101
|
Zuo H, Qu Q, Tong Y, Wang L, Wang X, Wu S, Zhou M. Electroacupuncture alleviates acute myocardial ischemic injury in mice by regulating the β 1 adrenergic receptor and post-receptor protein kinase A signaling pathway. Acupunct Med 2024; 42:342-355. [PMID: 39579035 PMCID: PMC11633077 DOI: 10.1177/09645284241298716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/25/2024]
Abstract
OBJECTIVE To determine the effect of electroacupuncture (EA) on β1-adrenergic receptor (β1-AR) and post-receptor protein kinase A (PKA) signaling pathway after acute myocardial ischemia (MI). METHODS An MI model was established by ligating the left anterior descending coronary artery of wild-type (WT) C57/BL and β1-AR+/- mice (heterozygous for β1-AR gene deletion). EA treatment was administered at HT5-HT7 or LU9-LU8. We evaluated cardiac function by measuring ST segment displacement, ischemic area and serum levels of creatine kinase (CK)-MB and lactate dehydrogenase (LDH). Pathological morphology/apoptosis of myocardial tissue were examined using hematoxylin-eosin and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Norepinephrine (NE) levels in myocardial tissue were detected by ELISA. Levels of β1 and post-receptor PKA signaling components were evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting. RESULTS EA stimulation at HT7-HT5 could better regulate the level of β1-AR in myocardial tissue than that at LU9-LU8. Following EA, the ST segment, serum CK-MB/ LDH and area of myocardial infarction were decreased in WT mice, and the degree of myocardial pathology/apoptosis and expression of cleaved caspase-3 were decreased. Myocardial levels of Gs protein (Gs), adenylate cyclase (AC), cyclic adenosine monophosphate (cAMP), phosphorylated protein kinase A (p-PKA), L-type voltage-gated calcium channel α1C (Cav1.2), serine phosphate 16-phospholamban (p-PLBs16) and sarcoplasmic reticulum Ca2+-adenosine triphosphate (ATP)ase 2a (SERCA2a) increased after EA. However, these effects of EA were not replicated in β1-AR+/- mice. Interestingly, myocardial NE content decreased after EA in WT and β1-AR+/- mice. CONCLUSION EA may enhance cardiac function and reduced MI area/apoptosis by restoring the activity of β1-AR and post-receptor PKA signaling.
Collapse
Affiliation(s)
- Haiyan Zuo
- Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Anhui University of Chinese Medicine, Hefei, China
| | - Qiaoyu Qu
- Anhui Sanlian University, Hefei, China
| | - Yan Tong
- Xinwu District Hospital of Traditional Chinese Medicine, Wuxi, China
| | - Lei Wang
- Changfeng Hospital of Traditional Chinese Medicine, Hefei, China
| | - Xiaoxiao Wang
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Shengbing Wu
- Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Anhui University of Chinese Medicine, Hefei, China
| | - Meiqi Zhou
- Institute of Acupuncture and Meridian, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Meridian Viscera Correlationship, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
102
|
Wu XQ, Zhao L, Zhao YL, He XY, Zou L, Zhao YY, Li X. Traditional Chinese medicine improved diabetic kidney disease through targeting gut microbiota. PHARMACEUTICAL BIOLOGY 2024; 62:423-435. [PMID: 38757785 PMCID: PMC11104709 DOI: 10.1080/13880209.2024.2351946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT Diabetic kidney disease (DKD) affects nearly 40% of diabetic patients, often leading to end-stage renal disease that requires renal replacement therapies, such as dialysis and transplantation. The gut microbiota, an integral aspect of human evolution, plays a crucial role in this condition. Traditional Chinese medicine (TCM) has shown promising outcomes in ameliorating DKD by addressing the gut microbiota. OBJECTIVE This review elucidates the modifications in gut microbiota observed in DKD and explores the impact of TCM interventions on correcting microbial dysregulation. METHODS We searched relevant articles from databases including Web of Science, PubMed, ScienceDirect, Wiley, and Springer Nature. The following keywords were used: diabetic kidney disease, diabetic nephropathy, gut microbiota, natural product, TCM, Chinese herbal medicine, and Chinese medicinal herbs. Rigorous criteria were applied to identify high-quality studies on TCM interventions against DKD. RESULTS Dysregulation of the gut microbiota, including Lactobacillus, Streptococcus, and Clostridium, has been observed in individuals with DKD. Key indicators of microbial dysregulation include increased uremic solutes and decreased short-chain fatty acids. Various TCM therapies, such as formulas, tablets, granules, capsules, and decoctions, exhibit unique advantages in regulating the disordered microbiota to treat DKD. CONCLUSION This review highlights the importance of targeting the gut-kidney axis to regulate microbial disorders, their metabolites, and associated signaling pathways in DKD. The Qing-Re-Xiao-Zheng formula, the Shenyan Kangfu tablet, the Huangkui capsule, and the Bekhogainsam decoction are potential candidates to address the gut-kidney axis. TCM interventions offer a significant therapeutic approach by targeting microbial dysregulation in patients with DKD.
Collapse
Affiliation(s)
- Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Lei Zhao
- Department of General Practice, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Yan-Long Zhao
- Faculty of Life Science & Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Xin-Yao He
- Faculty of Life Science & Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, Chengdu, Sichuan, China
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, Xi’an, Shaanxi, China
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xia Li
- Faculty of Life Science & Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of General Practice, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
103
|
Tang MY, Xie H, Tao JT, Zhang C, Luo YH, Zhang C, Peng SQ, Xie LX, Lv WB, Zhang C, Huang L. Pathophysiological relevance and therapeutic outlook of GPR43 in atherosclerosis. Biochem Cell Biol 2024; 102:418-429. [PMID: 39013204 DOI: 10.1139/bcb-2024-0053] [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] [Indexed: 07/18/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory arterial disorder that occurs due to the deposition of the excessive lipoprotein under the artery intima, mainly including low-density lipoprotein and other apolipoprotein B-containing lipoproteins. G protein-coupled receptors (GPCRs) play a crucial role in transmitting signals in physiological and pathophysiological conditions. GPCRs recognize inflammatory mediators, thereby serving as important players during chronic inflammatory processes. It has been demonstrated that free fatty acids can function as ligands for various GPCRs, such as free fatty acid receptor (FFAR)1/GPR40, FFAR2/GPR43, FFAR3/GPR41, FFAR4/GPR120, and the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). This review discusses GPR43 and its ligands in the pathogenesis of AS, especially focusing on its distinct role in regulating chronic vascular inflammation, inhibiting oxidative stress, ameliorating endothelial dysfunction and improving dyslipidemia. It is hoped that this review may provide guidance for further studies aimed at GPR43 as a promising target for drug development in the prevention and therapy of AS.
Collapse
Affiliation(s)
- Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hao Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jin-Tao Tao
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yao-Hua Luo
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Si-Qin Peng
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Xi Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Wen-Bo Lv
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
104
|
Dongdem JT, Etornam AE, Beletaa S, Alidu I, Kotey H, Wezena CA. The β 3-Adrenergic Receptor: Structure, Physiopathology of Disease, and Emerging Therapeutic Potential. Adv Pharmacol Pharm Sci 2024; 2024:2005589. [PMID: 39640497 PMCID: PMC11620816 DOI: 10.1155/2024/2005589] [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: 05/25/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
The discovery and characterization of the signal cascades of the β-adrenergic receptors have made it possible to effectively target the receptors for drug development. β-Adrenergic receptors are a class A rhodopsin type of G protein-coupled receptors (GPCRs) that are stimulated mainly by catecholamines and therefore mediate diverse effects of the parasympathetic nervous system in eliciting "fight or flight" type responses. They are detectable in several human tissues where they control a plethora of physiological processes and therefore contribute to the pathogenesis of several disease conditions. Given the relevance of the β-adrenergic receptor as a molecular target for many pathological conditions, this comprehensive review aims at providing an in-depth exploration of the recent advancements in β3-adrenergic receptor research. More importantly, we delve into the prospects of the β3-adrenergic receptor as a therapeutic target across a variety of clinical domains.
Collapse
Affiliation(s)
- Julius T. Dongdem
- Department of Chemical Pathology, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Axandrah E. Etornam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Solomon Beletaa
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Issah Alidu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Hassan Kotey
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Northern Region, Ghana
| | - Cletus A. Wezena
- Department of Microbiology, Faculty of Biosciences, University for Development Studies, Tamale, Northern Region, Ghana
| |
Collapse
|
105
|
Rivera K, Gonzalez L, Bravo L, Manjarres L, Andia ME. The Gut-Heart Axis: Molecular Perspectives and Implications for Myocardial Infarction. Int J Mol Sci 2024; 25:12465. [PMID: 39596530 PMCID: PMC11595032 DOI: 10.3390/ijms252212465] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Myocardial infarction (MI) remains the leading cause of death globally, imposing a significant burden on healthcare systems and patients. The gut-heart axis, a bidirectional network connecting gut health to cardiovascular outcomes, has recently emerged as a critical factor in MI pathophysiology. Disruptions in this axis, including gut dysbiosis and compromised intestinal barrier integrity, lead to systemic inflammation driven by gut-derived metabolites like lipopolysaccharides (LPSs) and trimethylamine N-oxide (TMAO), both of which exacerbate MI progression. In contrast, metabolites such as short-chain fatty acids (SCFAs) from a balanced microbiota exhibit protective effects against cardiac damage. This review examines the molecular mediators of the gut-heart axis, considering the role of factors like sex-specific hormones, aging, diet, physical activity, and alcohol consumption on gut health and MI outcomes. Additionally, we highlight therapeutic approaches, including dietary interventions, personalized probiotics, and exercise regimens. Addressing the gut-heart axis holds promise for reducing MI risk and improving recovery, positioning it as a novel target in cardiovascular therapy.
Collapse
Affiliation(s)
- Katherine Rivera
- Doctoral Program in Medical Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 8331010, Chile;
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Leticia Gonzalez
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Liena Bravo
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Laura Manjarres
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Marcelo E. Andia
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| |
Collapse
|
106
|
Donati Zeppa S, Gervasi M, Bartolacci A, Ferrini F, Patti A, Sestili P, Stocchi V, Agostini D. Targeting the Gut Microbiota for Prevention and Management of Type 2 Diabetes. Nutrients 2024; 16:3951. [PMID: 39599740 PMCID: PMC11597803 DOI: 10.3390/nu16223951] [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: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder with a heterogeneous etiology encompassing societal and behavioral risk factors in addition to genetic and environmental susceptibility. The cardiovascular consequences of diabetes account for more than two-thirds of mortality among people with T2D. Not only does T2D shorten life expectancy, but it also lowers quality of life and is associated with extremely high health expenditures since diabetic complications raise both direct and indirect healthcare costs. An increasing body of research indicates a connection between T2D and gut microbial traits, as numerous alterations in the intestinal microorganisms have been noted in pre-diabetic and diabetic individuals. These include pro-inflammatory bacterial patterns, increased intestinal permeability, endotoxemia, and hyperglycemia-favoring conditions, such as the alteration of glucagon-like peptide-1 (GLP-1) secretion. Restoring microbial homeostasis can be very beneficial for preventing and co-treating T2D and improving antidiabetic therapy outcomes. This review summarizes the characteristics of a "diabetic" microbiota and the metabolites produced by microbial species that can worsen or ameliorate T2D risk and progression, suggesting gut microbiota-targeted strategies to restore eubiosis and regulate blood glucose. Nutritional supplementation, diet, and physical exercise are known to play important roles in T2D, and here their effects on the gut microbiota are discussed, suggesting non-pharmacological approaches that can greatly help in diabetes management and highlighting the importance of tailoring treatments to individual needs.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
- Department of Human Science for Promotion of Quality of Life, University San Raffaele, 00166 Rome, Italy;
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Antonino Patti
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90144 Palermo, Italy;
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, University San Raffaele, 00166 Rome, Italy;
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (S.D.Z.); (A.B.); (P.S.); (D.A.)
| |
Collapse
|
107
|
Xue Y, Lin S, Chen M, Ke J, Zhang J, Fan Q, Chen Y, Chen F. Altered colonic microflora and its metabolic profile in mice with acute viral myocarditis induced by coxsackievirus B3. Virol J 2024; 21:295. [PMID: 39550578 PMCID: PMC11568606 DOI: 10.1186/s12985-024-02571-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 11/05/2024] [Indexed: 11/18/2024] Open
Abstract
Mounting evidence suggests that the gut-heart axis is critical in the pathogenesis of cardiovascular diseases. The gut serves as the primary pathway through which Coxsackievirus B3 (CVB3) infects its host, leading to acute viral myocarditis (AVMC). However, little is known about the role of gut microflora and its metabolites in the development of AVMC. The AVMC model was established by intraperitoneal injection of CVB3 in mice. Then, 16S ribosomal RNA (16S rRNA) gene sequencing and ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) untargeted metabolomics profiling were performed to analyze the microflora composition and metabolic profile of colonic contents. Compared to the Control mice, the AVMC mice displayed a significant reduction in gut microflora richness and diversity, as revealed by an increased abundance of Proteobacteria and a decreased abundance of Cyanobacteria and Desulfobacterota. LEfSe analysis indicated that the main genera differing between the two groups were Escherichia-Shigella, Lactobacillus, Clostridium_sensu_stricto_1, Prevotellaceae_UCG-001, and Odoribacter. Based on the criterion of OPLS-DA VIP ≥ 1.0 and p-value < 0.05, a total of 198 differential metabolites (DMs) were identified in the gut, including 79 upregulated and 119 downregulated metabolites, of which lipids and lipid-like molecules accounted for the largest proportion. Notably, both altered gut bacterial taxa and metabolites were significantly enriched in the Lipid metabolism pathway, with Traumatic acid (TA), Alpha-Linolenic acid (ALA), Eicosapentaenoic acid (EPA), and Docosahexaenoic acid (DHA) being the key DMs in the pathway. Additionally, significant positive correlations (|r| > 0.80 and p < 0.05) were found between TA levels and Anaerotruncus and Bilophila abundance, between EPA levels and Clostridium_sensu_stricto_1 abundance, and between DHA levels and Escherichia-Shigella abundance, respectively. CVB3 infection leads to notable alterations in gut microflora composition and its metabolic profile, which may participate in AVMC development. Our findings provide important clues for future in-depth studies on AVMC etiology.
