1
|
Xu S, Hu C, Han J, Luo W, Huang L, Jiang Y, Samorodov AV, Wang Y, Huang J. Schisandrin B alleviates angiotensin II-induced cardiac inflammatory remodeling by inhibiting the recruitment of MyD88 to TLRs in mouse cardiomyocytes. Int Immunopharmacol 2024; 139:112660. [PMID: 39018688 DOI: 10.1016/j.intimp.2024.112660] [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: 03/04/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/19/2024]
Abstract
Cardiac tissue remodeling is characterized by altered heart tissue architecture and dysfunction, leading to heart failure. Sustained activation of the renin-angiotensin-aldosterone system (RAAS) greatly promotes the development of myocardial remodeling. Angiotensin II (Ang II), which is the major component of RAAS, can directly lead to cardiac remodeling by inducing an inflammatory response. Schisandrin B (Sch B), the active component extracted from the fruit of Schisandra chinensis (Turcz.) Baill has been shown to exhibit anti-inflammatory activity through its ability to target TLR4 and its adaptor protein, MyD88. In this study, we explored whether Sch B alleviates Ang II-induced myocardial inflammation and remodeling via targeting MyD88. Sch B significantly suppressed Ang II-induced inflammation as well as increased the expression of several genes of tissue remodeling (β-Mhc, Tgfb, Anp, α-Ska) both in vivo and in vitro. These protective effects of Sch B were due to the inhibition of recruitment of MyD88 to TLR2 and TLR4, suppressing the Ang II-induced NF-κB activation and reducing the following inflammatory responses. Moreover, the knockdown of Myd88 in cardiomyocytes abrogated the Ang II-induced increases in the production of inflammatory cytokines and expression of remodeling genes. These findings provide new evidence that the mechanism of Sch B protection was attributed to selective inhibition of MyD88 signaling. This finding could pave the way for novel therapeutic strategies for myocardial inflammatory diseases.
Collapse
Affiliation(s)
- Sujing Xu
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, China; Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Chenghong Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Jibo Han
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lijiang Huang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, China
| | - Yongsheng Jiang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, China
| | | | - Yi Wang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, China; School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China.
| | - Jianxiong Huang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, China.
| |
Collapse
|
2
|
Grijaldo-Alvarez SJB, Alvarez MRS, Schindler RL, Oloumi A, Hernandez N, Seales T, Angeles JGC, Nacario RC, Completo GC, Zivkovic AM, Bruce German J, Lebrilla CB. N-Glycan profile of the cell membrane as a probe for lipopolysaccharide-induced microglial neuroinflammation uncovers the effects of common fatty acid supplementation. Food Funct 2024; 15:8258-8273. [PMID: 39011570 DOI: 10.1039/d4fo01598c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Altered N-glycosylation of proteins on the cell membrane is associated with several neurodegenerative diseases. Microglia are an ideal model for studying glycosylation and neuroinflammation, but whether aberrant N-glycosylation in microglia can be restored by diet remains unknown. Herein, we profiled the N-glycome, proteome, and glycoproteome of the human microglia following lipopolysaccharide (LPS) induction to probe the impact of dietary and gut microbe-derived fatty acids-oleic acid, lauric acid, palmitic acid, valeric acid, butyric acid, isobutyric acid, and propionic acid-on neuroinflammation using liquid chromatography-tandem mass spectrometry. LPS changed N-glycosylation in the microglial glycocalyx altering high mannose and sialofucosylated N-glycans, suggesting the dysregulation of mannosidases, fucosyltransferases, and sialyltransferases. The results were consistent as we observed the restoration effect of the fatty acids, especially oleic acid, on the LPS-treated microglia, specifically on the high mannose and sialofucosylated glycoforms of translocon-associated proteins, SSRA and SSRB along with the cell surface proteins, CD63 and CD166. In addition, proteomic analysis and in silico modeling substantiated the potential of fatty acids in reverting the effects of LPS on microglial N-glycosylation. Our results showed that N-glycosylation is likely affected by diet by restoring alterations following LPS challenge, which may then influence the disease state.
Collapse
Affiliation(s)
- Sheryl Joyce B Grijaldo-Alvarez
- Department of Chemistry, University of California, Davis, 95616, USA.
- Institute of Chemistry, University of the Philippines Los Baños, Philippines, 4031.
| | | | | | - Armin Oloumi
- Department of Chemistry, University of California, Davis, 95616, USA.
| | - Noah Hernandez
- Department of Chemistry, University of California, Davis, 95616, USA.
| | - Tristan Seales
- Department of Chemistry, University of California, Davis, 95616, USA.
| | - Jorge Gil C Angeles
- Philippine Genome Center - Program for Agriculture, Livestock, Fisheries and Forestry, University of the Philippines Los Baños, Philippines, 4031.
| | - Ruel C Nacario
- Institute of Chemistry, University of the Philippines Los Baños, Philippines, 4031.
| | - Gladys C Completo
- Institute of Chemistry, University of the Philippines Los Baños, Philippines, 4031.
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, 95616, USA.
