrosis. Conclusion Myocardial bioavailability of ANP is a major determinant of peptide efficacy in reducing cardiac fibrosis and improving pump function during diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jules Joel Bakhos
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Youakim Saliba
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Joelle Hajal
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Guy Achkouty
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Hrag Oskaridjian
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Miguel Albuquerque
- INSERM, Centre de Recherche sur L'inflammation, UMR 1149, Université Paris-Cité, 45 Rue des Saints-Pères 75006 Paris, France
- Service d'Anatomie Pathologique, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 100 Bd du Général Leclerc, 92110 Clichy, France
| | - Chloé Azevedo
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Albert Semaan
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Nadine Suffee
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Elise Balse
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Stéphane N Hatem
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| |
Collapse
|
3
|
Brotman SM, El-Sayed Moustafa JS, Guan L, Broadaway KA, Wang D, Jackson AU, Welch R, Currin KW, Tomlinson M, Vadlamudi S, Stringham HM, Roberts AL, Lakka TA, Oravilahti A, Fernandes Silva L, Narisu N, Erdos MR, Yan T, Bonnycastle LL, Raulerson CK, Raza Y, Yan X, Parker SCJ, Kuusisto J, Pajukanta P, Tuomilehto J, Collins FS, Boehnke M, Love MI, Koistinen HA, Laakso M, Mohlke KL, Small KS, Scott LJ. Adipose tissue eQTL meta-analysis highlights the contribution of allelic heterogeneity to gene expression regulation and cardiometabolic traits. Nat Genet 2025; 57:180-192. [PMID: 39747594 DOI: 10.1038/s41588-024-01982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/11/2024] [Indexed: 01/04/2025]
Abstract
Complete characterization of the genetic effects on gene expression is needed to elucidate tissue biology and the etiology of complex traits. In the present study, we analyzed 2,344 subcutaneous adipose tissue samples and identified 34,774 conditionally distinct expression quantitative trait locus (eQTL) signals at 18,476 genes. Over half of eQTL genes exhibited at least two eQTL signals. Compared with primary eQTL signals, nonprimary eQTL signals had lower effect sizes, lower minor allele frequencies and less promoter enrichment; they corresponded to genes with higher heritability and higher tolerance for loss of function. Colocalization of eQTLs with genome-wide association study (GWAS) signals for 28 cardiometabolic traits identified 1,835 genes. Inclusion of nonprimary eQTL signals increased discovery of colocalized GWAS-eQTL signals by 46%. Furthermore, 21 genes with ≥2 colocalized GWAS-eQTL signals showed a mediating gene dosage effect on the GWAS trait. Thus, expanded eQTL identification reveals more mechanisms underlying complex traits and improves understanding of the complexity of gene expression regulation.
Collapse
Affiliation(s)
- Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Li Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dongmeng Wang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Max Tomlinson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Yasrab Raza
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Xinyu Yan
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Stephen C J Parker
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics and Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK.
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Erciyes D, Bora ES, Tekindal MA, Erbaş O. Demonstration of the Protective Effect of Vinpocetine in Diabetic Cardiomyopathy. J Clin Med 2024; 13:4637. [PMID: 39200779 PMCID: PMC11354616 DOI: 10.3390/jcm13164637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Diabetic cardiomyopathy (DCM) poses a significant risk for heart failure in individuals with diabetes, yet its underlying mechanisms remain incompletely understood. Elevated blood sugar levels initiate harmful processes, including apoptosis, collagen accumulation, and fibrosis in the heart. Vinpocetine, a derivative of Vinca minor L., has demonstrated diverse pharmacological effects, including vasodilation, anti-inflammatory properties, and enhanced cellular metabolism. This study aims to investigate Vinpocetine's protective and remodeling effects in diabetic cardiomyopathy by evaluating biochemical and histopathological parameters. Methods: Twenty-one adult male Wistar rats were induced with diabetes using streptozocin and divided into Diabetes and Diabetes + Vinpocetine groups. Histopathological analyses, TGF-β1 immunoexpression, and measurements of plasma markers (TGF-β, pro-BNP, Troponin T) were performed. Biochemical analyses included HIF-1 alpha and neuregulin-1β quantification and evaluation of lipid peroxidation. Results: Vinpocetine significantly reduced cardiac muscle thickness, TGF-β1 expression, and plasma in diabetic rats. HIF-1 alpha and neuregulin-1β levels increased with Vinpocetine treatment. Histopathological observations confirmed reduced fibrosis and structural abnormalities in Vinpocetine-treated hearts. Conclusions: This study provides comprehensive evidence supporting the protective effects of Vinpocetine against diabetic cardiomyopathy. Vinpocetine treatment improved cardiac morphology, immunohistochemistry, and modulation of biochemical markers, suggesting its potential as a therapeutic intervention to attenuate the negative impact of diabetes on heart function.