Collapse
Affiliation(s)
- Yimin Xue
- Fourth Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Shirong Lin
- Department of Emergency, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Mingguang Chen
- Fourth Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Jun Ke
- Department of Emergency, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Jiuyun Zhang
- Department of Emergency, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Qiaolian Fan
- Fourth Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Yimei Chen
- Department of Emergency, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China
| | - Feng Chen
- Department of Emergency, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
108
|
Ferreira Alves G, Oliveira JG, Nakashima MA, Delfrate G, Sordi R, Assreuy J, da Silva-Santos JE, Collino M, Fernandes D. Cardiovascular effects of Roflumilast during sepsis: Risks or benefits? Eur J Pharmacol 2024; 983:177015. [PMID: 39332796 DOI: 10.1016/j.ejphar.2024.177015] [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: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Phosphodiesterase-4 (PDE4) is responsible for terminating cyclic adenosine monophosphate (cAMP) signalling. PDE4 inhibitors, such as roflumilast (RFM), have anti-inflammatory activity and have been studied in inflammation-induced tissue damage in sepsis. However, the role of RFM on cardiovascular derangements induced by sepsis is still unknown. Thus, we aimed to evaluate the potential effects of RFM on cardiovascular collapse and multiorgan damage caused by sepsis. METHODS Sepsis was induced by cecal ligation and puncture (CLP) in male rats. Six hours after the CLP or sham procedure, animals were randomly assigned to receive either RFM (0.3 mg/kg) or vehicle subcutaneously, and cardiovascular parameters were assessed 24 h after the surgery and organ/plasma samples were collected for further analyses. RESULTS Sepsis induced hypotension, tachycardia, reduced renal blood flow (RBF) and hyporeactivity to vasoconstrictors both in vivo and ex vivo. RFM treatment increased systemic cAMP levels and RBF. RFM also attenuated hypoperfusion and liver damage induced by CLP. Furthermore, RFM reduced systemic nitric oxide (NO) levels in septic rats, while there were no changes in hepatic NOS-2 expression. Nevertheless, RFM exacerbated sepsis-induced hypotension and tachycardia without ameliorating vascular hyporeactivity. CONCLUSION Our data show that PDE-4 inhibition protects septic rats from hepatic injury and improves renal perfusion. However, RFM worsened hemodynamic parameters and showed no protection against sepsis-induced cardiovascular dysfunction and mortality. Thus, despite the anti-inflammatory benefits of RFM, its application in sepsis should be approached cautiously.
Collapse
Affiliation(s)
- Gustavo Ferreira Alves
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | | | | | - Gabrielle Delfrate
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Regina Sordi
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Jamil Assreuy
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Massimo Collino
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Daniel Fernandes
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil.
| |
Collapse
|
109
|
Li L, Jin L, You L, Liu Q, Yan L. The association of preoperative serum free fatty acid levels with survival in breast cancer patients. Discov Oncol 2024; 15:629. [PMID: 39511004 PMCID: PMC11543952 DOI: 10.1007/s12672-024-01499-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Serum free fatty acids (FFA) are associated with various types of cancer. However, the prognostic value of preoperative serum FFA levels and breast cancer (BC) remains unclear. This study aimed to elucidate the specific relationship between FFA levels and BC outcomes. METHODS A retrospective review was conducted on 4133 patients with BC admitted to Sun Yat-sen Memorial Hospital from January 2015 to October 2021. Preoperative serum FFA levels were detected by the enzymatic endpoint method. The relationship between serum FFA levels and clinical characteristics was analyzed based on FFA interquartile range. Restricted cubic splines and multivariate Cox regression analyses were used to assess the relationship between preoperative serum FFA levels and overall survival (OS) in patients with BC. The hazard ratios (HRs) and 95% confidence intervals (95% CIs) were calculated. RESULTS According to the FFA interquartile range, FFA levels were significantly correlated with OS (years) (p < 0.001). Restricted cubic spline analysis might reveal a U-shaped relationship between preoperative serum FFA levels and OS, after adjusting for other variables. According to the cutoff points for FFAs, multivariate Cox regression analyses showed that patients with low FFA levels (≤ 250 µmol/L) had higher rates of all-cause mortality and cancer-specific mortality than those with high FFA levels (530-700 µmol/L) in the total population and patients with a BMI of 18.5-24.0 kg/m2. A trend was observed indicating that elevated FFA levels (≥ 715 µmol/L) were associated with worse prognosis; however, this association failed to reach statistical significance. CONCLUSIONS There might be a nonlinear U-shaped relationship between preoperative serum FFA levels and survival in breast cancer patients, with lower FFA levels associated with worse OS. The effect of elevated FFA levels on prognosis requires further investigation.
Collapse
Affiliation(s)
- Liuran Li
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou key laboratory for Metabolic Diseases, Guangzhou, Guangdong, China
| | - Liang Jin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lili You
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou key laboratory for Metabolic Diseases, Guangzhou, Guangdong, China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Clinical Research Center for Metabolic Diseases, Guangzhou key laboratory for Metabolic Diseases, Guangzhou, Guangdong, China.
| |
Collapse
|
110
|
Tóth AD, Turu G, Hunyady L. Functional consequences of spatial, temporal and ligand bias of G protein-coupled receptors. Nat Rev Nephrol 2024; 20:722-741. [PMID: 39039165 DOI: 10.1038/s41581-024-00869-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/24/2024]
Abstract
G protein-coupled receptors (GPCRs) regulate every aspect of kidney function by mediating the effects of various endogenous and exogenous substances. A key concept in GPCR function is biased signalling, whereby certain ligands may selectively activate specific pathways within the receptor's signalling repertoire. For example, different agonists may induce biased signalling by stabilizing distinct active receptor conformations - a concept that is supported by advances in structural biology. However, the processes underlying functional selectivity in receptor signalling are extremely complex, involving differences in subcellular compartmentalization and signalling dynamics. Importantly, the molecular mechanisms of spatiotemporal bias, particularly its connection to ligand binding kinetics, have been detailed for GPCRs critical to kidney function, such as the AT1 angiotensin receptor (AT1R), V2 vasopressin receptor (V2R) and the parathyroid hormone 1 receptor (PTH1R). This expanding insight into the multifaceted nature of biased signalling paves the way for innovative strategies for targeting GPCR functions; the development of novel biased agonists may represent advanced pharmacotherapeutic approaches to the treatment of kidney diseases and related systemic conditions, such as hypertension, diabetes and heart failure.
Collapse
MESH Headings
- Humans
- Ligands
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/physiology
- Receptors, Vasopressin/metabolism
- Receptors, Vasopressin/physiology
- Animals
- Receptor, Parathyroid Hormone, Type 1/metabolism
- Receptor, Parathyroid Hormone, Type 1/physiology
- Kidney Diseases/metabolism
- Kidney/metabolism
Collapse
Affiliation(s)
- András D Tóth
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Gábor Turu
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - László Hunyady
- Institute of Molecular Life Sciences, Centre of Excellence of the Hungarian Academy of Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
111
|
Singh H, Nair A, Mahajan SD. Impact of genetic variations of gene involved in regulation of metabolism, inflammation and coagulation on pathogenesis of cardiac injuries associated with COVID-19. Pathol Res Pract 2024; 263:155608. [PMID: 39447244 DOI: 10.1016/j.prp.2024.155608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/29/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND SARS-CoV-2 infection can result in long-term chronic cardiovascular (CV) damage after the acute phase of the illness. COVID-19 frequently causes active myocarditis, SARS-CoV-2 can directly infect and kill cardiac cells, causing severe pathology and dysfunction across the organs and cells. Till now, the pathogenesis of COVID-19-associated cardiac injuries has not been understood, but there are several factors that contribute to the progression of cardiac injuries, such as genetic, dietary, and environmental. Among them ranges of host genetic factor including metabolizing, inflammation, and coagulation related genes have a role to contribute the cardiac injuries induced by COVID-19. Hereditary DNA sequence variations contribute to the risk of illness in almost all of these diseases. Hence, we comprehended the occurrence of genetic variations of metabolizing, inflammation and coagulation-related genes in the general population, their expression in various diseases, and their impact on cardiac injuries induced by COVID-19. METHOD We utilized multiple databases, including PubMed (Medline), EMBASE, and Google Scholar, for literature searches. DESCRIPTION The genes involved in metabolism (APOE, MTHFR), coagulation (PAI-1, ACE2), and immune factors (CRP, ESR, and troponin I) may have a role in the progression of COVID-19-associated cardiac injuries. The risk factors for CVD are significantly varied between and within different regions. In healthy individuals, the ACE I allele is responsible for the predisposition to CAD, but the ACE D haplotype is responsible for susceptibility and severity, which ultimately leads to heart failure. Patients who carry the T allele of rs12329760 in the TMPRSS2 gene are at risk for developing the severe form of COVID-19. IL-6 (rs1800796/rs1800795) polymorphism is associated with an increased mortality rate and susceptibility to severe COVID-19 disease. While the putative role of IL-6 associated with chronic, inflammatory diseases like cardiac and cerebrovascular disease is well known. CONCLUSION The occurrence of genetic variations in the ACE-2, AGT, DPP-IV, TMPRSS2, FUIRN, IL-4, IL-6, IFN-γ, and CYP2D6 genes is varied among different populations. Examining the correlation between these variations and their protein levels and cardiac injuries induced by COVID-19 may provide valuable insights into the pathogenesis of cardiac injuries induced by COVID-19.
Collapse
Affiliation(s)
- HariOm Singh
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India.
| | - Aishwarya Nair
- Department of Molecular Biology, National AIDS Research Institute, Pune 411026, India
| | - Supriya D Mahajan
- Department of Medicine, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo's Clinical Translational Research Center, 875 Ellicott Street, Buffalo, NY 14203, USA
| |
Collapse
|
112
|
Ni D, Lin X, Deng C, Yuan L, Li J, Liu Y, Liang P, Jiang B. Energy metabolism: from physiological changes to targets in sepsis-induced cardiomyopathy. Hellenic J Cardiol 2024; 80:96-106. [PMID: 38734307 DOI: 10.1016/j.hjc.2024.05.010] [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: 01/17/2024] [Revised: 03/07/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by a variety of dysregulated responses to host infection with life-threatening multi-organ dysfunction. Among the injuries or dysfunctions involved in the course of sepsis, cardiac injury and dysfunction often occur and are associated with the pathogenesis of hemodynamic disturbances, also defined as sepsis-induced cardiomyopathy (SIC). The process of myocardial metabolism is tightly regulated and adapts to various cardiac output demands. The heart is a metabolically flexible organ capable of utilizing all classes of energy substrates, including carbohydrates, lipids, amino acids, and ketone bodies, to produce ATP. The demand of cardiac cells for energy metabolism changes substantially in septic cardiomyopathy, with distinct etiological causes and different times. This review describes changes in cardiomyocyte energy metabolism under normal physiological conditions and some features of myocardial energy metabolism in septic cardiomyopathy and briefly outlines the role of the mitochondria as a center of energy metabolism in the septic myocardium, revealing that changes in energy metabolism can serve as a potential future therapy for infectious cardiomyopathy.