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, 95616, USA.
| | | |
Collapse
|
3
|
Jiang Y, Zhang S, Pan L, Leng J, Zhou T, Liu M, Li L, Zhao W. β-Glucan-based superabsorbent hydrogel acts as a gastrointestinal exoskeleton enhancing satiety and interfering fat hydrolysis. Int J Biol Macromol 2024; 275:133333. [PMID: 38945724 DOI: 10.1016/j.ijbiomac.2024.133333] [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: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/02/2024]
Abstract
Fat and its hydrolysis products, fatty acids, are indispensable nutritional components; however, prolonged excessive fat consumption, particularly in western diets, contributes to the onset of obesity and multiple metabolic disorders. In this study, we propose a daily-ingestible hydrogel (denoted as βC-MA hydrogel) composed of natural β-glucan and sodium carboxymethylcellulose crosslinked by malic acid at 120 °C. This hydrogel exhibits rapid swelling performance, up to 24-fold within 1 min and 176-fold after 1 h in deionized water. It also lengthens gastric retention and increases endogenous satiety signal levels, potentially controlling appetite and reducing food intake. Furthermore, βC-MA hydrogels that enter the small intestine can effectively inhibit fat hydrolysis and decrease triglyceride synthesis and transport. Specifically, the hydrogels inhibit the release of free fatty acids (FFAs) by approximately 50 % during digestion, influence the translocation of triglycerides and FFAs across the intestinal epithelium, and reduce the serum triglyceride levels by 22.2 %. These findings suggest that βC-MA hydrogels could serve as a noninvasive gastrointestinal device for weight control, with the advantage of reducing food intake and restoring lipid metabolism homeostasis.
Collapse
Affiliation(s)
- Yiming Jiang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Shiqi Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Linfan Pan
- School of Design, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Juncai Leng
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Tingyi Zhou
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Mingxuan Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Li Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wei Zhao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
4
|
Zhang C, Shi Y, Liu C, Sudesh SM, Hu Z, Li P, Liu Q, Ma Y, Shi A, Cai H. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol 2024; 23:169. [PMID: 38750502 PMCID: PMC11097480 DOI: 10.1186/s12933-024-02273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.
Collapse
Affiliation(s)
- Chaoyue Zhang
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunke Shi
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Changzhi Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shivon Mirza Sudesh
- Faculty of Medicine, St. George University of London, London, UK
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus
| | - Zhao Hu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pengyang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Qi Liu
- Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Yiming Ma
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ao Shi
- Faculty of Medicine, St. George University of London, London, UK.
- University of Nicosia Medical School, University of Nicosia, Nicosia, Cyprus.
| | - Hongyan Cai
- Cardiovascular Clinical Medical Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Heymsfield SB, Shapses SA. Guidance on Energy and Macronutrients across the Life Span. N Engl J Med 2024; 390:1299-1310. [PMID: 38598796 DOI: 10.1056/nejmra2214275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Steven B Heymsfield
- From the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge (S.B.H.); and the Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition, and Health, Rutgers University, and the Department of Medicine, Rutgers-Robert Wood Johnson School of Medicine - both in New Brunswick (S.A.S.)
| | - Sue A Shapses
- From the Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge (S.B.H.); and the Department of Nutritional Sciences and the New Jersey Institute for Food, Nutrition, and Health, Rutgers University, and the Department of Medicine, Rutgers-Robert Wood Johnson School of Medicine - both in New Brunswick (S.A.S.)
| |
Collapse
|
6
|
Salama RM, Eissa N, Doghish AS, Abulsoud AI, Abdelmaksoud NM, Mohammed OA, Abdel Mageed SS, Darwish SF. Decoding the secrets of longevity: unraveling nutraceutical and miRNA-Mediated aging pathways and therapeutic strategies. FRONTIERS IN AGING 2024; 5:1373741. [PMID: 38605867 PMCID: PMC11007187 DOI: 10.3389/fragi.2024.1373741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024]
Abstract
MicroRNAs (miRNAs) are short RNA molecules that are not involved in coding for proteins. They have a significant function in regulating gene expression after the process of transcription. Their participation in several biological processes has rendered them appealing subjects for investigating age-related disorders. Increasing data indicates that miRNAs can be influenced by dietary variables, such as macronutrients, micronutrients, trace minerals, and nutraceuticals. This review examines the influence of dietary factors and nutraceuticals on the regulation of miRNA in relation to the process of aging. We examine the present comprehension of miRNA disruption in age-related illnesses and emphasize the possibility of dietary manipulation as a means of prevention or treatment. Consolidating animal and human research is essential to validate the significance of dietary miRNA control in living organisms, despite the abundance of information already provided by several studies. This review elucidates the complex interaction among miRNAs, nutrition, and aging, offering valuable insights into promising areas for further research and potential therapies for age-related disorders.
Collapse
Affiliation(s)
- Rania M. Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| | - Nermin Eissa
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | - Ahmed I. Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | | | - Osama A. Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| | - Sherif S. Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Samar F. Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| |
Collapse
|
7
|
Zhu J, Bao Z, Hu Z, Wu S, Tian C, Zhou Y, Ding Z, Tan X. Myricetin alleviates diabetic cardiomyopathy by regulating gut microbiota and their metabolites. Nutr Diabetes 2024; 14:10. [PMID: 38472186 PMCID: PMC10933338 DOI: 10.1038/s41387-024-00268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The gut microbiota is involved in the pathogenesis of diabetic cardiomyopathy (DCM). Myricetin protects cardiac function in DCM. However, the low bioavailability of myricetin fails to explain its pharmacological mechanisms thoroughly. Research has shown that myricetin has a positive effect on the gut microbiota. We hypothesize that myricetin improves the development of DCM via regulating gut microbiota. METHODS DCM mice were induced with streptozotocin and fed a high-fat diet, and then treated with myricetin by gavage and high-fat diet for 16 weeks. Indexes related to gut microbiota composition, cardiac structure, cardiac function, intestinal barrier function, and inflammation were detected. Moreover, the gut contents were transplanted to DCM mice, and the effect of fecal microbiota transplantation (FMT) on DCM mice was assessed. RESULTS Myricetin could improve cardiac function in DCM mice by decreasing cardiomyocyte hypertrophy and interstitial fibrosis. The composition of gut microbiota, especially for short-chain fatty acid-producing bacteria involving Roseburia, Faecalibaculum, and Bifidobacterium, was more abundant by myricetin treatment in DCM mice. Myricetin increased occludin expression and the number of goblet cells in DCM mice. Compared with DCM mice unfed with gut content, the cardiac function, number of goblet cells, and expression of occludin in DCM mice fed by gut contents were elevated, while cardiomyocyte hypertrophy and TLR4/MyD88 pathway-related proteins were decreased. CONCLUSIONS Myricetin can prevent DCM development by increasing the abundance of beneficial gut microbiota and restoring the gut barrier function.