Collapse
Affiliation(s)
- Demet Erciyes
- Department of Cardiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, 35620 Izmir, Türkiye
| | - Mustafa Agah Tekindal
- Department of Basic Medical Sciences Biostatistics, Faculty of Medicine, İzmir Katip Çelebi Unıversity, 35620 Izmir, Türkiye;
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, 34394 Istanbul, Türkiye;
| |
Collapse
|
5
|
Liu YB, Wang Q, Song YL, Song XM, Fan YC, Kong L, Zhang JS, Li S, Lv YJ, Li ZY, Dai JY, Qiu ZK. Abnormal phosphorylation / dephosphorylation and Ca 2+ dysfunction in heart failure. Heart Fail Rev 2024; 29:751-768. [PMID: 38498262 DOI: 10.1007/s10741-024-10395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Heart failure (HF) can be caused by a variety of causes characterized by abnormal myocardial systole and diastole. Ca2+ current through the L-type calcium channel (LTCC) on the membrane is the initial trigger signal for a cardiac cycle. Declined systole and diastole in HF are associated with dysfunction of myocardial Ca2+ function. This disorder can be correlated with unbalanced levels of phosphorylation / dephosphorylation of LTCC, endoplasmic reticulum (ER), and myofilament. Kinase and phosphatase activity changes along with HF progress, resulting in phased changes in the degree of phosphorylation / dephosphorylation. It is important to realize the phosphorylation / dephosphorylation differences between a normal and a failing heart. This review focuses on phosphorylation / dephosphorylation changes in the progression of HF and summarizes the effects of phosphorylation / dephosphorylation of LTCC, ER function, and myofilament function in normal conditions and HF based on previous experiments and clinical research. Also, we summarize current therapeutic methods based on abnormal phosphorylation / dephosphorylation and clarify potential therapeutic directions.
Collapse
Affiliation(s)
- Yan-Bing Liu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
- Medical College, Qingdao University, Qingdao, China
| | - Qian Wang
- Medical College, Qingdao University, Qingdao, China
| | - Yu-Ling Song
- Department of Pediatrics, Huantai County Hospital of Traditional Chinese Medicine, Zibo, China
| | | | - Yu-Chen Fan
- Medical College, Qingdao University, Qingdao, China
| | - Lin Kong
- Medical College, Qingdao University, Qingdao, China
| | | | - Sheng Li
- Medical College, Qingdao University, Qingdao, China
| | - Yi-Ju Lv
- Medical College, Qingdao University, Qingdao, China
| | - Ze-Yang Li
- Medical College, Qingdao University, Qingdao, China
| | - Jing-Yu Dai
- Department of Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| | - Zhen-Kang Qiu
- Interventional Medical Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
6
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
7
|
Brotman SM, El-Sayed Moustafa JS, Guan L, Broadaway KA, Wang D, Jackson AU, Welch R, Currin KW, Tomlinson M, Vadlamudi S, Stringham HM, Roberts AL, Lakka TA, Oravilahti A, Silva LF, Narisu N, Erdos MR, Yan T, Bonnycastle LL, Raulerson CK, Raza Y, Yan X, Parker SCJ, Kuusisto J, Pajukanta P, Tuomilehto J, Collins FS, Boehnke M, Love MI, Koistinen HA, Laakso M, Mohlke KL, Small KS, Scott LJ. Adipose tissue eQTL meta-analysis reveals the contribution of allelic heterogeneity to gene expression regulation and cardiometabolic traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.563798. [PMID: 37961277 PMCID: PMC10634839 DOI: 10.1101/2023.10.26.563798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Complete characterization of the genetic effects on gene expression is needed to elucidate tissue biology and the etiology of complex traits. Here, we analyzed 2,344 subcutaneous adipose tissue samples and identified 34K conditionally distinct expression quantitative trait locus (eQTL) signals in 18K genes. Over half of eQTL genes exhibited at least two eQTL signals. Compared to primary signals, non-primary signals had lower effect sizes, lower minor allele frequencies, and less promoter enrichment; they corresponded to genes with higher heritability and higher tolerance for loss of function. Colocalization of eQTL with conditionally distinct genome-wide association study signals for 28 cardiometabolic traits identified 3,605 eQTL signals for 1,861 genes. Inclusion of non-primary eQTL signals increased colocalized signals by 46%. Among 30 genes with ≥2 pairs of colocalized signals, 21 showed a mediating gene dosage effect on the trait. Thus, expanded eQTL identification reveals more mechanisms underlying complex traits and improves understanding of the complexity of gene expression regulation.