Collapse
Affiliation(s)
- Dan Ni
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Xiaofang Lin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Chuanhuan Deng
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Ludong Yuan
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Jing Li
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Yuxuan Liu
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bimei Jiang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
113
|
Wang X, Nie X, Wang H, Ren Z. Roles of small GTPases in cardiac hypertrophy (Review). Mol Med Rep 2024; 30:208. [PMID: 39301654 PMCID: PMC11425065 DOI: 10.3892/mmr.2024.13332] [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: 04/10/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Cardiac hypertrophy results from the heart reacting and adapting to various pathological stimuli and its persistent development is a major contributing factor to heart failure. However, the molecular mechanisms of cardiac hypertrophy remain unclear. Small GTPases in the Ras, Rho, Rab, Arf and Ran subfamilies exhibit GTPase activity and play crucial roles in regulating various cellular responses. Previous studies have shown that Ras, Rho and Rab are closely linked to cardiac hypertrophy and that their overexpression can induce cardiac hypertrophy. Here, we review the functions of small GTPases in cardiac hypertrophy and provide additional insights and references for the prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xin Wang
- School of Mathematics and Statistics, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xinwen Nie
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hao Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
114
|
Torres DB, Lopes A, Rodrigues AJ, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA, Lopes MG. Early morphological and neurochemical changes of the bed nucleus of stria terminalis (BNST) in gestational protein-restricted male offspring. Nutr Neurosci 2024; 27:1250-1268. [PMID: 38576309 DOI: 10.1080/1028415x.2024.2320498] [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] [Indexed: 04/06/2024]
Abstract
BACKGROUND The bed nucleus of the stria terminalis (BNST) is a structure with a peculiar neurochemical composition involved in modulating anxietylike behavior and fear. AIM The present study investigated the effects on the BNST neurochemical composition and neuronal structure in critical moments of the postnatal period in gestational protein-restricted male rats' offspring. METHODS Dams were maintained during the pregnancy on isocaloric rodent laboratory chow with standard protein content [NP, 17%] or low protein content [LP, 6%]. BNST from male NP and age-matched LP offspring was studied using the isotropic fractionator method, Neuronal 3D reconstruction, dendritic-tree analysis, blotting analysis, and high-performance liquid chromatography. RESULTS Serum corticosterone levels were higher in male LP offspring than NP rats in 14-day-old offspring, without any difference in 7-day-old progeny. The BNST total cell number and anterodorsal BNST division volume in LP progeny were significantly reduced on the 14th postnatal day compared with NP offspring. The BNST HPLC analysis from 7 days-old LP revealed increased norepinephrine levels compared to NP progeny. The BNST blot analysis from 7-day-old LP revealed reduced levels of GR and BDNF associated with enhanced CRF1 expression compared to NP offspring. 14-day-old LP offspring showed reduced expression of MR and 5HT1A associated with decreased DOPAC and DOPA turnover levels relative to NP rats. In Conclusion, the BNST cellular and neurochemical changes may represent adaptation during development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition alters the BNST content and structure and contributes to already-known behavioral changes.
Collapse
Affiliation(s)
- D B Torres
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A Lopes
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J A R Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - P A Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | | |
Collapse
|
115
|
Stoicovy RA, Cora N, Perez A, Nagliya D, Del Calvo G, Lopez TB, Weinstein EC, Borges JI, Maning J, Lymperopoulos A. Cyclic adenosine monophosphate critically modulates cardiac GLP-1 receptor's anti-inflammatory effects. Inflamm Res 2024; 73:2043-2056. [PMID: 39305297 DOI: 10.1007/s00011-024-01950-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Glucagon-like peptide (GLP)-1 receptor (GLP1R) agonists exert a multitude of beneficial cardiovascular effects beyond control of blood glucose levels and obesity reduction. They also have anti-inflammatory actions through both central and peripheral mechanisms. GLP1R is a G protein-coupled receptor (GPCR), coupling to adenylyl cyclase (AC)-stimulatory Gs proteins to raise cyclic 3`-5`-adenosine monophosphate (cAMP) levels in cells. cAMP exerts various anti-apoptotic and anti-inflammatory effects via its effectors protein kinase A (PKA) and Exchange protein directly activated by cAMP (Epac). However, the precise role and importance of cAMP in mediating GLP1R`s anti-inflammatory actions, at least in the heart, remains to be determined. To this end, we tested the effects of the GLP1R agonist liraglutide on lipopolysaccharide (LPS)-induced acute inflammatory injury in H9c2 cardiac cells, either in the absence of cAMP production (AC inhibition) or upon enhancement of cAMP levels via phosphodiesterase (PDE)-4 inhibition with roflumilast. METHODS & RESULTS Liraglutide dose-dependently inhibited LPS-induced apoptosis and increased cAMP levels in H9c2 cells, with roflumilast but also PDE8 inhibition further enhancing cAMP production by liraglutide. GLP1R-stimulated cAMP markedly suppressed the LPS-dependent induction of pro-inflammatory tumor necrosis factor (TNF)-a, interleukin (IL)-1b, and IL-6 cytokine expression, of inducible nitric oxide synthase (iNOS) expression and nuclear factor (NF)-kB activity, of matrix metalloproteinases (MMP)-2 and MMP-9 levels and activities, and of myocardial injury markers in H9c2 cardiac cells. The effects of liraglutide were mediated by the GLP1R since they were abolished by the GLP1R antagonist exendin(9-39). Importantly, AC inhibition completely abrogated liraglutide`s suppression of LPS-dependent inflammatory injury, whereas roflumilast significantly enhanced the protective effects of liraglutide against LPS-induced inflammation. Finally, PKA inhibition or Epac1/2 inhibition alone only partially blocked liraglutide`s suppression of LPS-induced inflammation in H9c2 cardiac cells, but, together, PKA and Epac1/2 inhibition fully prevented liraglutide from reducing LPS-dependent inflammation. CONCLUSIONS cAMP, via activation of both PKA and Epac, is essential for GLP1R`s anti-inflammatory signaling in cardiac cells and that cAMP levels crucially regulate the anti-inflammatory efficacy of GLP1R agonists in the heart. Strategies that elevate cardiac cAMP levels, such as PDE4 inhibition, may potentiate the cardiovascular, including anti-inflammatory, benefits of GLP1R agonist drugs.
Collapse
Affiliation(s)
- Renee A Stoicovy
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Natalie Cora
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Arianna Perez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Deepika Nagliya
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Giselle Del Calvo
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Teresa Baggio Lopez
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Emma C Weinstein
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Jordana I Borges
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
| | - Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328-2018, USA.
- , University Dr., HPD (Terry) Bldg./Room 1350, Fort Lauderdale, FL, 33328-2018, USA.
| |
Collapse
|
116
|
Kovačević M, Paudel A, Planinšek O, Bertoni S, Passerini N, Zupančič O, Alva C, German Ilić I, Zvonar Pobirk A. The comparison of melt technologies based on mesoporous carriers for improved carvedilol dissolution. Eur J Pharm Sci 2024; 202:106880. [PMID: 39181171 DOI: 10.1016/j.ejps.2024.106880] [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: 04/25/2024] [Revised: 07/10/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
High-shear (HS) melt granulation and hot melt extrusion (HME) were compared as perspective melt-based technologies for preparation of amorphous solid dispersions (ASDs). ASDs were prepared using mesoporous carriers (SyloidⓇ 244FP or NeusilinⓇ US2), which were loaded with carvedilol dispersed in polymeric matrix (polyethylene glycol 6000 or SoluplusⓇ). Formulations with high carvedilol content were obtained either by HME (11 extrudates with polymer:carrier ratio 1:1) or HS granulation (6 granulates with polymer:carrier ratio 3:1). DSC and XRD analysis confirmed the absence of crystalline carvedilol for the majority of prepared ADSs, thus confirming the stabilizing effect of selected polymers and carriers over amorphous carvedilol. HME produced larger particles compared to HS melt granulation, which was in line with better flow time and Carr index of extrudates. Moreover, SEM images revealed smoother surface of ASDs obtained by HME, contributing to less obstructed flow. The rougher and more porous surface of HS granules was correlated to larger granule specific surface area, manifesting in faster carvedilol release from SyloidⓇ 244FP-based granules, as compared to their HME counterparts. Regarding dissolution, the two HS-formulations performed superior to pure crystalline carvedilol, thereby confirming the suitability of HS melt granulation for developing dosage forms with improved carvedilol dissolution.
Collapse
Affiliation(s)
- Mila Kovačević
- University of Ljubljana Faculty of Pharmacy, Department of Pharmaceutical Technology, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Amrit Paudel
- Research Centre for Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Odon Planinšek
- University of Ljubljana Faculty of Pharmacy, Department of Pharmaceutical Technology, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Serena Bertoni
- University of Bologna, Department of Pharmacy and BioTechnology, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Nadia Passerini
- University of Bologna, Department of Pharmacy and BioTechnology, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Ožbej Zupančič
- Research Centre for Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Carolina Alva
- Research Centre for Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Ilija German Ilić
- University of Ljubljana Faculty of Pharmacy, Department of Pharmaceutical Technology, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| | - Alenka Zvonar Pobirk
- University of Ljubljana Faculty of Pharmacy, Department of Pharmaceutical Technology, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
117
|
O'Rorke J, Butler WG, Mason K. Complex Management of Respiratory Failure, Atrial Fibrillation, Ventricular Tachycardia, and Stenotrophomonas maltophilia in a Patient Following Osteomyelitis Amputation: A Case of Multisystem Complications Occurring Postoperatively. Cureus 2024; 16:e73505. [PMID: 39677254 PMCID: PMC11642728 DOI: 10.7759/cureus.73505] [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: 10/01/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Stenotrophomonas maltophilia is an opportunistic, multidrug-resistant gram-negative bacterium often affecting patients with significant comorbidities. This case report examines the hospital course of a 75-year-old male with a history of atrial fibrillation and heart failure with preserved ejection fraction (HFpEF), who presented with compromised respiratory status and recurrent infections, highlighting the complexities of clinical management in the setting of multidrug-resistant HFpEF organisms and postoperative complications. The patient was admitted following an episode of ventricular tachycardia and acute respiratory failure, requiring rapid airway management and intensive clinical intervention. His recent hospitalization for sepsis, pneumonia, and osteomyelitis complicated his clinical profile, particularly given his recurrent urinary tract infections (UTIs), which prevented the use of sodium-glucose cotransporter-2 inhibitor therapy for heart failure management. Respiratory cultures confirmed the presence of S. maltophilia, prompting treatment with minocycline and piperacillin-tazobactam. This case highlights the significant risks associated with postoperative arrhythmias in patients with underlying cardiac disease, particularly when superimposed with sepsis. Furthermore, the management of recurrent UTIs posed a barrier to optimizing heart failure therapy, further complicating the patient's clinical stability. The need for vigilant monitoring and tailored therapeutic strategies is essential to improve outcomes in this vulnerable patient population. The interplay between multidrug-resistant infections, arrhythmias, and comorbidities emphasizes the importance of comprehensive clinical management and the need for further research to develop targeted therapies and clinical plans for at-risk populations.
Collapse
Affiliation(s)
- Jesse O'Rorke
- Medicine, Lee Health, Fort Myers, USA
- Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, USA
| | - W Greyson Butler
- Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, USA
| | - Keri Mason
- Internal Medicine, Cape Coral Hospital, Fort Myers, USA
| |
Collapse
|
118
|
Xu Y, He Z, Rao Z, Li Z, Hu Y, Zhang Z, Zhou J, Zhou T, Wang H. The role of β2-AR/PI3K/AKT pathway in the proliferation, migration and invasion of THLE-2 cells induced by nicotine. Toxicology 2024; 508:153924. [PMID: 39147091 DOI: 10.1016/j.tox.2024.153924] [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: 05/31/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Nicotine, the primary constituent of tobacco, is one of the important factors that induce the occurrence of hepatocellular carcinoma (HCC). The β2-adrenergic receptor (β2-AR) is implicated in the growth and advancement of tumors. However, the role of β2-AR and its mediated cascades in nicotine-induced HCC remains unclear. This present study aims to observe the effects of nicotine on the proliferation, migration, and invasion of immortalized human liver epithelial (THLE-2) cells, as well as to explore the underlying mechanisms of action. The results of cell counting kit-8 (CCK-8) assay showed that 0.3125 μM nicotine had the ability to promote the proliferation of THLE-2 cells with a significant time-dependent manner. Therefore, THLE-2 cells were mainly selected for chronic treatment with 0.3125 μM nicotine in the later stage to cause transformation. After 30 passages of THLE-2 cells with 0.3125 μM nicotine treatment, chronic exposure to nicotine significantly enhanced the proliferation, metastasis, and invasion of cells. Besides, it also upregulated the intracellular levels of β2-AR, phosphoinositide 3-kinase (PI3K), AKT, matrix metalloproteinase-2 (MMP-2) and Cyclin D1, as well as downregulated the expression of p53. More importantly, the β2-AR/PI3K/AKT pathway was found to mediate the expression of MMP-2, Cyclin D1, and p53 in THLE-2 cells, playing a crucial role in their proliferation, migration, and invasion after continuous exposure to nicotine. Simply put, it demonstrated the role of β2-AR/PI3K/AKT pathway in the transformation of THLE-2 cells induced by nicotine. This study could provide valuable insights into the relationship between nicotine and HCC. Additionally, it lays the groundwork for investigating potential anticancer treatments for liver cancer linked to tobacco consumption.