Collapse
Affiliation(s)
- Jinxiu Zhu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Longgang Maternity and Child Institute of Shantou University Medical College (Longgang District Maternity & Child Healthcare Hospital of Shenzhen City), 518172, Shenzhen, Guangdong, China
| | - Zhijun Bao
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Zuoqi Hu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Shenglin Wu
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Cuihong Tian
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Yueran Zhou
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Zipeng Ding
- Institute of Clinical Electrocardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China
| | - Xuerui Tan
- Department of Cardiology, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China.
- Clinical Research Center, the First Affiliated Hospital of Shantou University Medical College, 515041, Shantou, Guangdong, China.
| |
Collapse
|
8
|
Li X, Mai K, Ai Q. Palmitic acid activates NLRP3 inflammasome through NF-κB and AMPK-mitophagy-ROS pathways to induce IL-1β production in large yellow croaker (Larimichthys crocea). Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159428. [PMID: 38029958 DOI: 10.1016/j.bbalip.2023.159428] [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: 09/05/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023]
Abstract
Studies on marine fish showed that vegetable oils substituted for excessive fish oil increased interleukin-1β (IL-1β) production. However, whether the nucleotide-binding oligomerization domain, leucine-rich repeat-containing family, pyrin domain-containing-3 (NLRP3) inflammasome has a substantial role in fatty acid-induced IL-1β production in fish remains unclear. The associated specific mechanism is also unknown. In this study, nlrp3, caspase-1 and apoptosis-associated speck-like protein containing a CARD (asc) were successfully cloned, and NLRP3 inflammasome consisted of NLRP3, caspase-1 and ASC in large yellow croaker. Primary hepatocytes of fish incubated with palmitic acid (PA) exhibited the highest expression of pro-inflammatory genes (il-1β and tnfα) and NLRP3 inflammasome related genes (nlrp3, caspase-1 and asc), caspase-1 activity and IL-1β production among different treatments. Furthermore, PA-induced NLRP3 inflammasome activation was confirmed to require two signals: the first signal was that PA promoted the NF-κB (P65) protein into the nucleus, and NF-κB increased NLRP3 promoter activity and nlrp3 transcription. The second signal was that PA inhibited AMPK phosphorylation and decreased mitophagy by inhibiting the expression of PINK and parkin proteins, thereby damaging the mitochondria that could not be effectively cleared. Mitochondrial damage generated excessive amounts of reactive oxygen species, which activated the NLRP3 inflammasome and then induced caspase-1 activity and IL-1β production. Therefore, excessive dietary PA activated NLRP3 inflammasome through NF-κB and AMPK-mitophagy-ROS pathways to induce IL-1β production, thereby leading to inflammation in fish.
Collapse
Affiliation(s)
- Xueshan Li
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, People's Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong 266237, People's Republic of China.
| |
Collapse
|
9
|
Ali M, Kumari T, Gupta A, Akhtar S, Verma RD, Ghosh JK. Identification of a 10-mer peptide from the death domain of MyD88 which attenuates inflammation and insulin resistance and improves glucose metabolism. Biochem J 2024; 481:191-218. [PMID: 38224573 DOI: 10.1042/bcj20230369] [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: 08/26/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/17/2024]
Abstract
Insulin resistance (IR) is the key pathophysiological cause of type 2 diabetes, and inflammation has been implicated in it. The death domain (DD) of the adaptor protein, MyD88 plays a crucial role in the transduction of TLR4-associated inflammatory signal. Herein, we have identified a 10-residue peptide (M10), from the DD of MyD88 which seems to be involved in Myddosome formation. We hypothesized that M10 could inhibit MyD88-dependent TLR4-signaling and might have effects on inflammation-associated IR. Intriguingly, 10-mer M10 showed oligomeric nature and reversible self-assembly property indicating the peptide's ability to recognize its own amino acid sequence. M10 inhibited LPS-induced nuclear translocation of NF-κB in L6 myotubes and also reduced LPS-induced IL-6 and TNF-α production in peritoneal macrophages of BALB/c mice. Remarkably, M10 inhibited IL-6 and TNF-α secretion in diabetic, db/db mice. Notably, M10 abrogated IR in insulin-resistant L6 myotubes, which was associated with an increase in glucose uptake and a decrease in Ser307-phosphorylation of IRS1, TNF-α-induced JNK activation and nuclear translocation of NF-κB in these cells. Alternate day dosing with M10 (10 and 20 mg/kg) for 30 days in db/db mice significantly lowered blood glucose and improved glucose intolerance after loading, 3.0 g/kg glucose orally. Furthermore, M10 increased insulin and adiponectin secretion in db/db mice. M10-induced glucose uptake in L6 myotubes involved the activation of PI3K/AKT/GLUT4 pathways. A scrambled M10-analog was mostly inactive. Overall, the results show the identification of a 10-mer peptide from the DD of MyD88 with anti-inflammatory and anti-diabetic properties, suggesting that targeting of TLR4-inflammatory pathway, could lead to the discovery of molecules against IR and diabetes.