Collapse
Affiliation(s)
- Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | | | - Li Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - K Alaine Broadaway
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dongmeng Wang
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Max Tomlinson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Roberts
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Anniina Oravilahti
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Erdos
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lori L Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Yasrab Raza
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Xinyu Yan
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Stephen C J Parker
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics and Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Heikki A Koistinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
- Department of Medicine and Clinical Research, Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Jacques M, Landen S, Romero JA, Hiam D, Schittenhelm RB, Hanchapola I, Shah AD, Voisin S, Eynon N. Methylome and proteome integration in human skeletal muscle uncover group and individual responses to high-intensity interval training. FASEB J 2023; 37:e23184. [PMID: 37698381 DOI: 10.1096/fj.202300840rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/17/2023] [Accepted: 08/24/2023] [Indexed: 09/13/2023]
Abstract
Exercise is a major beneficial contributor to muscle metabolism, and health benefits acquired by exercise are a result of molecular shifts occurring across multiple molecular layers (i.e., epigenome, transcriptome, and proteome). Identifying robust, across-molecular level targets associated with exercise response, at both group and individual levels, is paramount to develop health guidelines and targeted health interventions. Sixteen, apparently healthy, moderately trained (VO2 max = 51.0 ± 10.6 mL min-1 kg-1 ) males (age range = 18-45 years) from the Gene SMART (Skeletal Muscle Adaptive Responses to Training) study completed a longitudinal study composed of 12-week high-intensity interval training (HIIT) intervention. Vastus lateralis muscle biopsies were collected at baseline and after 4, 8, and 12 weeks of HIIT. DNA methylation (~850 CpG sites) and proteomic (~3000 proteins) analyses were conducted at all time points. Mixed models were applied to estimate group and individual changes, and methylome and proteome integration was conducted using a holistic multilevel approach with the mixOmics package. A total of 461 proteins significantly changed over time (at 4, 8, and 12 weeks), whilst methylome overall shifted with training only one differentially methylated position (DMP) was significant (adj.p-value < .05). K-means analysis revealed cumulative protein changes by clusters of proteins that presented similar changes over time. Individual responses to training were observed in 101 proteins. Seven proteins had large effect-sizes >0.5, among them are two novel exercise-related proteins, LYRM7 and EPN1. Integration analysis showed bidirectional relationships between the methylome and proteome. We showed a significant influence of HIIT on the epigenome and more so on the proteome in human muscle, and uncovered groups of proteins clustering according to similar patterns across the exercise intervention. Individual responses to exercise were observed in the proteome with novel mitochondrial and metabolic proteins consistently changed across individuals. Future work is required to elucidate the role of these proteins in response to exercise.