Collapse
Affiliation(s)
- Yuqin Xu
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zihan He
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zihan Rao
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zihan Li
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Yuxin Hu
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Zhongwei Zhang
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Jianming Zhou
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Tong Zhou
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China
| | - Huai Wang
- School of Public Health, Jiangxi Medical College, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, No. 461 Ba Yi Avenue, Nanchang, Jiangxi 330006, PR China; Chongqing Research Institute of Nanchang University, Tai Bai Road, Tongnan, Chongqing 402679, PR China.
| |
Collapse
|
119
|
Zhou Z, Wang Y, Xing Y, Pan S, Wang W, Yang J, Wu W, Zhou J, Huang L, Liang Q, Zhang D, Kong L. Magnolol Inhibits High Fructose-Induced Podocyte Inflammation via Downregulation of TKFC/Sp1/HDAC4/Notch1 Activation. Pharmaceuticals (Basel) 2024; 17:1416. [PMID: 39598328 PMCID: PMC11597211 DOI: 10.3390/ph17111416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/06/2024] [Accepted: 10/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES High fructose has been implicated as an important trigger of kidney inflammation in patients and experimental models. Magnolol, isolated from Magnolia officinalis, has an anti-inflammatory effect, but its protective role in podocytes remains underexplored. This study explored the protective effects and underlying mechanism of magnolol against high fructose-induced podocyte inflammation. METHODS The effects of magnolol on high fructose-induced podocyte inflammation were assessed in male Sprague Dawley rats administered 10% (w/v) fructose water for 12 weeks and heat-sensitive human podocyte cell lines (HPCs) exposed to 5 mM fructose. Podocyte foot processes were examined using transmission electron microscopy. The expression levels of nephrin, podocin, tumor necrosis factor-α (TNF-α), Notch1 intracellular domain (NICD1), triokinase/FMN cyclase (TKFC), specificity protein 1 (Sp1) and histone deacetylase 4 (HDAC4) were determined by Western blot, immunofluorescence and real-time quantitative polymerase chain reaction (qRT-PCR). The chromatin immunoprecipitation (ChIP) assay was performed to evaluate the interaction between Sp1 and the promoter region of HDAC4. RESULTS Magnolol mitigated the impairment of glomerular filtration function in high fructose-fed rats. Besides, it significantly alleviated the inflammatory responses in glomeruli and HPCs, evidenced by decreased protein levels of TNF-α and NICD1. Increased protein levels of TKFC, Sp1 and HDAC4 were observed in high fructose-stimulated HPCs and rat glomeruli. TMP195, an HDAC4 inhibitor, reduced TNF-α and NICD1 protein levels in high fructose-exposed HPCs. The increased Sp1 was shown to associate with the promoter region of HDAC4, promoting HDAC4 protein expression in high fructose-exposed HPCs. The knockdown of TKFC in HPCs by TKFC siRNA decreased Sp1, HDAC4 and NICD1 protein levels, alleviating podocyte inflammatory response. Furthermore, magnolol inhibited TKFC/Sp1/HDAC4/Notch1 activation in vivo and in vitro. CONCLUSIONS Magnolol attenuated high fructose-induced podocyte inflammation possibly through the suppression of TKFC/Sp1/HDAC4/Notch1 activation, providing new evidence for its potential role in podocyte protection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Dongmei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China; (Z.Z.); (Y.W.); (Y.X.); (S.P.); (W.W.); (J.Y.); (W.W.); (J.Z.); (L.H.); (Q.L.)
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Chinese Medicine, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China; (Z.Z.); (Y.W.); (Y.X.); (S.P.); (W.W.); (J.Y.); (W.W.); (J.Z.); (L.H.); (Q.L.)
| |
Collapse
|
120
|
Chaudhary S, Kaur P, Singh TA, Bano KS, Vyas A, Mishra AK, Singh P, Mehdi MM. The dynamic crosslinking between gut microbiota and inflammation during aging: reviewing the nutritional and hormetic approaches against dysbiosis and inflammaging. Biogerontology 2024; 26:1. [PMID: 39441393 DOI: 10.1007/s10522-024-10146-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The early-life gut microbiota (GM) is increasingly recognized for its contributions to human health and disease over time. Microbiota composition, influenced by factors like race, geography, lifestyle, and individual differences, is subject to change. The GM serves dual roles, defending against pathogens and shaping the host immune system. Disruptions in microbial composition can lead to immune dysregulation, impacting defense mechanisms. Additionally, GM aids digestion, releasing nutrients and influencing physiological systems like the liver, brain, and endocrine system through microbial metabolites. Dysbiosis disrupts intestinal homeostasis, contributing to age-related diseases. Recent studies are elucidating the bacterial species that characterize a healthy microbiota, defining what constitutes a 'healthy' colonic microbiota. The present review article focuses on the importance of microbiome composition for the development of homeostasis and the roles of GM during aging and the age-related diseases caused by the alteration in gut microbial communities. This article might also help the readers to find treatments targeting GM for the prevention of various diseases linked to it effectively.
Collapse
Affiliation(s)
- Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Pardeep Kaur
- Department of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Thokchom Arjun Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Kaniz Shahar Bano
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
121
|
Wu J, Ai T, He P, Shi Q, Li Y, Zhang Z, Chen M, Huang Z, Wu S, Chen W, Han J. Cecal necroptosis triggers lethal cardiac dysfunction in TNF-induced severe SIRS. Cell Rep 2024; 43:114778. [PMID: 39325617 DOI: 10.1016/j.celrep.2024.114778] [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: 03/11/2024] [Revised: 07/25/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
Tumor necrosis factor (TNF) induces systemic inflammatory response syndrome (SIRS), and severe SIRS can serve as a model for studying animal death caused by organ failure. Through strategic cecectomy, we demonstrate that necroptosis in the cecum initiates the death process in TNF-treated mice, but it is not the direct cause of death. Instead, we show that it is the cardiac dysfunction downstream of cecum damage that ultimately leads to the death of TNF-treated mice. By in vivo and ex vivo physiological analyses, we reveal that TNF and the damage-associated molecular patterns (DAMPs) released from necroptotic cecal cells jointly target cardiac endothelial cells, triggering caspase-8 activation and subsequent cardiac endothelial damage. Cardiac endothelial damage is a primary cause of the deterioration of diastolic function in the heart of TNF-treated mice. Our research provides insights into the pathophysiological process of TNF-induced lethality.
Collapse
Affiliation(s)
- Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Ai
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Peng He
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Qilin Shi
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Yangxin Li
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China
| | - Ziguan Zhang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Minwei Chen
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Zhengrong Huang
- Xiamen Key Laboratory of Cardiac Electrophysiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361102, China
| | - Suqin Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanze Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518000, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian 361102, China; Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian 361102, China; Research Unit of Cellular Stress of Chinese Academy of Medical Sciences, Xiang'an Hospital of Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
122
|
Maluleke TT, Manilall A, Shezi N, Baijnath S, Millen AME. Acute exposure to LPS induces cardiac dysfunction via the activation of the NLRP3 inflammasome. Sci Rep 2024; 14:24378. [PMID: 39420211 PMCID: PMC11487256 DOI: 10.1038/s41598-024-76066-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] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic inflammation contributes to left ventricular (LV) dysfunction, however the role of the NLRP3 inflammasome in LV dysfunction in acute inflammatory conditions is unclear. This study investigated the role of the NLRP3 inflammasome in acute (24 h) cardiac structural and functional changes in vivo and in vitro in lipopolysaccharide (LPS)-induced inflammation. LPS-treated Sprague-Dawley (SD) rats showed increased LPS metabolite abundance in their LVs as measured by atmospheric pressure matrix-assisted laser desorption ionisation (AP-MALDI) mass spectrometry imaging (MSI). Echocardiography and histology showed that in LPS-exposed rats, LV internal diameter was decreased, with evidence of macrophage infiltration and oedema. However, there were no changes in LV wall thickness or collagen volume. Additionally, LPS-exposed rats exhibited impaired LV relaxation, potentially contributing to decreased stroke volume. While global systolic function was preserved, LPS exposure in SD rats resulted in impaired myocardial deformation assessed by speckle-tracking echocardiography. Exposure to LPS resulted in upregulation of the expression of components of the NLRP3 inflammasome in rodents. In vitro LPS exposure resulted in increased gene expression of NLRP3 and downstream cytokines IL-1β and IL-18, antioxidant SOD2, and elevated markers of pyroptosis (GSDMD) which were inhibited by treatment with a NLRP3 antagonist. However, LPS-induced increases in the gene expression of apoptosic markers (BAX/Bcl2) were not impacted by NLRP3 antagonism. These findings suggest that inflammation induced adverse cardiac structural and functional changes is, at least in part, mediated by the NLRP3 inflammasome in acute, high-grade inflammatory states. In addition, in vitro findings suggest that while the NLRP3 inflammasome mediates pyroptotic pathways, regulation of apoptosis that is independent of the inflammasome.
Collapse
Affiliation(s)
- Tshiamo T Maluleke
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Ashmeetha Manilall
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Nandi Shezi
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Sooraj Baijnath
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa
| | - Aletta M E Millen
- Wits Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, Wits Health Consortium (PTY) Ltd, University of The Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
123
|
Hu H, Xu Z, Zhang Z, Song P, Stull F, Xu P, Tang H. Rational design of a flavoenzyme for aerobic nicotine catabolism. mBio 2024; 15:e0205024. [PMID: 39191403 PMCID: PMC11481913 DOI: 10.1128/mbio.02050-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Enzymatic therapy with nicotine-degrading enzyme is a new strategy in treating nicotine addiction, which can reduce nicotine concentrations and weaken withdrawal in the rat model. However, when O2 is used as the electron acceptor, no satisfactory performance has been achieved with one of the most commonly studied and efficient nicotine-catabolizing enzymes, NicA2. To obtain more efficient nicotine-degrading enzyme, we rationally designed and engineered a flavoenzyme Pnao, which shares high structural similarity with NicA2 (RMSD = 1.143 Å) and efficiently catalyze pseudooxynicotine into 3-succinoyl-semialdehyde pyridine using O2. Through amino acid alterations with NicA2, five Pnao mutants were generated, which can degrade nicotine in Tris-HCl buffer and retain catabolic activity on its natural substrate. Nicotine-1'-N-oxide was identified as one of the reaction products. Four of the derivative mutants showed activity in rat serum and Trp220 and Asn224 were found critical for enzyme specificity. Our findings offer a novel avenue for research into aerobic nicotine catabolism and provide a promising method of generating additional nicotine-catalytic enzymes. IMPORTANCE Nicotine, the main active substance in tobacco, results in cigarette addiction and various diseases. There have been some attempts at using nicotine oxidoreductase, NicA2, as a therapeutic for nicotine cessation. However, it uses cytochrome c as it is electron acceptor, which is impractical for therapeutic use compared with using O2 as an oxidant. Thus, amino acid alteration was performed on Pnao using NicA2 as model. Five of the mutants generated degraded nicotine at a rate similar to NicA2, and one of the catabolic compounds was identified as nicotine-1'-N-oxide. Our research highlights a new direction in developing enzymes that efficiently catabolize nicotine without co-enzymes and suggests that structure-similar human original MAOA (or B) may assist with nicotine cessation after being engineered.
Collapse
Affiliation(s)
- Haiyang Hu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoyong Xu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyao Zhang
- Department of Chemistry, Western Michigan University, Kalamazoo, Michigan, USA
| | - Peizhi Song
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Frederick Stull
- Department of Chemistry, Western Michigan University, Kalamazoo, Michigan, USA
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hongzhi Tang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
124
|
Lymperopoulos A, Stoicovy RA. RGS Proteins in Sympathetic Nervous System Regulation: Focus on Adrenal RGS4. FRONT BIOSCI-LANDMRK 2024; 29:355. [PMID: 39473413 DOI: 10.31083/j.fbl2910355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 01/11/2025]
Abstract
The sympathetic nervous system (SNS) consists largely of two different types of components: neurons that release the neurotransmitter norepinephrine (NE, noradrenaline) to modulate homeostasis of the innevrvated effector organ or tissue and adrenal chromaffin cells, which synthesize and secrete the hormone epinephrine (Epi, adrenaline) and some NE into the blood circulation to act at distant organs and tissues that are not directly innervated by the SNS. Like almost every physiological process in the human body, G protein-coupled receptors (GPCRs) tightly modulate both NE release from sympathetic neuronal terminals and catecholamine (CA) secretion from the adrenal medulla. Regulator of G protein Signaling (RGS) proteins, acting as guanosine triphosphatase (GTPase)-activating proteins (GAPs) for the Gα subunits of heterotrimeric guanine nucleotide-binding proteins (G proteins), play a central role in silencing G protein signaling from a plethora of GPCRs. Certain RGS proteins and, in particular, RGS4, have been implicated in regulation of SNS activity and of adrenal chromaffin cell CA secretion. More specifically, recent studies have implicated RGS4 in regulation of NE release from cardiac sympathetic neurons by means of terminating free fatty acid receptor (FFAR)-3 calcium signaling and in regulation of NE and Epi secretion from the adrenal medulla by means of terminating cholinergic calcium signaling in adrenal chromaffin cells. Thus, in this review, we provide an overview of the current literature on the involvement of RGS proteins, with a particular focus on RGS4, in these two processes, i.e., NE release from sympathetic nerve terminals & CA secretion from adrenal chromaffin cells. We also highlight the therapeutic potential of RGS4 pharmacological manipulation for diseases characterized by sympathetic dysfunction or SNS hyperactivity, such as heart failure and hypertension.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Department of Pharmaceutical Sciences, Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Davie/Fort Lauderdale, FL 33328-2018, USA
| | - Renee A Stoicovy
- Department of Pharmaceutical Sciences, Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University Barry and Judy Silverman College of Pharmacy, Davie/Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
125
|
Parichatikanond W, Duangrat R, Kurose H, Mangmool S. Regulation of β-Adrenergic Receptors in the Heart: A Review on Emerging Therapeutic Strategies for Heart Failure. Cells 2024; 13:1674. [PMID: 39451192 PMCID: PMC11506672 DOI: 10.3390/cells13201674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
The prolonged overstimulation of β-adrenergic receptors (β-ARs), a member of the G protein-coupled receptor (GPCR) family, causes abnormalities in the density and functionality of the receptor and contributes to cardiac dysfunctions, leading to the development and progression of heart diseases, especially heart failure (HF). Despite recent advancements in HF therapy, mortality and morbidity rates continue to be high. Treatment with β-AR antagonists (β-blockers) has improved clinical outcomes and reduced overall hospitalization and mortality rates. However, several barriers in the management of HF remain, providing opportunities to develop new strategies that focus on the functions and signal transduction of β-ARs involved in the pathogenesis of HF. As β-AR can signal through multiple pathways influenced by different receptor subtypes, expression levels, and signaling components such as G proteins, G protein-coupled receptor kinases (GRKs), β-arrestins, and downstream effectors, it presents a complex mechanism that could be targeted in HF management. In this narrative review, we focus on the regulation of β-ARs at the receptor, G protein, and effector loci, as well as their signal transductions in the physiology and pathophysiology of the heart. The discovery of potential ligands for β-AR that activate cardioprotective pathways while limiting off-target signaling is promising for the treatment of HF. However, applying findings from preclinical animal models to human patients faces several challenges, including species differences, the genetic variability of β-ARs, and the complexity and heterogeneity of humans. In this review, we also summarize recent updates and future research on the regulation of β-ARs in the molecular basis of HF and highlight potential therapeutic strategies for HF.