Collapse
Affiliation(s)
- Mehmood Ali
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Tripti Kumari
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
| | - Arvind Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Sariyah Akhtar
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
| | - Rahul Dev Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226 031 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 India
| |
Collapse
|
10
|
Qian C, Xu D, Wang J, Luo Y, Jin T, Huang L, Zhou Y, Cai Z, Jin B, Bao H, Wang Y. Toll-like receptor 2 deficiency ameliorates obesity-induced cardiomyopathy via inhibiting NF-κB signaling pathway. Int Immunopharmacol 2024; 128:111551. [PMID: 38278067 DOI: 10.1016/j.intimp.2024.111551] [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: 11/12/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
Growing evidence demonstrates that chronic low-grade inflammation, which is induced by high-fat diet (HFD) or saturated fatty acid, plays an important role in the obesity-induced cardiomyopathy (OIC) process. Moreover, obesity is associated with the activation of different inflammatory pathways, including nuclear factor-κB (NF-κB), Toll-like-receptor-2 (TLR2) and Toll-like-receptor-4 (TLR4). In this study, we established an HFD-induced cardiac injury mouse model and palmitate (PA)-induced myocardial cell model to evaluate the role of TLR2 in OIC. Our data show that TLR2 blockade using TLR2 knockout (KO) mice or a TLR2-specific inhibitor, C29, markedly ameliorated HFD- or PA-induced inflammation, myocardial fibrosis, and hypertrophy both in vivo and in vitro. Moreover, the PA-induced myocardial cell injury was mediated via inducing the formation of TLR2-MyD88 complex in a TLR4-independent manner in cardiomyocytes. Our data prove the critical role of cardiac TLR2 in the pathogenesis of HFD- and saturated fatty acid-induced myocarditis, fibrosis, myocardial hypertrophy, and cardiac dysfunction. Inhibition of TLR2 pathway may be a therapeutic strategy of OIC.
Collapse
Affiliation(s)
- Chenchen Qian
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyun Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiong Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianyang Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lijiang Huang
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Yafen Zhou
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Zhaohong Cai
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
| | - Bo Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongdan Bao
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China.
| | - Yi Wang
- Joint Research Center on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Ji L, Yang X, Jin Y, Li L, Yang B, Zhu W, Xu M, Wang Y, Wu G, Luo W, Lee K, Liang G. Blockage of DCLK1 in cardiomyocytes suppresses myocardial inflammation and alleviates diabetic cardiomyopathy in streptozotocin-induced diabetic mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166900. [PMID: 37778481 DOI: 10.1016/j.bbadis.2023.166900] [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: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a pathophysiological condition triggered by diabetes mellitus and can lead to heart failure. Doublecortin-like kinase protein 1 (DCLK1) is a multifunctional protein kinase involved in the regulation of cell proliferation, differentiation, survival, and migration. Current studies on DCLK1 mainly focus on cancer development; however, its role in non-tumor diseases such as DCM is yet to be deciphered. Our analysis revealed that DCLK1 was upregulated in cardiomyocytes of streptozotocin (STZ)-induced type 1 diabetic mouse, suggesting a correlation between DCLK1 and DCM progression. It was further demonstrated that either cardiomyocyte-specific DCLK1 knockout or pharmacological DCLK1 inhibitor DCLK1-IN-1 significantly alleviated cardiac hypertrophy and fibrosis in STZ-induced diabetic mice. RNA-seq analysis of heart tissues revealed that DCLK1 regulated the nuclear factor kappa B (NF-κB)-mediated inflammatory response in DCM. In vitro, DCLK1 activated NF-κB and the inflammatory response by inducing the IKKβ phosphorylation in high-concentration glucose (HG)-challenged cardiomyocytes. DCLK1-IN-1 also prevented HG-induced IKKβ/NF-κB activation and inflammatory injuries in cardiomyocytes. In conclusion, this study highlights the novel role of cardiomyocyte DCLK1 in regulating IKKβ/NF-κB, which aggravates inflammation to promote the pathogenesis of DCM. DCLK1 may serve as a new target for DCM treatment.
Collapse
Affiliation(s)
- Lijun Ji
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Xiaojing Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yiyi Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Li Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weiwei Zhu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mingjiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gaojun Wu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wu Luo
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Kwangyoul Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Guang Liang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Zhejiang Provincial People's Hospital, the Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
12
|
Facci L, Bolego C, Chemello C, Yasser R, Fusco M, Barbierato M, Giusti P, Moro S, Zusso M. 2-Pentadecyl-2-oxazoline inhibits lipopolysaccharide-induced microglia activation interfering with TLR4 signaling. Life Sci 2023; 335:122242. [PMID: 37952834 DOI: 10.1016/j.lfs.2023.122242] [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: 06/14/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023]
Abstract
AIM 2-Pentadecyl-2-oxazoline (PEA-OXA), the oxazoline derivative of N-palmitoylethanolamine, exerts anti-inflammatory activity; however, very little is known about the molecular mechanisms underlying this effect. Here, we tested the anti-neuroinflammatory effect of PEA-OXA in primary microglia and we also investigated the possible interaction of the molecule with the Toll-like receptor 4 (TLR4)-myeloid differentiation protein-2 (MD-2) complex. MAIN METHODS The anti-inflammatory effect of PEA-OXA was analyzed by measuring the expression and release of pro-inflammatory mediators in primary microglia by real-time PCR and ELISA, respectively. The effect of PEA-OXA on the activation of TLR4 signaling was assessed using two stably TLR4-transfected cell lines (i.e., HEK-293 and Ba/F3 cells). Finally, the putative binding mode of PEA-OXA to TLR4-MD-2 was investigated by molecular docking simulations. KEY FINDINGS Treatment with PEA-OXA resulted in the following effects: (i) it down-regulated gene expression of several pro-inflammatory molecules and the secretion of pro-inflammatory cytokines in LPS stimulated microglia cells; (ii) it did not prevent microglia activation after stimulation with TLR2 ligands; (iii) it prevented TLR4/NF-κB activation triggered by LPS in HEK-Blue™ hTLR4 cells; and (iv) it interfered with the binding of LPS to TLR4-MD-2 complex. Furthermore, molecular docking studies suggested that PEA-OXA could bind MD-2 with a 1:3 (MD-2/PEA-OXA) stoichiometry. CONCLUSION We show for the first time that the anti-neuroinflammatory effect of PEA-OXA involves its activity against TLR4 signaling, making this molecule a valuable tool for the development of new compounds directed to control neuroinflammation via inhibiting TLR4 signaling.