Collapse
Affiliation(s)
- Macsue Jacques
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Shanie Landen
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Javier Alvarez Romero
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Danielle Hiam
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Institute of Nutrition and Health Sciences, Deakin University, Melbourne, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Iresha Hanchapola
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Anup D Shah
- Monash Proteomics & Metabolomics Facility, Monash University, Melbourne, Victoria, Australia
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nir Eynon
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Wang W, Yao W, Tan Q, Li S, Duan H, Tian X, Xu C, Zhang D. Identification of key DNA methylation changes on fasting plasma glucose: a genome-wide DNA methylation analysis in Chinese monozygotic twins. Diabetol Metab Syndr 2023; 15:159. [PMID: 37461060 PMCID: PMC10351111 DOI: 10.1186/s13098-023-01136-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Elevated fasting plasma glucose (FPG) levels can increase morbidity and mortality even when it is below the diagnostic threshold of type 2 diabetes mellitus (T2DM). We conducted a genome-wide DNA methylation analysis to detect DNA methylation (DNAm) variants potentially related to FPG in Chinese monozygotic twins. METHODS Genome-wide DNA methylation profiling in whole blood of twins was performed using Reduced Representation Bisulfite Sequencing (RRBS), yielding 551,447 raw CpGs. Association between DNAm of single CpG and FPG was tested using a generalized estimation equation. Differentially methylated regions (DMRs) were identified using comb-P approach. ICE FALCON method was utilized to perform the causal inference. Candidate CpGs were quantified and validated using Sequenom MassARRAY platform in a community population. Weighted gene co-expression network analysis (WGCNA) was conducted using gene expression data from twins. RESULTS The mean age of 52 twin pairs was 52 years (SD: 7). The relationship between DNAm of 142 CpGs and FPG reached the genome-wide significance level. Thirty-two DMRs within 24 genes were identified, including TLCD1, MRPS31P5, CASZ1, and CXADRP3. The causal relationship of top CpGs mapped to TLCD1, MZF1, PTPRN2, SLC6A18, ASTN2, IQCA1, GRIN1, and PDE2A genes with FPG were further identified using ICE FALCON method. Pathways potentially related to FPG were also identified, such as phospholipid-hydroperoxide glutathione peroxidase activity and mitogen-activated protein kinase p38 binding. Three CpGs mapped to SLC6A18 gene were validated in a community population, with a hypermethylated direction in diabetic patients. The expression levels of 18 genes (including SLC6A18 and TLCD1) were positively correlated with FPG levels. CONCLUSIONS We detect many DNAm variants that may be associated with FPG in whole blood, particularly the loci within SLC6A18 gene. Our findings provide important reference for the epigenetic regulation of elevated FPG levels and diabetes.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, Qingdao, 266071 Shandong Province China
| | - Wenqin Yao
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, Qingdao, 266071 Shandong Province China
- Shandong Province Center for Disease Control and Prevention, Shandong, China
| | - Qihua Tan
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Shuxia Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, No. 308 Ningxia Road, Qingdao, 266071 Shandong Province China
| |
Collapse
|
10
|
Chaoul V, Hanna R, Hachem P, El Hayek MS, Nour‐Eldine W, Abou‐Khalil P, Abi‐Ramia E, Vandecasteele G, Abi‐Gerges A. Differential changes in cyclic adenosine 3′‐5′ monophosphate (
cAMP
) effectors and major Ca
2+