Collapse
Affiliation(s)
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Hitoshi Kurose
- Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan;
- Pharmacology for Life Sciences, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Supachoke Mangmool
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
126
|
Amicone S, Impellizzeri A, Tattilo FP, Ryabenko K, Asta C, Belà R, Suma N, Canton L, Fedele D, Bertolini D, Bodega F, Cavallo D, Marinelli V, Ciarlantini M, Pastore G, Iuorio OD, Alvarez MC, Bavuso LL, Salerno J, Vasumini N, Maida A, Armillotta M, Angeli F, Sansonetti A, Bergamaschi L, Foà A, Squeri A, Dall'Ara G, Pizzi C. Noninvasive Assessment in Takotsubo Syndrome: A Diagnostic Challenge. Echocardiography 2024; 41:e15953. [PMID: 39387111 DOI: 10.1111/echo.15953] [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: 08/21/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Takotsubo syndrome (TTS) is a clinical syndrome characterized by a transient left ventricular dysfunction whose diagnosis can be challenging due to its resemblance to acute myocardial infarction (AMI). Despite the growing recognition of TTS, acute complications and long-term mortality rates are comparable to those observed in AMI patients. In this context, a systematic diagnostic approach is imperative for an accurate patient assessment, with due consideration of the distinctive characteristics and optimal timing of each imaging modality. Coronary angiography with ventriculography may be reserved for cases presenting with ST-segment elevation, whereas in all other cases, the use of multimodality noninvasive imaging allows for a comprehensive evaluation of typical diagnostic features and detection of acute complications while also providing prognostic insights. The aim of this review is to evaluate the current research on non-invasive modalities and to propose a diagnostic algorithm that will facilitate the identification and management of TTS.
Collapse
Affiliation(s)
- Sara Amicone
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Andrea Impellizzeri
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Francesco Pio Tattilo
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Khrystyna Ryabenko
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Claudio Asta
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Rebecca Belà
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Nicole Suma
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Lisa Canton
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Damiano Fedele
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Davide Bertolini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Francesca Bodega
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Daniele Cavallo
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Virginia Marinelli
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Mariachiara Ciarlantini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giuseppe Pastore
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Ornella Di Iuorio
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Marcello Casuso Alvarez
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Leonardo Luca Bavuso
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Jessica Salerno
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Nicolò Vasumini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Angelo Maida
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Matteo Armillotta
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Francesco Angeli
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Angelo Sansonetti
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Luca Bergamaschi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alberto Foà
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Angelo Squeri
- Department of Cardiology, Maria Cecilia Hospital, GVM Care and Research, Cotignola (RA), Italy
| | - Gianni Dall'Ara
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Cardiology Unit, Morgagni Pierantoni Hospital, Forlì, Italy
| | - Carmine Pizzi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences - DIMEC; Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
127
|
Xu Z, Li M, Lyu D, Xiao H, Li S, Li Z, Li M, Xiao J, Huang H. Cinnamaldehyde activates AMPK/PGC-1α pathway via targeting GRK2 to ameliorate heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155894. [PMID: 39089090 DOI: 10.1016/j.phymed.2024.155894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/25/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND According to recent research, treating heart failure (HF) by inhibiting G protein-coupled receptor kinase 2 (GRK2) to improve myocardial energy metabolism has been identified as a potential approach. Cinnamaldehyde (CIN), a phenylpropyl aldehyde compound, has been demonstrated to exhibit beneficial effects in cardiovascular diseases. However, whether CIN inhibits GRK2 to ameliorate myocardial energy metabolism in HF is still unclear. PURPOSE This study examines the effects of CIN on GRK2 and myocardial energy metabolism to elucidate its underlying mechanism to treat HF. METHODS The isoproterenol (ISO) induced HF model in vivo and in vitro were constructed using Sprague-Dawley (SD) rats and primary neonatal rat cardiomyocytes (NRCMs). Based on this, the effects of CIN on myocardial energy metabolism and GRK2 were investigated. Additionally, validation experiments were conducted after interfering and over-expressing GRK2 in ISO-induced NRCMs to verify the regulatory effect of CIN on GRK2. Furthermore, binding capacity between GRK2 and CIN was explored by Cellular Thermal Shift Assay (CETSA) and Microscale Thermophoresis (MST). RESULTS In vivo and in vitro, CIN significantly improved HF as demonstrated by reversing abnormal changes in myocardial injury markers, inhibiting myocardial hypertrophy and decreasing myocardial fibrosis. Additionally, CIN promoted myocardial fatty acid metabolism to ameliorate myocardial energy metabolism disorder by activating AMPK/PGC-1α signaling pathway. Moreover, CIN reversed the inhibition of myocardial fatty acid metabolism and AMPK/PGC-1α signaling pathway by GRK2 over-expression in ISO-induced NRCMs. Meanwhile, CIN had no better impact on the stimulation of cardiac fatty acid metabolism and the AMPK/PGC-1α signaling pathway in ISO-induced NRCMs when GRK2 was disrupted. Noticeably, CETSA and MST confirmed that CIN binds to GRK2 directly. The binding of CIN and GRK2 promoted the ubiquitination degradation of GRK2 mediated by murine double mimute 2. CONCLUSION This study demonstrates that CIN exerts a protective intervention in HF by targeting GRK2 and promoting its ubiquitination degradation to activate AMPK/PGC-1α signaling pathway, ultimately improving myocardial fatty acid metabolism.
Collapse
Affiliation(s)
- Zhanchi Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China
| | - Minghui Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongxin Lyu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuoming Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junhui Xiao
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| | - Heqing Huang
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| |
Collapse
|
128
|
Liao L, Wang T, Zhang L, Wei Y, Fan X. Protective Mechanisms of SGLTi in Ischemic Heart Disease. J Cardiovasc Transl Res 2024; 17:1018-1035. [PMID: 38767796 DOI: 10.1007/s12265-024-10513-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 05/22/2024]
Abstract
Ischemic heart disease (IHD) is a common clinical cardiovascular disease with high morbidity and mortality. Sodium glucose cotransporter protein inhibitor (SGLTi) is a novel hypoglycemic drug. To date, both clinical trials and animal experiments have shown that SGLTi play a protective role in IHD, including myocardial infarction (MI) and ischemia/reperfusion (I/R). The protective effects may be involved in mechanisms of energy metabolic conversion, anti-inflammation, anti-fibrosis, ionic homeostasis improvement, immune cell development, angiogenesis and functional regulation, gut microbiota regulation, and epicardial lipids. Thus, this review summarizes the above mechanisms and aims to provide theoretical evidence for therapeutic strategies for IHD.
Collapse
Affiliation(s)
- Lei Liao
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tong Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lu Zhang
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Xinrong Fan
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
129
|
Chen J, Zeng JC, Feng Y, Wei WT, Li SZ, Wu MD, Mo CJ, Huang JW, Yang CW, Hu SQ, Ni H. Carnosic acid, a novel food-source AT1R antagonist and its anti-hypertension mechanism. Int J Biol Macromol 2024; 278:135012. [PMID: 39181360 DOI: 10.1016/j.ijbiomac.2024.135012] [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: 06/21/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Hypertension is the most prevalent non-communicable disease, affecting billions of people worldwide. Discovery and development of natural antihypertensive lead compounds or drugs are important to resolve the limitations of existing antihypertensive drug safety and resistance. This investigation verified that carnosic acid (CA), an important active ingredient of rosemary, an edible spice plant, indicates a significant anti-hypertensive activity in spontaneous hypertension rats by targeting AT1R. Moreover, our research indicated that CA shared a comparable antagonistic mechanism with established synthetic angiotensin II receptor blockers (ARBs), as it occupies the binding sites of Angiotensin II (AngII) at His6 and Pro7 within the AT1R's ligand-binding pocket. Notably, CA exerted better anti-hypertensive activity since it could not break the Asn1113.35-Asn2957.46 hydrogen bond to stabilize the AT1R inactive state. As the first potent AT1R antagonist identified in a natural food source, CA is poised to become a novel anti-hypertensive lead compound, distinguished by its unique skeleton structure different from conventional ARBs. This research lays a valuable theoretical groundwork for the future exploration of CA and rosemary extract in both fundamental studies and clinical applications.
Collapse
Affiliation(s)
- Jie Chen
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jia-Cheng Zeng
- Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu Feng
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Wan-Ting Wei
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Shang-Ze Li
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Meng-Die Wu
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Chang-Jia Mo
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jun-Wen Huang
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Cheng-Wei Yang
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Song-Qing Hu
- School of Food Sciences and Engineering, South China University of Technology, Guangzhou 510641, China.
| | - He Ni
- Guangdong Provincial Key Lab of Biotechnology for Plant Development, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
130
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
131
|
Cuervo L, McAlpine PL, Olano C, Fernández J, Lombó F. Low-Molecular-Weight Compounds Produced by the Intestinal Microbiota and Cardiovascular Disease. Int J Mol Sci 2024; 25:10397. [PMID: 39408727 PMCID: PMC11477366 DOI: 10.3390/ijms251910397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is the main cause of mortality in industrialized countries, with over 500 million people affected worldwide. In this work, the roles of low-molecular-weight metabolites originating from the gut microbiome, such as short-chain fatty acids, hydrogen sulfide, trimethylamine, phenylacetic acid, secondary bile acids, indoles, different gases, neurotransmitters, vitamins, and complex lipids, are discussed in relation to their CVD-promoting or preventing activities. Molecules of mixed microbial and human hepatic origin, such as trimethylamine N-oxide and phenylacetylglutamine, are also presented. Finally, dietary agents with cardioprotective effects, such as probiotics, prebiotics, mono- and poly-unsaturated fatty acids, carotenoids, and polyphenols, are also discussed. A special emphasis is given to their gut microbiota-modulating properties.
Collapse
Affiliation(s)
- Lorena Cuervo
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Patrick L. McAlpine
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Research Group BIOMIC (Biosynthesis of Antitumor Molecules), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain; (L.C.); (C.O.)
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
| | - Javier Fernández
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Felipe Lombó
- IUOPA (Instituto Universitario de Oncología del Principado de Asturias), 33006 Oviedo, Spain
- ISPA (Instituto de Investigación Sanitaria del Principado de Asturias), 33006 Oviedo, Spain
- Research Group BIONUC (Biotechnology of Nutraceuticals and Bioactive Compounds), Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
132
|
Huang C, Zhao Y, Hu J. Endocrine-Disruptive Effects of Adenylate Cyclase Activator Forskolin: In Vitro and In Vivo Evidence. TOXICS 2024; 12:701. [PMID: 39453121 PMCID: PMC11510926 DOI: 10.3390/toxics12100701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/14/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
Forskolin (FSK) is a potent adenylate cyclase activator and may display endocrine-disruptive effects via the disruption of steroidogenesis. Here, we tested this hypothesis by use of the in vitro H295R steroidogenesis assay and the in vivo long-term medaka (Oryzias latipes) exposure assay. The results from the H295R assay demonstrated that the transcriptional levels of a series of genes involved in steroidogenesis, including HSD3B2, CYP11A, CYP11B2, CYP17, CYP19, and CYP21, were remarkably up-regulated. Meanwhile, the productions of estrogens (17β-estradiol (17β-E2) and estrone (E1)) and progestins (progesterone (PGT) and 17-hydroxyprogesterone (17-HPT)) were significantly increased, and those of androgens (androstenedione (ADD) and testosterone (TTR)) were significantly inhibited. After waterborne exposure of medaka to FSK for 100 days, the gene expressions of HMGR, HSD17B1, CYP17B, CYP19A, and CYP21A were significantly enhanced in the gonads of male medaka. 17β-E2 was remarkably induced, although without statistical significance. In addition, the biomarker genes for estrogenicity, including VTG-I, VTG-II, CHG-H, and CHG-L, were significantly induced in male medaka livers. Pathological damage to their gonads was further identified. Therefore, the results demonstrated that FSK modulates the transcriptions of steroidogenesis genes and alters hormone levels in vitro and in vivo, which is a mark of endocrine disruption in organisms.