Collapse
Affiliation(s)
- Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Chiara Chemello
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Reem Yasser
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Mariella Fusco
- Scientific Information and Documentation Center, Epitech Group SpA, Padua, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Stefano Moro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy.
| |
Collapse
|
13
|
Chen Y, Lin W, Zhong L, Fang Z, Ye B, Wang Z, Chattipakorn N, Huang W, Liang G, Wu G. Bicyclol Attenuates Obesity-Induced Cardiomyopathy via Inhibiting NF-κB and MAPK Signaling Pathways. Cardiovasc Drugs Ther 2023; 37:1131-1141. [PMID: 35750941 DOI: 10.1007/s10557-022-07356-6] [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] [Accepted: 06/08/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE Schisandra is a well-known traditional Chinese medicine in East Asia. As a traditional Chinese medicine derivative with Schisandra chinensis as raw material, bicyclol is well known for its significant anti-inflammatory effect. Chronic inflammation plays a significant part in obesity-induced cardiomyopathy. Our purpose was to explore the effect and mechanism of bicyclol on obesity-induced cardiomyopathy. METHODS Mice fed with a high-fat diet (HFD) and cardiomyocytes stimulated by palmitic acid (PA) were used as models of obesity-related cardiomyopathy in vivo and in vitro, respectively. The therapeutic effect of bicyclol on pathological changes such as myocardial hypertrophy and fibrosis was evaluated by staining cardiac tissue sections. PCR was used to detect inflammatory factors in H9c2 cells and animal heart tissue after bicyclol treatment. Then, we used western blotting to detect the expression levels of the myocardial hypertrophy related protein, myocardial fibrosis related protein, NF-κB and MAPK pathways. RESULTS Our results indicated that bicyclol treatment significantly alleviates HFD-induced myocardial inflammation, fibrosis, and hypertrophy by inhibiting the MAPK and NF-κB pathways. Similar to animal level results, bicyclol could significantly inhibit PA-induced inflammation and prevent NF-κB and MAPK pathways from being activated. CONCLUSION Our results showed that bicyclol has potential as a drug to treat obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Yanghao Chen
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
| | - Wante Lin
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
| | - Lingfeng Zhong
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
| | - Zimin Fang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
| | - Bozhi Ye
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China
| | - Zhe Wang
- Department of Pharmacy, the Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325000, Wenzhou, China
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China.
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, 311399, Hangzhou, China.
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325035, Wenzhou, China.
| |
Collapse
|
14
|
Kim S, Lee S, Kim TY, Lee SH, Seo SU, Kweon MN. Newly isolated Lactobacillus paracasei strain modulates lung immunity and improves the capacity to cope with influenza virus infection. MICROBIOME 2023; 11:260. [PMID: 37996951 PMCID: PMC10666316 DOI: 10.1186/s40168-023-01687-8] [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: 05/23/2023] [Accepted: 10/01/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND The modulation of immune responses by probiotics is crucial for local and systemic immunity. Recent studies have suggested a correlation between gut microbiota and lung immunity, known as the gut-lung axis. However, the evidence and mechanisms underlying this axis remain elusive. RESULTS In this study, we screened various Lactobacillus (L.) strains for their ability to augment type I interferon (IFN-I) signaling using an IFN-α/β reporter cell line. We identified L. paracasei (MI29) from the feces of healthy volunteers, which showed enhanced IFN-I signaling in vitro. Oral administration of the MI29 strain to wild-type B6 mice for 2 weeks resulted in increased expression of IFN-stimulated genes and pro-inflammatory cytokines in the lungs. We found that MI29-treated mice had significantly increased numbers of CD11c+PDCA-1+ plasmacytoid dendritic cells and Ly6Chi monocytes in the lungs compared with control groups. Pre-treatment with MI29 for 2 weeks resulted in less weight loss and lower viral loads in the lung after a sub-lethal dose of influenza virus infection. Interestingly, IFNAR1-/- mice did not show enhanced viral resistance in response to oral MI29 administration. Furthermore, metabolic profiles of MI29-treated mice revealed changes in fatty acid metabolism, with MI29-derived fatty acids contributing to host defense in a Gpr40/120-dependent manner. CONCLUSIONS These findings suggest that the newly isolated MI29 strain can activate host defense immunity and prevent infections caused by the influenza virus through the gut-lung axis. Video Abstract.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sohyeon Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Tae-Young Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Su-Hyun Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Tepavcevic S, Romic S, Zec M, Culafic T, Stojiljkovic M, Ivkovic T, Pantelic M, Kostic M, Stanisic J, Koricanac G. Effects of Walnut-Rich Diet on Cation-Handling Proteins in the Heart of Healthy and Metabolically Compromised Male Rats. J Med Food 2023; 26:849-857. [PMID: 37889606 DOI: 10.1089/jmf.2022.0157] [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: 10/29/2023] Open
Abstract
The transport of cations in the cardiomyocytes, crucial for the functioning of the heart, can be affected by walnut diet due to the high content of polyunsaturated fatty acids. Healthy and metabolically compromised rats (drinking 10% fructose solution) were subjected to a diet supplemented with 2.4 g of walnuts for 6 weeks to investigate the effect on proteins involved in cation transport in the heart cells. Fructose increased the level of the α1 subunit of Na+/K+-ATPase and the phosphorylation of extracellular signal-regulated kinase 1/2 in the heart of control and walnut-eating rats, while elevated L-type calcium channel α (LTCCα), sodium-calcium exchanger 1 (NCX1), and Maxi Kα level were observed only in rats that did not consume walnuts. However, walnuts significantly increased the cardiac content of LTCC, NCX1, and Maxi Kα, as well as Kir6.1 and SUR2B subunits of KATP channel, but only in fructose-naive rats. In animals that drank fructose, a significant increasing effect of walnuts was observed only in Akt kinase phosphorylation, which may be a part of the antiarrhythmic mechanism of decreasing cation currents in cardiomyocytes. The walnut diet-induced increase in LTCC and NCX1 expression in healthy rats may indicate intense cardiac calcium turnover, whereas the effect on Kir6.1 and SUR2B subunits suggests stimulation of KATP channel transport in the cardiac vasculature. The effects of walnuts on the cation-handling proteins in the heart, mostly limited to healthy animals, suggest the possible use of a walnut-supplemented diet in the prevention rather than the treatment of cardiological channelopathies.