handling proteins during diabetic cardiomyopathy. J Cell Mol Med 2023; 27:1277-1289. [PMID: 36967707 PMCID: PMC10148055 DOI: 10.1111/jcmm.17733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is associated with differential and time-specific regulation of β-adrenergic receptors and cardiac cyclic nucleotide phosphodiesterases with consequences for total cyclic adenosine 3'-5' monophosphate (cAMP) levels. We aimed to investigate whether these changes are associated with downstream impairments in cAMP and Ca2+ signalling in a type 1 diabetes (T1D)-induced DCM model. T1D was induced in adult male rats by streptozotocin (65 mg/kg) injection. DCM was assessed by cardiac structural and molecular remodelling. We delineated sequential changes affecting the exchange protein (Epac1/2), cAMP-dependent protein kinase A (PKA) and Ca2+ /Calmodulin-dependent kinase II (CaMKII) at 4, 8 and 12 weeks following diabetes, by real-time quantitative PCR and western blot. Expression of Ca2+ ATPase pump (SERCA2a), phospholamban (PLB) and Troponin I (TnI) was also examined. Early upregulation of Epac1 transcripts was noted in diabetic hearts at Week 4, followed by increases in Epac2 mRNA, but not protein levels, at Week 12. Expression of PKA subunits (RI, RIIα and Cα) remained unchanged regardless of the disease stage, whereas CaMKII increased at Week 12 in DCM. Moreover, PLB transcripts were upregulated in diabetic hearts, whereas SERCA2a and TnI gene expression was unchanged irrespective of the disease evolution. PLB phosphorylation at threonine-17 was increased in DCM, whereas phosphorylation of both PLB at serine-16 and TnI at serine-23/24 was unchanged. We show for the first time differential and time-specific regulations in cardiac cAMP effectors and Ca2+ handling proteins, data that may prove useful in proposing new therapeutic approaches in T1D-induced DCM.
Collapse
Affiliation(s)
- Victoria Chaoul
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| | - Rita Hanna
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| | - Pia Hachem
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| | - Magali Samia El Hayek
- Signaling and Cardiovascular Pathophysiology, UMR‐S1180Université Paris‐SaclayOrsay91400France
| | - Wared Nour‐Eldine
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| | - Pamela Abou‐Khalil
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| | - Elias Abi‐Ramia
- School of Arts and Sciences, Department of Natural SciencesLebanese American UniversityByblosLebanon
| | - Grégoire Vandecasteele
- Signaling and Cardiovascular Pathophysiology, UMR‐S1180Université Paris‐SaclayOrsay91400France
| | - Aniella Abi‐Gerges
- Gilbert and Rose‐Marie Chagoury School of MedicineLebanese American UniversityP.O. Box 36ByblosLebanon
| |
Collapse
|
11
|
Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiac hypertrophy and heart failure. Nat Rev Cardiol 2023; 20:90-108. [PMID: 36050457 DOI: 10.1038/s41569-022-00756-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate the neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple PDE isozymes with different enzymatic properties and subcellular localization regulate local pools of cyclic nucleotides and specific functions. This organization is heavily perturbed during cardiac hypertrophy and heart failure (HF), which can contribute to disease progression. Clinically, PDE inhibition has been considered a promising approach to compensate for the catecholamine desensitization that accompanies HF. Although PDE3 inhibitors, such as milrinone or enoximone, have been used clinically to improve systolic function and alleviate the symptoms of acute HF, their chronic use has proved to be detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as new potential targets to treat HF, each having a unique role in local cyclic nucleotide signalling pathways. In this Review, we describe cAMP and cGMP signalling in cardiomyocytes and present the various PDE families expressed in the heart as well as their modifications in pathological cardiac hypertrophy and HF. We also appraise the evidence from preclinical models as well as clinical data pointing to the use of inhibitors or activators of specific PDEs that could have therapeutic potential in HF.