Collapse
Affiliation(s)
- Chong Huang
- MOE Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China; (C.H.); (J.H.)
| | - Yanbin Zhao
- MOE Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China; (C.H.); (J.H.)
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianying Hu
- MOE Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China; (C.H.); (J.H.)
| |
Collapse
|
133
|
Yang ES, Jung JY, Kang CK. Regulation of Cerebral Blood Flow Velocity by Transcutaneous Electrical Nerve Stimulation: A Preliminary Study. Healthcare (Basel) 2024; 12:1908. [PMID: 39408088 PMCID: PMC11476227 DOI: 10.3390/healthcare12191908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
OBJECTIVES An excessive and abrupt increase in cerebral blood flow may cause blood vessel damage, leading to stroke. Therefore, appropriate methods to immediately regulate blood flow velocity are important. Through an analysis of 31 healthy adults, we therefore investigated whether stimulating the common carotid artery (CCA) using transcutaneous electrical nerve stimulation (TENS) could modulate blood flow velocity in the CCA. METHODS Three stimulation intensities (below-threshold, threshold, and above-threshold) were applied in a random order. Blood velocity changes were examined by the measurement of peak systolic velocity (PSV) with Doppler ultrasound before, during, and after TENS stimulation. To evaluate arterial stiffness, pulse wave velocity (PWV) was calculated using CCA diameter, and blood pressure was measured before and after stimulation. RESULTS PSV changes in the below-threshold level were significant (p = 0.028). The PSV after below-threshold stimulation was significantly decreased by 2.23% compared to that before stimulation (p = 0.031). PWV showed no significant differences; however, a nonsignificant increase was observed immediately after stimulation only in the above-threshold condition. Above-threshold stimulation can increase vascular tone by activating the sympathetic nerve, possibly triggering vasoconstriction. CONCLUSIONS A decrease in blood flow velocity may not be expected upon the above-threshold stimulation. In contrast, the below-threshold stimulation immediately reduces blood flow velocity, without significantly affecting hemodynamic function, such as arterial flexibility. Therefore, this short-term and low electrical stimulation technique can help to lower vascular resistance and prevent vascular damage from rapid blood flow velocity.
Collapse
Affiliation(s)
- Eun-Seon Yang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon 21936, Republic of Korea;
| | - Ju-Yeon Jung
- Institute for Human Health and Science Convergence, Gachon University, Incheon 21936, Republic of Korea
| | - Chang-Ki Kang
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon 21936, Republic of Korea;
- Institute for Human Health and Science Convergence, Gachon University, Incheon 21936, Republic of Korea
- Department of Radiological Science, College of Medical Science, Gachon University, Incheon 21936, Republic of Korea
| |
Collapse
|
134
|
Fu ZP, Lee S, Wang RY, Wang YQ. Cronobacter sakazakii induced sepsis-associated arrhythmias through its outer membrane vesicles. iScience 2024; 27:110572. [PMID: 39228788 PMCID: PMC11369384 DOI: 10.1016/j.isci.2024.110572] [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: 03/14/2024] [Revised: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
Sepsis-induced arrhythmia, linked to sudden cardiac death, is associated with gut microbiota, though the exact relationship is unclear. This study aimed to elucidate the relationship between Cronobacter sakazakii (C. sakazakii) and arrhythmia. The relative abundance of C. sakazakii was increased in cecal ligation and puncture (CLP)-induced septic mice. Live C. sakazakii, supernatant, and outer membrane vesicles (OMVs) resulted in premature ventricular beat (PVB), sinus arrhythmia (SA), and increased arrhythmia and mortality in sepsis model through dysregulated ion channel proteins. Moreover, short-chain fatty acids (SCFAs) showed antibacterial effects in vitro. We confirmed sodium acetate (C2) and sodium butyrate (C4) protect from C. sakazakii-induced arrhythmia, and C2 and C4 protected from septic arrhythmia by activating free fatty acid receptor 2 and 3 (FFAR2 and FFAR3) in mice. These findings point to how C. sakazakii's OMVs trigger arrhythmia, and SCFAs may be a treatment for septic arrhythmia.
Collapse
Affiliation(s)
- Zhi-ping Fu
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Shuang Lee
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Rui-yao Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yu-qing Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| |
Collapse
|
135
|
Liccardo D, Valletta A, Spagnuolo G, Vinciguerra C, Lauria MR, Perrotta A, Del Giudice C, De Luca F, Rengo G, Rengo S, Rengo C, Cannavo A. Porphyromonas gingivalis virulence factors induce toxic effects in SH-SY5Y neuroblastoma cells: GRK5 modulation as a protective strategy. J Biotechnol 2024; 393:7-16. [PMID: 39033880 DOI: 10.1016/j.jbiotec.2024.07.009] [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: 11/07/2023] [Revised: 07/02/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Periodontitis (PDS) is a chronic inflammatory disease initiated by a dysbiosis of oral pathogenic bacterial species, such as Porphyromonas gingivalis (Pg). These bacteria can penetrate the bloodstream, releasing various endo and exotoxins that fuel the infection, and stimulate toxic inflammation in different compartments, including the brain. However, the specific mechanisms by which PDS/Pg contribute to brain disorders, such as Alzheimer's disease (AD), remain unclear. This study assessed the effects of Pg's virulence factors - lipopolysaccharide (LPS-Pg) and gingipains (gps) K (Kgp) and Rgp - on SH-SY5Y cells. Our results demonstrated that LPS-Pg activated signaling through the Toll-like receptor (TLR)-2/4 induced a significant downregulation of G protein-coupled receptor kinase 5 (GRK5). Additionally, LPS-Pg stimulation resulted in a robust increase in Tau phosphorylation (pTau) and p53 levels, while causing a marked reduction in Bcl2 and increased cell death compared to unstimulated cells (Ns). LPS-Pg also elevated inducible nitric oxide synthase (iNOS) expression, leading to oxidative damage. In cells overexpressing GRK5 via Adenovirus, LPS-Pg failed to increase iNOS and pTau levels compared to GFP control cells. High GRK5 levels also prevented the nuclear accumulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Furthermore, the overexpression of a GRK5 mutant form lacking the nuclear localization signal (ΔNLS) nearly abolished LPS-Pg induced p53 and iNOS upregulation. Finally, we tested whether Kgp and Rgp mediated similar effects and our data showed that both gps caused a marked downregulation of GRK5 leading to increased p53 and pTau levels. In conclusion, this study provides further insight into the toxic effects elicited by Pg in cells and suggests that preventing GRK5 deficiency may be a valid strategy to mitigate Pg-induced toxic effects (i.e. cell death, oxidative damage, and Tau hyperphosphorylation) in SH-SY5Y cells, which are typical molecular hallmarks of neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniela Liccardo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Alessandra Valletta
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Gianrico Spagnuolo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Caterina Vinciguerra
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Maria Rosaria Lauria
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Alessia Perrotta
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Carmela Del Giudice
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Francesca De Luca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy; Istituti Clinici Scientifici Maugeri IRCCS - Scientific Institute of Telese Terme (BN), Italy
| | - Sandro Rengo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples 80131, Italy
| | - Carlo Rengo
- Dental School of Periodontology, University of Naples Federico II, Napoli 80127, Italy.
| | - Alessandro Cannavo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples 80131, Italy.
| |
Collapse
|
136
|
Wang J, Xue H, He J, Deng L, Tian J, Jiang Y, Feng J. Therapeutic potential of finerenone for diabetic cardiomyopathy: focus on the mechanisms. Diabetol Metab Syndr 2024; 16:232. [PMID: 39289758 PMCID: PMC11409712 DOI: 10.1186/s13098-024-01466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a kind of myocardial disease that occurs in diabetes patients and cannot be explained by hypertensive heart disease, coronary atherosclerotic heart disease and other heart diseases. Its pathogenesis may be closely related to programmed cell death, oxidative stress, intestinal microbes and micro-RNAs. The excessive activation of mineralocorticoid receptors (MR) in DCM can cause damage to the heart and kidneys. The third-generation non-steroidal mineralocorticoid receptor antagonist (MRA), finerenone, can effectively block MR, thus playing a role in protecting the heart and kidneys. This review mainly introduces the classification of MRA, and the mechanism of action, applications and limitations of finerenone in DCM, in order to provide reference for the study of treatment plans for DCM patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University; Southwest Medical University Affiliated Hospital Medical Group Gulin Hospital (Gulin County People's Hospital), Luzhou, Sichuan, China
| | - Haojie Xue
- Department of Cardiology, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University; Southwest Medical University Affiliated Hospital Medical Group Gulin Hospital (Gulin County People's Hospital), Luzhou, Sichuan, China
| | - Jinyu He
- Department of Cardiology, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University; Southwest Medical University Affiliated Hospital Medical Group Gulin Hospital (Gulin County People's Hospital), Luzhou, Sichuan, China
| | - Li Deng
- Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Julong Tian
- Department of Cardiology, The Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Yang Jiang
- Department of Cardiology, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University; Southwest Medical University Affiliated Hospital Medical Group Gulin Hospital (Gulin County People's Hospital), Luzhou, Sichuan, China.
| | - Jian Feng
- Department of Cardiology, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University; Southwest Medical University Affiliated Hospital Medical Group Gulin Hospital (Gulin County People's Hospital), Luzhou, Sichuan, China.
| |
Collapse
|
137
|
Montecino-Garrido H, Trostchansky A, Espinosa-Parrilla Y, Palomo I, Fuentes E. How Protein Depletion Balances Thrombosis and Bleeding Risk in the Context of Platelet's Activatory and Negative Signaling. Int J Mol Sci 2024; 25:10000. [PMID: 39337488 PMCID: PMC11432290 DOI: 10.3390/ijms251810000] [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: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Platelets are small cell fragments that play a crucial role in hemostasis, requiring fast response times and fine signaling pathway regulation. For this regulation, platelets require a balance between two pathway types: the activatory and negative signaling pathways. Activatory signaling mediators are positive responses that enhance stimuli initiated by a receptor in the platelet membrane. Negative signaling regulates and controls the responses downstream of the same receptors to roll back or even avoid spontaneous thrombotic events. Several blood-related pathologies can be observed when these processes are unregulated, such as massive bleeding in activatory signaling inhibition or thrombotic events for negative signaling inhibition. The study of each protein and metabolite in isolation does not help to understand the role of the protein or how it can be contrasted; however, understanding the balance between active and negative signaling could help develop effective therapies to prevent thrombotic events and bleeding disorders.
Collapse
Affiliation(s)
- Hector Montecino-Garrido
- Centro de Estudios en Alimentos Procesados (CEAP), ANID-Regional, Gore Maule R0912001, Talca 3480094, Chile
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Yolanda Espinosa-Parrilla
- Interuniversity Center for Healthy Aging (CIES), Centro Asistencial, Docente e Investigación-CADI-UMAG, Escuela de Medicina, Universidad de Magallanes, Punta Arenas 6210427, Chile
| | - Iván Palomo
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Eduardo Fuentes
- Thrombosis and Healthy Aging Research Center, Interuniversity Center for Healthy Aging (CIES), Interuniversity Network of Healthy Aging in Latin America and Caribbean (RIES-LAC), Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| |
Collapse
|
138
|
Erta G, Gersone G, Jurka A, Tretjakovs P. The Link between Salivary Amylase Activity, Overweight, and Glucose Homeostasis. Int J Mol Sci 2024; 25:9956. [PMID: 39337444 PMCID: PMC11432655 DOI: 10.3390/ijms25189956] [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: 08/18/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Butyrate, a short-chain fatty acid (SCFA) produced by the fermentation of dietary fibers in the colon, plays a pivotal role in regulating metabolic health, particularly by enhancing insulin sensitivity. Given the rising incidence of metabolic disorders, understanding the factors that influence butyrate production is of significant interest. This study explores the link between salivary amylase activity and butyrate levels in overweight women of reproductive age. Participants were categorized into low (LSA) and high (HSA) salivary amylase activity groups and further divided into two subgroups: one followed a low-starch diet (LS), and the other underwent caloric restriction (CR). We assessed salivary amylase activity and measured serum butyrate concentrations to examine their associations. Our findings showed a significant, though weak, positive correlation (ρ = 0.0486, p < 0.05), suggesting a link between salivary amylase activity and butyrate levels. The statistical significance, despite the weak correlation, implies that this relationship is not random. Moreover, higher baseline butyrate levels were observed in women with elevated salivary amylase activity. Also, women with low salivary amylase activity on a low-starch diet experienced a more pronounced increase in butyrate levels compared to those on caloric restriction. These results suggest that salivary amylase activity and dietary intake interact to influence butyrate production, with potential implications for improving insulin sensitivity and metabolic health. The study underscores the potential of butyrate in enhancing insulin sensitivity and promoting overall metabolic well-being. Further research is necessary to clarify the mechanisms involved and to understand the long-term effects of butyrate on metabolic health across different populations.