Collapse
Affiliation(s)
- Snezana Tepavcevic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Snjezana Romic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Manja Zec
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Tijana Culafic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mojca Stojiljkovic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Ivkovic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Pantelic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milan Kostic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Stanisic
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Goran Koricanac
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Zhang Q, Zhu W, Lou S, Bao H, Zhou Y, Cai Z, Ye J, Cui Y, Wang M, Jin L, Liang G, Luo W, Wang Y. Compound c17 alleviates inflammatory cardiomyopathy in streptozotocin-induced diabetic mice by targeting MyD88. Int Immunopharmacol 2023; 124:110863. [PMID: 37703787 DOI: 10.1016/j.intimp.2023.110863] [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: 07/02/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a common complication of diabetes mellitus and is associated with increased morbidity and mortality due to cardiac dysfunction. Chronic inflammation plays a significant role in the development of DCM, making it a promising target for novel pharmacological strategies. Our previous study has synthesized a novel compound, c17, which exhibited strong anti-inflammatory activity by specifically targeting to myeloid differentiation primary response 88 (MyD88). In this study, we evaluated the therapeutic effect of c17 in DCM. METHODS The small molecular selective MyD88 inhibitor, c17, was used to evaluate the effect of MyD88 on DCM in both high concentration of glucose- and palmitic acid-stimulated macrophages and streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM) mice. RESULTS The treatment of c17 in T1DM mice resulted in improved heart function and reduced cardiac hypertrophy, inflammation and fibrogenesis. RNA sequencing analysis of the heart tissues revealed that c17 effectively suppressed the inflammatory response by regulating the MyD88-dependent pathway. Co-immunoprecipitation experiments further confirmed that c17 disrupted the interaction between MyD88 and Toll-like receptor 4 (TLR4), consequently inhibiting downstream NF-κB activation. In vitro studies demonstrated that c17 exhibited similar anti-inflammatory activity by targeting MyD88 in macrophages, which are the primary regulators of cardiac inflammation. Furthermore, conditioned medium derived from c17-treated macrophages showed reduced capacity to induce hypertrophy, pro-fibrotic reactions, and secondary inflammation in cardiomyocytes. CONCLUSIONS In conclusion, the small-molecule MyD88 inhibitor, c17, effectively combated the inflammatory DCM, therefore could be a potential candidate for the treatment of this disease.
Collapse
Affiliation(s)
- Qianhui Zhang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Weiwei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Shuaijie Lou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Hongdan Bao
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, Zhejiang, China
| | - Yafen Zhou
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, Zhejiang, China
| | - Zhaohong Cai
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, Zhejiang, China
| | - Jiaxi Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yaqian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Leiming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, Zhejiang, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| | - Yi Wang
- Joint Research Center on Medicine, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo 315700, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; School of Pharmacy, Hangzhou Normal University, Hangzhou 311399, Zhejiang, China.
| |
Collapse
|
17
|
Chattergoon N, Louey S, Jonker SS, Thornburg KL. Thyroid hormone increases fatty acid use in fetal ovine cardiac myocytes. Physiol Rep 2023; 11:e15865. [PMID: 38010207 PMCID: PMC10680578 DOI: 10.14814/phy2.15865] [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: 09/12/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/29/2023] Open
Abstract
Cardiac metabolic substrate preference shifts at parturition from carbohydrates to fatty acids. We hypothesized that thyroid hormone (T3 ) and palmitic acid (PA) stimulate fetal cardiomyocyte oxidative metabolism capacity. T3 was infused into fetal sheep to a target of 1.5 nM. Dispersed cardiomyocytes were assessed for lipid uptake and droplet formation with BODIPY-labeled fatty acids. Myocardial expression levels were assessed PCR. Cardiomyocytes from naïve fetuses were exposed to T3 and PA, and oxygen consumption was measured with the Seahorse Bioanalyzer. Cardiomyocytes (130-day gestational age) exposed to elevated T3 in utero accumulated 42% more long-chain fatty acid droplets than did cells from vehicle-infused fetuses. In utero T3 increased myocardial mRNA levels of CD36, CPT1A, CPT1B, LCAD, VLCAD, HADH, IDH, PDK4, and caspase 9. In vitro exposure to T3 increased maximal oxygen consumption rate in cultured cardiomyocytes in the absence of fatty acids, and when PA was provided as an acute (30 min) supply of cellular energy. Longer-term exposure (24 and 48 h) to PA abrogated increased oxygen consumption rates stimulated by elevated levels of T3 in cultured cardiomyocytes. T3 contributes to metabolic maturation of fetal cardiomyocytes. Prolonged exposure of fetal cardiomyocytes to PA, however, may impair oxidative capacity.