Collapse
|
12
|
Phosphodiesterase 5a Signalling in Skeletal Muscle Pathophysiology. Int J Mol Sci 2022; 24:ijms24010703. [PMID: 36614143 PMCID: PMC9820699 DOI: 10.3390/ijms24010703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Phosphodiesterase 5A (PDE5A) is involved in cGMP hydrolysis, regulating many physiological processes. Increased activity of PDE5A has been found in several pathological conditions, and the pharmacological inhibition of PDE5 has been demonstrated to have several therapeutic applications. We have identified the presence of three different Pde5a isoforms in cardiomyocytes, and we have found that the expression of specific Pde5a isoforms may have a causal role in the onset of pathological responses in these cells. In our previous study, we demonstrated that PDE5A inhibition could ameliorate muscular dystrophy by acting at different levels, as assessed by the altered genomic response of muscular cells following treatment with the PDE5A inhibitor tadalafil. Thus, considering the importance of PDE5A in various pathophysiological conditions, we further investigated the regulation of this enzyme. Here, we analysed the expression of Pde5a isoforms in the pathophysiology of skeletal muscle. We found that skeletal muscle tissues and myogenic cells express Pde5a1 and Pde5a2 isoforms, and we observed an increased expression of Pde5a1 in damaged skeletal muscles, while Pde5a2 levels remained unchanged. We also cloned and characterized the promoters that control the transcription of Pde5a isoforms, investigating which of the transcription factors predicted by bioinformatics analysis could be involved in their modulation. In conclusion, we found an overexpression of Pde5a1 in compromised muscle and identified an involvement of MyoD and Runx1 in Pde5a1 transcriptional activity.
Collapse
|
13
|
Cao N, Li X, Zhang W, Wang Q, Liang Y, Zhou F, Xiao X. Research progress of signaling pathways of the natural substances intervene dyslipidemia (Review). Exp Ther Med 2022; 24:494. [PMID: 35813312 PMCID: PMC9257764 DOI: 10.3892/etm.2022.11421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/25/2022] [Indexed: 11/27/2022] Open
Abstract
Dyslipidemia is an umbrella term for a range of lipid metabolic disorders in the body. This condition has been widely reported to greatly increase the risk of cardiovascular diseases, threatening human health. In recent years, advances in molecular biology have deepened understanding of the dyslipidemia-related signaling pathways and specific mechanisms underlying dyslipidemia. Signaling pathways possess the ability to transmit an extracellular signal to the inside of the cell, leading to specific biological effects. Lipid metabolism disorders and lipid levels in the blood are frequently affected by aberrant alterations in the dyslipidemia-related signaling pathways. Therefore, further investigations into these pathways are required for the prevention and treatment of dyslipidemia. The present review summarizes the characteristics of six dyslipidemia-associated signaling pathways: Peroxisome proliferator-activated receptor, adenosine monophosphate-activated protein kinase, farnesoid X receptor, forkhead box O, adipocytokine and cyclic adenosine monophosphate signaling pathways. In particular, specific focus was placed on previous experimental studies and reports on the intervention effects of natural substances (compounds from animals, plants, marine organisms and microorganisms) on dyslipidemia.
Collapse
Affiliation(s)
- Ningning Cao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| | - Xiaoxuan Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| | - Wanjing Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| | - Qingguo Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| | - Yujuan Liang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| | - Fujun Zhou
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Binhai, Tianjin 300301, P.R. China
| | - Xuefeng Xiao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Jinghai, Tianjin 301617, P.R. China
| |
Collapse
|
14
|
Nour-Eldine W, Sayyed K, Harhous Z, Dagher-Hamalian C, Mehanna S, Achkouti D, ElKazzaz H, Khnayzer RS, Kobeissy F, Khalil C, Abi-Gerges A. Gasoline fume inhalation induces apoptosis, inflammation, and favors Th2 polarization in C57BL/6 mice. J Appl Toxicol 2022; 42:1178-1191. [PMID: 35001415 DOI: 10.1002/jat.4286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/11/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Abstract