Collapse
Affiliation(s)
- Gita Erta
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Gita Gersone
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Antra Jurka
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Pēteris Tretjakovs
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| |
Collapse
|
139
|
Yu Q, Fu M, Hou Z, Wang Z. Developing a prediction model for preoperative acute heart failure in elderly hip fracture patients: a retrospective analysis. BMC Musculoskelet Disord 2024; 25:736. [PMID: 39277727 PMCID: PMC11401261 DOI: 10.1186/s12891-024-07843-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Hip fractures in the elderly are a common traumatic injury. Due to factors such as age and underlying diseases, these patients exhibit a high incidence of acute heart failure prior to surgery, severely impacting surgical outcomes and prognosis. OBJECTIVE This study aims to explore the potential risk factors for acute heart failure before surgery in elderly patients with hip fractures and to establish an effective clinical prediction model. METHODS This study employed a retrospective cohort study design and collected baseline and preoperative variables of elderly patients with hip fractures. Strict inclusion and exclusion criteria were adopted to ensure sample consistency. Statistical analyses were carried out using SPSS 24.0 and R software. A prediction model was developed using least absolute shrinkage and selection operator (LASSO) regression and multivariate logistic regression. The accuracy of the model was evaluated by analyzing the area under the receiver operating characteristic (ROC) curve (AUC) and a calibration curve was plotted to assess the model's calibration. RESULTS Between 2018 and 2019, 1962 elderly fracture patients were included in the study. After filtering, 1273 were analyzed. Approximately 25.7% of the patients experienced acute heart failure preoperatively. Through LASSO and logistic regression analyses, predictors for preoperative acute heart failure in elderly patients with hip fractures were identified as Gender was male (OR = 0.529, 95% CI: 0.381-0.734, P < 0.001), Age (OR = 1.760, 95% CI: 1.251-2.479, P = 0.001), Coronary Heart Disease (OR = 1.977, 95% CI: 1.454-2.687, P < 0.001), Chronic Obstructive Pulmonary Disease (COPD) (OR = 2.484, 95% CI: 1.154-5.346, P = 0.020), Complications (OR = 1.516, 95% CI: 1.033-2.226, P = 0.033), Anemia (OR = 2.668, 95% CI: 1.850-3.847, P < 0.001), and Hypoalbuminemia (OR 2.442, 95% CI: 1.682-3.544, P < 0.001). The linear prediction model of acute heart failure was Logit(P) = -2.167-0.637×partial regression coefficient for Gender was male + 0.566×partial regression coefficient for Age + 0.682×partial regression coefficient for Coronary heart disease + 0.910×partial regression coefficient for COPD + 0.416×partial regression coefficient for Complications + 0.981×partial regression coefficient for Anemia + 0.893×partial regression coefficient for Hypoalbuminemia, and the nomogram prediction model was established. The AUC of the predictive model was 0.763, indicating good predictive performance. Decision curve analysis revealed that the prediction model offers the greatest net benefit when the threshold probability ranges from 4 to 62%. CONCLUSION The prediction model we developed exhibits excellent accuracy in predicting the onset of acute heart failure preoperatively in elderly patients with hip fractures. It could potentially serve as an effective and useful clinical tool for physicians in conducting clinical assessments and individualized treatments.
Collapse
Affiliation(s)
- Qili Yu
- Department of Geriatric Orthopedics, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Mingming Fu
- Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China.
| | - Zhiqian Wang
- Department of Geriatric Orthopedics, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
140
|
Li H, Li X, Xu G, Zhan F. Minocycline alleviates lipopolysaccharide-induced cardiotoxicity by suppressing the NLRP3/Caspase-1 signaling pathway. Sci Rep 2024; 14:21180. [PMID: 39261543 PMCID: PMC11390881 DOI: 10.1038/s41598-024-72133-4] [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: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Minocycline (Min), as an antibiotic, possesses various beneficial properties such as anti-inflammatory, antioxidant, and anti-apoptotic effects. Despite these known qualities, the precise cardioprotective effect and mechanism of Min in protecting against sepsis-induced cardiotoxicity (SIC) remain unspecified. To address this, our study sought to assess the protective effects of Min on the heart. Lipopolysaccharide (LPS) was utilized to establish a cardiotoxicity model both in vivo and in vitro. Min was pretreated in the models. In the in vivo setting, evaluation of heart tissue histopathological injury was performed using hematoxylin and eosin (H&E) staining and TUNEL. Immunohistochemistry (IHC) was employed to evaluate the expression levels of NLRP3 and Caspase-1 in the heart tissue of mice. During in vitro experiments, the viability of H9c2 cells was gauged utilizing the CCK8 assay kit. Intracellular ROS levels in H9c2 cells were quantified using a ROS assay kit. Both in vitro and in vivo settings were subjected to measurement of oxidative stress indexes, encompassing glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. Additionglly, myocardial injury markers like lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB) activity were quantified using appropriate assay kits. Western blotting (WB) analysis was conducted to detect the expression levels of NOD-like receptor protein-3 (NLRP3), caspase-1, IL-18, and IL-1β, alongside apoptosis-related proteins such as Bcl-2 and Bax, and antioxidant proteins including superoxide dismutase-1 (SOD-1) and antioxidant proteins including superoxide dismutase-1 (SOD-2), both in H9c2 cells and mouse heart tissues. In vivo, Min was effective in reducing LPS-induced inflammation in cardiac tissue, preventing cell damage and apoptosis in cardiomyocytes. The levels of LDH and CK-MB were significantly reduced with Min treatment. In vitro studies showed that Min improved the viability of H9C2 cells, reduced apoptosis, and decreased ROS levels in these cells. Further analysis indicated that Min decreased the protein levels of NLRP3, Caspase-1, IL-18, and IL-1β, while increasing the levels of SOD-1 and SOD-2 both in vivo and in vitro. Min alleviates LPS-induced SIC by suppressing the NLRP3/Caspase-1 signalling pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Huijuan Li
- Department of Anesthesiology, Wuhan Third Hospital, Wuhan, 430074, China
| | - Xiaozhong Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China.
| |
Collapse
|
141
|
Fang Z, Zang Q, Chen J, Li Z, Yang D, Wu C, Yang H, Guo N. Whole-body mass spectrometry imaging reveals the systemic metabolic disorder and catecholamines biosynthesis alteration on heart-gut axis in heart failure rat. J Adv Res 2024:S2090-1232(24)00385-0. [PMID: 39270978 DOI: 10.1016/j.jare.2024.09.001] [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: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
INTRODUCTION Heart failure (HF) is a systemic metabolic disorder disease, across multiorgan investigations advancing knowledge of progression and treatment of HF. Whole-body MSI provides spatiotemporal information of metabolites in multiorgan and is expected to be a potent tool to dig out the complex mechanism of HF. OBJECTIVES This study aimed at exploring the systemic metabolic disorder in multiorgan and catecholamines biosynthesis alteration on heart-gut axis after HF. METHODS Whole-body MSI was used to characterize metabolic disorder of the whole rat body after HF. An integrated method by MSI, LC-MS/MS and ELISA was utilized to analyze key metabolites and enzymes on heart, small intestine, cecum and colon tissues of rat. Gut microbiota dysbiosis was investigated by 16S rDNA sequencing and metagenomic sequencing. Validation experiments and in vitro experiments were performed to verify the effect of catecholamines biosynthesis alteration on heart-gut axis after HF. RESULTS Whole-body MSI exhibited varieties of metabolites alteration in multiple organs. Remarkably, catecholamine biosynthesis was significantly altered in the serum, heart and intestines of rats. Furthermore, catecholamines and tyrosine hydroxylase were obviously upregulated in heart and colon tissue. Turicibacter_sanguinis was relevant to catecholamines of heart and colon. Validation experiments demonstrated excessive norepinephrine induced cardio-intestinal injury, including significantly elevating the levels of BNP, pro-BNP, LPS, DAO, and increased the abundance of Turicibacter_sanguinis. These alterations could be reversed by metoprolol treatment blocking the effect of norepinephrine. Additionally, in vitro studies demonstrated that norepinephrine promoted the growth of Turicibacter_sanguinis and Turicibacter_sanguinis could import and metabolize norepinephrine. Collectively, excessive norepinephrine exerted bidirectional effects on cardio-intestinal function to participate in the progression of HF. CONCLUSION Our study provides a new approach to elucidate multiorgan metabolic disorder and proposes new insights into heart-gut axis in HF development.
Collapse
Affiliation(s)
- Zhengyu Fang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingce Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiemei Chen
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zeyu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dawei Yang
- Zhong Yuan Academy of Biological Medicine, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Na Guo
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
142
|
Huang K, Lu J, Li Q, Wang C, Ding S, Xu X, Han L. The Role of Epicardial Adipose Tissue-Derived Proteins in Heart Failure with Preserved Ejection Fraction and Atrial Fibrillation: A Bioinformatics Analysis. J Inflamm Res 2024; 17:6093-6111. [PMID: 39257896 PMCID: PMC11385935 DOI: 10.2147/jir.s466203] [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: 03/29/2024] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
Background The accumulation of epicardial adipose tissue (EAT) is associated with cardiometabolic risks and adverse outcomes in heart failure with preserved ejection fraction (HFpEF) and atrial fibrillation (AF). This study aims to identify genes secreted by EAT that contribute to the shared pathogenesis of HFpEF and AF, potentially serving as biomarkers for diagnosis. Methods Data sets from the GEO database for HFpEF-EAT, HFpEF-heart tissue, AF-EAT, AF-PBMC, and AF-heart tissue were analyzed. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) identified key genes in EAT linked to HFpEF and AF. Functional enrichment and connectivity map analyses explored common pathways and therapeutic targets. Machine learning techniques, including LASSO regression, random forest, and support vector machine, identified shared biomarkers. CIBERSORT was used to assess immune cell infiltration, while gene set enrichment analysis identified pathways related to hub genes. Receiver operating characteristic (ROC) curve analysis and experimental validation assessed the bioinformatics findings. Results In the HFpEF dataset, 200 key genes were identified by intersecting HFpEF-EAT, HFpEF-heart tissue, WGCNA analyses, and secretory proteins. For AF, 232 related genes were identified through similar methods. Thirteen genes were common between HFpEF and AF, with two central genes, ITPKA and WNT9B, selected as potential biomarkers through machine learning and ROC analysis. Immune cell infiltration and gene set enrichment analysis revealed pathways related to ITPKA/WNT9B. These patterns were confirmed in human samples. Conclusion This study identified EAT-derived secretory proteins as potential biomarkers for HFpEF and AF, with ITPKA and WNT9B as central hub genes. These findings offer insights into potential diagnostic and therapeutic strategies for HFpEF and AF.
Collapse
Affiliation(s)
- Kai Huang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Jie Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Qin Li
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Chuyi Wang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Sufan Ding
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Xiangyang Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Lin Han
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
143
|
Frusciante L, Geminiani M, Shabab B, Olmastroni T, Scavello G, Rossi M, Mastroeni P, Nyong'a CN, Salvini L, Lamponi S, Parisi ML, Sinicropi A, Costa L, Spiga O, Trezza A, Santucci A. Exploring the Antioxidant and Anti-Inflammatory Potential of Saffron ( Crocus sativus) Tepals Extract within the Circular Bioeconomy. Antioxidants (Basel) 2024; 13:1082. [PMID: 39334741 PMCID: PMC11428576 DOI: 10.3390/antiox13091082] [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: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Repurposing saffron (Crocus sativus) waste presents a sustainable strategy for generating high-value products within the bioeconomy framework. Typically, flower components are discarded after stigma harvest, resulting in significant waste-350 kg of tepals per kilogram of stigmas. This research employed a comprehensive approach, integrating bioactivity studies (in vitro and in silico) with Life Cycle Assessment (LCA) evaluations, to extract and assess bioactive compounds from C. sativus tepals sourced in Tuscany, Italy. Phytochemical characterization using UPLC-MS/MS revealed a high abundance and variety of flavonoids in the hydro-ethanolic extract (CST). The antioxidant capacity was validated through various assays, and the ability to mitigate H2O2-induced oxidative stress and enhance fermentation was demonstrated in Saccharomyces cerevisiae. This study reports that C. sativus tepals extract reduces oxidative stress and boosts ethanol fermentation in yeast, paving the way for applications in the food and biofuels sectors. Further validation in RAW 264.7 macrophages confirmed CST's significant anti-inflammatory effects, indicating its potential for pharmaceutical, cosmeceutical, and nutraceutical applications. In silico studies identified potential targets involved in antioxidant and anti-inflammatory processes, shedding light on possible interaction mechanisms with Kaempferol 3-O-sophoroside (KOS-3), the predominant compound in the extract. The integration of LCA studies highlighted the environmental benefits of this approach. Overall, this research underscores the value of using waste-derived extracts through "green" methodologies, offering a model that may provide significant advantages for further evaluations compared to traditional methodologies and supporting the circular bioeconomy.