Collapse
Affiliation(s)
- Natasha Chattergoon
- Center for Developmental Health, Knight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Samantha Louey
- Center for Developmental Health, Knight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Sonnet S. Jonker
- Center for Developmental Health, Knight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Kent L. Thornburg
- Center for Developmental Health, Knight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
18
|
Yue Q, Liu Y, Ji J, Hu T, Lin T, Yu S, Li S, Wu N. Down-regulation of OIP5-AS1 inhibits obesity-induced myocardial pyroptosis and miR-22/NLRP3 inflammasome axis. Immun Inflamm Dis 2023; 11:e1066. [PMID: 37904706 PMCID: PMC10611552 DOI: 10.1002/iid3.1066] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Obesity can induce myocardial pyroptosis, but the exact mechanism is still unknown. A recent study reported the association of opa-interacting protein 5-antisense transcript 1 (OIP5-AS1), an evolutionarily conserved long noncoding RNA, with pyroptosis. Therefore, this study aimed to investigate the role of OIP5-AS1 in obesity-induced myocardial pyroptosis. METHODS OIP5-AS1 was downregulated in H9c2 cells, followed by treatment with 400 μM palmitic acid (PA). Propidium iodide (PI) staining, lactic dehydrogenase (LDH) release assay, caspase-1 activity assay, IL-1β, and IL-18 activity assay were performed to detect pyroptotic phenotype. The interaction between OIP5-AS1 and microRNAs (miRNAs) was analyzed using RNA pull-down and luciferase assay. The effect of OIP5-AS1 knockdown in high-fat diet (HFD)-induced obesity rat on cardiac function, myocardial hypertrophy, fibrosis, and remodeling was evaluated. RESULTS Fat deposition was observed in cardiomyocytes 24 h after PA treatment; moreover, PA-treated cardiomyocytes showed significant increase in the rate of pyroptotic cells, release of LDH, protein expressions of NLRP3 and cleaved caspase-1, and the activity of caspase-1, IL-1β, and IL-18 as well as OIP5-AS1 expression. These findings suggested that PA activated pyroptosis and induced OIP5-AS1 expression in cardiomyocytes. Moreover, OIP5-AS1 knockdown inhibited PA-induced pyroptosis. Mechanistically, OIP5-AS1 was found to specifically bind to miR-22 and to regulate NLRP3 inflammasome-mediated pyroptosis via miR-22. Furthermore, OIP5-AS1 knockdown ameliorated HFD-induced cardiac dysfunction, myocardial hypertrophy, fibrosis, remodeling, and pyroptosis. CONCLUSION Our results revealed that downregulation of OIP5-AS1 can inhibit obesity-induced myocardial pyroptosis via miR-22/NLRP3 inflammasome axis. This finding lays a foundation of gene therapy for heart disease targeting OIP5-AS1.
Collapse
Affiliation(s)
- Qingxiong Yue
- Department of UltrasoundDalian Municipal Central HospitalDalianLiaoning ProvinceChina
| | - Yan Liu
- Department of UltrasoundDalian Women and Children's Medical GroupDalianLiaoning ProvinceChina
| | - Jun Ji
- Department of Central LaboratoryDalian Municipal Central HospitalDalianLiaoning ProvinceChina
| | - Tao Hu
- Department of UltrasoundDalian Municipal Central HospitalDalianLiaoning ProvinceChina
| | - Tong Lin
- Department of UltrasoundDalian Municipal Central HospitalDalianLiaoning ProvinceChina
| | - Shuang Yu
- Department of Central LaboratoryFirst Affiliated Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Shijun Li
- Department of CardiologyDalian Municipal Central HospitalDalianLiaoning ProvinceChina
| | - Nan Wu
- Department of Central LaboratoryFirst Affiliated Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
19
|
Zhuang P, Liu X, Li Y, Wu Y, Li H, Wan X, Zhang L, Xu C, Jiao J, Zhang Y. Circulating fatty acids, genetic risk, and incident coronary artery disease: A prospective, longitudinal cohort study. SCIENCE ADVANCES 2023; 9:eadf9037. [PMID: 37738352 PMCID: PMC10881029 DOI: 10.1126/sciadv.adf9037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 08/11/2023] [Indexed: 09/24/2023]
Abstract
The role of fatty acids (FAs) in primary prevention of coronary artery disease (CAD) is highly debated, and the modification effect by genetic risk profiles remains unclear. Here, we report the prospective associations of circulating FAs and genetic predisposition with CAD development in 101,367 U.K. Biobank participants. A total of 3719 CAD cases occurred during a mean follow-up of 11.5 years. Plasma monounsaturated FAs (MUFAs) were positively associated with risk of CAD, whereas the risk was significantly lower with higher n-3 polyunsaturated FAs (PUFAs) and more reductions in risk were detected among TT carriers of rs174547. Furthermore, increased plasma saturated FAs (SFAs) and linoleic acid were related to a significant increase in CAD risk among participants with high genetic risk (genetic risk score > 90%). These findings suggest that individuals with high genetic risk need to reduce plasma SFAs levels for CAD prevention. Supplementation of n-3 PUFAs for CAD prevention may consider individuals' genetic makeup.