Gasoline exposure has been widely reported in the literature as being toxic to human health. However, the exact underlying molecular mechanisms triggered by its inhalation have not been thoroughly investigated. We herein present a model of sub-chronic, static gasoline vapor inhalation in adult female C57BL/6 mice. Animals were exposed daily to either gasoline vapors (0.86 g/animal/90 minutes) or ambient air for five days/week over seven consecutive weeks. At the end of the study period, toxic and molecular mechanisms, underlying the inflammatory, oxidative, and apoptotic effects triggered by gasoline vapors, were examined in the lungs and liver of gasoline exposed mice. Static gasoline exposure induced a significant increase (+21 %) in lungs/body weight ratio in gasoline-exposed (GE) versus control (CON) mice along with a pulmonary inflammation attested by histological staining. The latter was consistent with increases in the transcript levels of proinflammatory cytokines [Interleukins (ILs) 4 and 6], respectively by ~ 6-, 4-fold in the lungs of GE mice compared to CON. Interestingly, IL-10 expression was also increased by ~ 10-fold in the lungs of GE mice suggesting an attempt to counterbalance the established inflammation. Moreover, the pulmonary expression of IL-12 and TNF-α was downregulated by 2- and 4-fold, respectively, suggesting the skewing toward Th2 phenotype. Additionally, GE mice showed a significant upregulation in Bax/Bcl-2 ratio, caspases 3, 8 and 9 with no change in JNK expression in the lungs, suggesting the activation of both intrinsic and extrinsic apoptotic pathways. Static gasoline exposure over seven consecutive weeks had a minor hepatic portal inflammation attested by H&E staining along with an increase in the hepatic expression of the mitochondrial complexes in GE mice. Therefore, tissue damage biomarkers highlight the health risks associated with vapor exposure and may present potential therapeutic targets for recovery from gasoline intoxication.
Collapse
Affiliation(s)
- Wared Nour-Eldine
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Katia Sayyed
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Lebanon
| | - Zeina Harhous
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Carole Dagher-Hamalian
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Stephanie Mehanna
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut, Lebanon
| | - Donna Achkouti
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Hanan ElKazzaz
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Rony S Khnayzer
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut, Lebanon
| | - Firas Kobeissy
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Christian Khalil
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos, Lebanon
| | - Aniella Abi-Gerges
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
15
|
Peng M, Liu H, Ji Q, Ma P, Niu Y, Ning S, Sun H, Pang X, Yang Y, Zhang Y, Han J, Hao G. Fufang Xueshuantong Improves Diabetic Cardiomyopathy by Regulating the Wnt/ β-Catenin Pathway. Int J Endocrinol 2022; 2022:3919161. [PMID: 36237833 PMCID: PMC9553353 DOI: 10.1155/2022/3919161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/02/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is one of the main complications of diabetic patients and the major reason for the high prevalence of heart failure in diabetic patients. Fufang Xueshuantong (FXST) is a traditional Chinese medicine formula commonly used in the treatment of diabetic retinopathy and stable angina pectoris. However, the role of FXST in DCM has not yet been clarified. This study was conducted to investigate the effects of FXST on diabetic myocardial lesions and reveal its molecular mechanism. The rats were intraperitoneally injected with 65 mg/kg streptozotocin (STZ) to induce diabetes mellitus (DM). DM rats were given saline or FXST. The rats in the control group were intraperitoneally injected with an equal amount of sodium citrate buffer and gavaged with saline. After 12 weeks, echocardiography, heart weight index (HWI), and myocardial pathological changes were determined. The expression of transforming growth factor-beta1 (TGF-β1), collagen I, and collagen III was examined using immunofluorescence staining and western blot. The expressions of Wnt/β-catenin signaling pathway-related proteins and mRNA were detected by western blot and real-time PCR. The results showed that FXST significantly improved cardiac function, ameliorated histopathological changes, and decreased HWI in the DM rats. FXST significantly inhibited the expression of myocardial TGF-β1, collagen I, and collagen III in DM rats. Furthermore, FXST significantly inhibited the Wnt/β-catenin pathway. Taken together, FXST has a protective effect on DCM, which might be mediated by suppressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Meizhong Peng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hanying Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingxuan Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pan Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiting Niu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shangqiu Ning
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huihui Sun
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinxin Pang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqian Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Zhang
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Han
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Gansu Provincial Hospital of Traditional Chinese Medicine, Gansu, China
| |
Collapse
|