Collapse
Affiliation(s)
- Luisa Frusciante
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Michela Geminiani
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Behnaz Shabab
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Tommaso Olmastroni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Giorgia Scavello
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Martina Rossi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Pierfrancesco Mastroeni
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Collins Nyaberi Nyong'a
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Laura Salvini
- Fondazione Toscana Life Sciences, Strada del Petriccio e Belriguardo, 53100 Siena, Italy
| | - Stefania Lamponi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Maria Laura Parisi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- LifeCARES, Via Emilio Vezzosi 15, 52100 Arezzo, Italy
| | - Adalgisa Sinicropi
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- LifeCARES, Via Emilio Vezzosi 15, 52100 Arezzo, Italy
| | - Lorenzo Costa
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| | - Alfonso Trezza
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Annalisa Santucci
- Dipartimento di Biotecnologie Chimica e Farmacia, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- SienabioACTIVE, Università di Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| |
Collapse
|
144
|
Hu D, Sheeja Prabhakaran H, Zhang YY, Luo G, He W, Liou YC. Mitochondrial dysfunction in sepsis: mechanisms and therapeutic perspectives. Crit Care 2024; 28:292. [PMID: 39227925 PMCID: PMC11373266 DOI: 10.1186/s13054-024-05069-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] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/17/2024] [Indexed: 09/05/2024] Open
Abstract
Sepsis is a severe medical condition characterized by a systemic inflammatory response, often culminating in multiple organ dysfunction and high mortality rates. In recent years, there has been a growing recognition of the pivotal role played by mitochondrial damage in driving the progression of sepsis. Various factors contribute to mitochondrial impairment during sepsis, encompassing mechanisms such as reactive nitrogen/oxygen species generation, mitophagy inhibition, mitochondrial dynamics change, and mitochondrial membrane permeabilization. Damaged mitochondria actively participate in shaping the inflammatory milieu by triggering key signaling pathways, including those mediated by Toll-like receptors, NOD-like receptors, and cyclic GMP-AMP synthase. Consequently, there has been a surge of interest in developing therapeutic strategies targeting mitochondria to mitigate septic pathogenesis. This review aims to delve into the intricate mechanisms underpinning mitochondrial dysfunction during sepsis and its significant impact on immune dysregulation. Moreover, we spotlight promising mitochondria-targeted interventions that have demonstrated therapeutic efficacy in preclinical sepsis models.
Collapse
Affiliation(s)
- Dongxue Hu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Harshini Sheeja Prabhakaran
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Yuan-Yuan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China.
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore.
| |
Collapse
|
145
|
Wang Y, Feng W, Li S, Liu C, Jia L, Wang P, Li L, Du H, Yu W. Oxycodone attenuates lipopolysaccharide-induced myocardial injury by inhibiting inflammation, oxidation and pyroptosis via Nrf2/HO-1 signalling pathway. Clin Exp Pharmacol Physiol 2024; 51:e13910. [PMID: 39073215 DOI: 10.1111/1440-1681.13910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/27/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024]
Abstract
Myocardial injury and cardiovascular dysfunction are the most common complications of sepsis, and effective therapeutic candidate is still lacking. This study aims to investigate the protective effect of oxycodone in myocardial injury of lipopolysaccharide-induced sepsis and its related signalling pathways. Wild-type and nuclear factor erythroid 2-related factor 2 (Nrf2)-knockout mice, as well as H9c2 cardiomyocytes cultures treated with lipopolysaccharide (LPS) were used as models of septic myocardial injury. H9c2 cardiomyocytes culture showed that oxycodone protected cells from pyroptosis induced by LPS. Mice model confirmed that oxycodone pretreatment significantly attenuated myocardial pathological damage and improved cardiac function demonstrated by increased ejection fraction (EF) and fractional shortening (FS), as well as decreased cardiac troponin I (cTnI) and creatine kinase isoenzymes MB (CK-MB). Oxycodone also reduced the levels of inflammatory factors and oxidative stress damage induced by LPS, which involves pyroptosis-related proteins including: Nod-like receptor protein 3 (NLRP3), Caspase 1, Apoptosis-associated speck-like protein contain a CARD (ASC), and Gasdermin D (GSDMD). These changes were mediated by Nrf2 and heme oxygenase-1 (HO-1) because Nrf2-knockout mice or Nrf2 knockdown in H9c2 cells significantly reversed the beneficial effect of oxycodone on oxidative stress, inflammatory responses and NLRP3-mediated pyroptosis. Our findings yielded that oxycodone therapy reduces LPS-induced myocardial injury by suppressing NLRP3-mediated pyroptosis via the Nrf2/HO-1 signalling pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Yanting Wang
- The First Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Wei Feng
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaona Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cuicui Liu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Jia
- Department of Anesthesiology, Tianjin First Central Hospital, Tianjin, China
| | - Pei Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Linlin Li
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyin Du
- Tianjin Municipal Health Commission, Tianjin, China
| | - Wenli Yu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
146
|
Vecchiola A, Uslar T, Friedrich I, Aguirre J, Sandoval A, Carvajal CA, Tapia-Castillo A, Martínez-García A, Fardella CE. The role of sex hormones in aldosterone biosynthesis and their potential impact on its mineralocorticoid receptor. Cardiovasc Endocrinol Metab 2024; 13:e0305. [PMID: 38846628 PMCID: PMC11155591 DOI: 10.1097/xce.0000000000000305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/22/2024] [Indexed: 06/09/2024]
Abstract
Blood pressure (BP) regulation is a complex process involving various hormones, including aldosterone and its mineralocorticoid receptor. Mineralocorticoid receptor is expressed in several tissues, including the kidney, and plays a crucial role in regulating BP by controlling the sodium and water balance. During different stages of life, hormonal changes can affect mineralocorticoid receptor activity and aldosterone levels, leading to changes in BP. Increasing evidence suggests that sex steroids modulate aldosterone levels. Estrogens, particularly estradiol, mediate aldosterone biosynthesis by activating classical estrogen receptors and the G protein-coupled receptor. Progesterone acts as an anti-mineralocorticoid by inhibiting the binding of aldosterone to the mineralocorticoid receptor. Moreover, progesterone inhibits aldosterone synthase enzymes. The effect of testosterone on aldosterone synthesis is still a subject of debate. However, certain studies show that testosterone downregulates the mRNA levels of aldosterone synthase, leading to decreased plasma aldosterone levels.
Collapse
Affiliation(s)
- Andrea Vecchiola
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Thomas Uslar
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Isidora Friedrich
- Departamento de Endocrinologìa, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago
| | - Joaquin Aguirre
- Departamento de Endocrinologìa, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago
| | - Alejandra Sandoval
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cristian A. Carvajal
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Alejandra Tapia-Castillo
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Alejandra Martínez-García
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Carlos E. Fardella
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| |
Collapse
|
147
|
Yakoubi S. Synergistic integration of deep learning with protein docking in cardiovascular disease treatment strategies. IUBMB Life 2024; 76:666-696. [PMID: 38748776 DOI: 10.1002/iub.2819] [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: 02/11/2024] [Accepted: 03/13/2024] [Indexed: 08/31/2024]
Abstract
This research delves into the exploration of the potential of tocopherol-based nanoemulsion as a therapeutic agent for cardiovascular diseases (CVD) through an in-depth molecular docking analysis. The study focuses on elucidating the molecular interactions between tocopherol and seven key proteins (1O8a, 4YAY, 4DLI, 1HW9, 2YCW, 1BO9 and 1CX2) that play pivotal roles in CVD development. Through rigorous in silico docking investigations, assessment was conducted on the binding affinities, inhibitory potentials and interaction patterns of tocopherol with these target proteins. The findings revealed significant interactions, particularly with 4YAY, displaying a robust binding energy of -6.39 kcal/mol and a promising Ki value of 20.84 μM. Notable interactions were also observed with 1HW9, 4DLI, 2YCW and 1CX2, further indicating tocopherol's potential therapeutic relevance. In contrast, no interaction was observed with 1BO9. Furthermore, an examination of the common residues of 4YAY bound to tocopherol was carried out, highlighting key intermolecular hydrophobic bonds that contribute to the interaction's stability. Tocopherol complies with pharmacokinetics (Lipinski's and Veber's) rules for oral bioavailability and proves safety non-toxic and non-carcinogenic. Thus, deep learning-based protein language models ESM1-b and ProtT5 were leveraged for input encodings to predict interaction sites between the 4YAY protein and tocopherol. Hence, highly accurate predictions of these critical protein-ligand interactions were achieved. This study not only advances the understanding of these interactions but also highlights deep learning's immense potential in molecular biology and drug discovery. It underscores tocopherol's promise as a cardiovascular disease management candidate, shedding light on its molecular interactions and compatibility with biomolecule-like characteristics.
Collapse
Affiliation(s)
- Sana Yakoubi
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, Japan
- Alliance for Research on the Mediterranean North Africa (ARENA), University of Tsukuba, Ibaraki, Japan
- University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
148
|
Krukowski DC, Rao VN. Exposing Faults in the Foundation: Sodium-restrictive Guidance for Ambulatory Heart Failure. J Card Fail 2024; 30:1083-1085. [PMID: 39068983 DOI: 10.1016/j.cardfail.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Affiliation(s)
- Danielle C Krukowski
- Division of Cardiovascular Medicine, Medical University of South Carolina, Charleston, SC
| | - Vishal N Rao
- Division of Cardiovascular Medicine, Medical University of South Carolina, Charleston, SC; The Ralph H. Johnson Department of Veterans Affairs Medical Center, Charleston, SC.
| |
Collapse
|
149
|
Flori L, Benedetti G, Martelli A, Calderone V. Microbiota alterations associated with vascular diseases: postbiotics as a next-generation magic bullet for gut-vascular axis. Pharmacol Res 2024; 207:107334. [PMID: 39103131 DOI: 10.1016/j.phrs.2024.107334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The intestinal microbiota represents a key element in maintaining the homeostasis and health conditions of the host. Vascular pathologies and other risk factors such as aging have been recently associated with dysbiosis. The qualitative and quantitative alteration of the intestinal microbiota hinders correct metabolic homeostasis, causing structural and functional changes of the intestinal wall itself. Impairment of the intestinal microbiota, combined with the reduction of the barrier function, worsen the pathological scenarios of peripheral tissues over time, including the vascular one. Several experimental evidence, collected in this review, describes in detail the changes of the intestinal microbiota in dysbiosis associated with vascular alterations, such as atherosclerosis, hypertension, and endothelial dysfunction, the resulting metabolic disorders and how these can impact on vascular health. In this context, the gut-vascular axis is considered, for the first time, as a merged unit involved in the development and progression of vascular pathologies and as a promising target. Current approaches for the management of dysbiosis such as probiotics, prebiotics and dietary modifications act mainly on the intestinal district. Postbiotics, described as preparation of inanimate microorganisms and/or their components that confers health benefits on the host, represent an innovative strategy for a dual management of intestinal dysbiosis and vascular pathologies. In this context, this review has the further purpose of defining the positive effects of the supplementation of bacterial strains metabolites (short‑chain fatty acids, exopolysaccharides, lipoteichoic acids, gallic acid, and protocatechuic acid) restoring intestinal homeostasis and acting directly on the vascular district through the gut-vascular axis.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy.
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno, Pisa 6-56120, Italy; Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa 56120, Italy; Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa 56120, Italy.
| |
Collapse
|
150
|
Sun Y, Zhang R, Tian L, Pan Y, Sun X, Huang Z, Fan J, Chen J, Zhang K, Li S, Chen W, Bazzano LA, Kelly TN, He J, Bundy JD, Li C. Novel Metabolites Associated With Blood Pressure After Dietary Interventions. Hypertension 2024; 81:1966-1975. [PMID: 39005213 PMCID: PMC11324412 DOI: 10.1161/hypertensionaha.124.22999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The blood pressure (BP) etiologic study is complex due to multifactorial influences, including genetic, environmental, lifestyle, and their intricate interplays. We used a metabolomics approach to capture internal pathways and external exposures and to study BP regulation mechanisms after well-controlled dietary interventions. METHODS In the ProBP trail (Protein and Blood Pressure), a double-blinded crossover randomized controlled trial, participants underwent dietary interventions of carbohydrate, soy protein, and milk protein, receiving 40 g daily for 8 weeks, with 3-week washout periods. We measured plasma samples collected at baseline and at the end of each dietary intervention. Multivariate linear models were used to evaluate the association between metabolites and systolic/diastolic BP. Nominally significant metabolites were examined for enriching biological pathways. Significant ProBP findings were evaluated for replication among 1311 participants of the BHS (Bogalusa Heart Study), a population-based study conducted in the same area as ProBP. RESULTS After Bonferroni correction for 77 independent metabolite clusters (α=6.49×10-4), 18 metabolites were significantly associated with BP at baseline or the end of a dietary intervention, of which 11 were replicated in BHS. Seven emerged as novel discoveries, which are as follows: 1-linoleoyl-GPE (18:2), 1-oleoyl-GPE (18:1), 1-stearoyl-2-linoleoyl-GPC (18:0/18:2), 1-palmitoyl-2-oleoyl-GPE (16:0/18:1), maltose, N-stearoyl-sphinganine (d18:0/18:0), and N6-carbamoylthreonyladenosine. Pathway enrichment analyses suggested dietary protein intervention might reduce BP through pathways related to G protein-coupled receptors, incretin function, selenium micronutrient network, and mitochondrial biogenesis. CONCLUSIONS Seven novel metabolites were identified to be associated with BP at the end of different dietary interventions. The beneficial effects of protein interventions might be mediated through specific metabolic pathways.
Collapse
Affiliation(s)
- Yixi Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Ling Tian
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Yang Pan
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Xiao Sun
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Jia Fan
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Kai Zhang
- Department of Environmental Health Sciences, University of Albany, State University of New York, Rensselaer (K.Z.)
| | - Shengxu Li
- Children's Minnesota Research Institute, Children's Minnesota, Minneapolis (S.L.)
| | - Wei Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Joshua D Bundy
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| |
Collapse
|