Collapse
Affiliation(s)
- Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yin Li
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuqi Wu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haoyu Li
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuzhi Wan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lange Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Tanaka Y, Nagoshi T, Takahashi H, Oi Y, Yasutake R, Yoshii A, Kimura H, Kashiwagi Y, Tanaka TD, Shimoda M, Yoshimura M. URAT1 is expressed in cardiomyocytes and dotinurad attenuates the development of diet-induced metabolic heart disease. iScience 2023; 26:107730. [PMID: 37694143 PMCID: PMC10483053 DOI: 10.1016/j.isci.2023.107730] [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: 11/28/2022] [Revised: 05/17/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023] Open
Abstract
We recently reported that the selective inhibition of urate transporter-1 (URAT1), which is primarily expressed in the kidneys, ameliorates insulin resistance by attenuating hepatic steatosis and improving brown adipose tissue function in diet-induced obesity. In this study, we evaluated the effects of dotinurad, a URAT1-selective inhibitor, on the hearts of high-fat diet (HFD)-fed obese mice for 16-20 weeks and on neonatal rat cardiomyocytes (NRCMs) exposed to palmitic acid. Outside the kidneys, URAT1 was also expressed in cardiomyocytes and indeed worked as a uric acid transporter. Dotinurad substantially attenuated HFD-induced cardiac fibrosis, inflammatory responses, and cardiac dysfunction. Intriguingly, among various factors related to the pathophysiology of diet-induced obesity, palmitic acid significantly increased URAT1 expression in NRCMs and subsequently induced apoptosis, oxidative stress, and inflammatory responses via MAPK pathway, all of which were reduced by dotinurad. These results indicate that URAT1 is a potential therapeutic target for metabolic heart disease.
Collapse
Affiliation(s)
- Yoshiro Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hirotake Takahashi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yuhei Oi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Rei Yasutake
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Akira Yoshii
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Haruka Kimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yusuke Kashiwagi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toshikazu D. Tanaka
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
21
|
Muniz-Santos R, Lucieri-Costa G, de Almeida MAP, Moraes-de-Souza I, Brito MADSM, Silva AR, Gonçalves-de-Albuquerque CF. Lipid oxidation dysregulation: an emerging player in the pathophysiology of sepsis. Front Immunol 2023; 14:1224335. [PMID: 37600769 PMCID: PMC10435884 DOI: 10.3389/fimmu.2023.1224335] [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: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 08/22/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by abnormal host response to infection. Millions of people are affected annually worldwide. Derangement of the inflammatory response is crucial in sepsis pathogenesis. However, metabolic, coagulation, and thermoregulatory alterations also occur in patients with sepsis. Fatty acid mobilization and oxidation changes may assume the role of a protagonist in sepsis pathogenesis. Lipid oxidation and free fatty acids (FFAs) are potentially valuable markers for sepsis diagnosis and prognosis. Herein, we discuss inflammatory and metabolic dysfunction during sepsis, focusing on fatty acid oxidation (FAO) alterations in the liver and muscle (skeletal and cardiac) and their implications in sepsis development.
Collapse
Affiliation(s)
- Renan Muniz-Santos
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giovanna Lucieri-Costa
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus Augusto P. de Almeida
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabelle Moraes-de-Souza
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Adriana Ribeiro Silva
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratory of Immunopharmacology, Department of Physiology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroscience Graduate Program, Federal Fluminense University, Niteroi, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Petrenko V, Sinturel F, Riezman H, Dibner C. Lipid metabolism around the body clocks. Prog Lipid Res 2023; 91:101235. [PMID: 37187314 DOI: 10.1016/j.plipres.2023.101235] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
Lipids play important roles in energy metabolism along with diverse aspects of biological membrane structure, signaling and other functions. Perturbations of lipid metabolism are responsible for the development of various pathologies comprising metabolic syndrome, obesity, and type 2 diabetes. Accumulating evidence suggests that circadian oscillators, operative in most cells of our body, coordinate temporal aspects of lipid homeostasis. In this review we summarize current knowledge on the circadian regulation of lipid digestion, absorption, transportation, biosynthesis, catabolism, and storage. Specifically, we focus on the molecular interactions between functional clockwork and biosynthetic pathways of major lipid classes comprising cholesterol, fatty acids, triacylglycerols, glycerophospholipids, glycosphingolipids, and sphingomyelins. A growing body of epidemiological studies associate a socially imposed circadian misalignment common in modern society with growing incidence of metabolic disorders, however the disruption of lipid metabolism rhythms in this connection has only been recently revealed. Here, we highlight recent studies that unravel the mechanistic link between intracellular molecular clocks, lipid homeostasis and development of metabolic diseases based on animal models of clock disruption and on innovative translational studies in humans. We also discuss the perspectives of manipulating circadian oscillators as a potentially powerful approach for preventing and managing metabolic disorders in human patients.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland
| | - Flore Sinturel
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, Geneva 1211, Switzerland
| | - Charna Dibner
- Thoracic and Endocrine Surgery Division, Department of Surgery, University Hospital of Geneva, Geneva 1211, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), Geneva 1211, Switzerland.
| |
Collapse
|
23
|
Najjar RS. The Impacts of Animal-Based Diets in Cardiovascular Disease Development: A Cellular and Physiological Overview. J Cardiovasc Dev Dis 2023; 10:282. [PMID: 37504538 PMCID: PMC10380617 DOI: 10.3390/jcdd10070282] [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: 05/24/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the United States, and diet plays an instrumental role in CVD development. Plant-based diets have been strongly tied to a reduction in CVD incidence. In contrast, animal food consumption may increase CVD risk. While increased serum low-density lipoprotein (LDL) cholesterol concentrations are an established risk factor which may partially explain the positive association with animal foods and CVD, numerous other biochemical factors are also at play. Thus, the aim of this review is to summarize the major cellular and molecular effects of animal food consumption in relation to CVD development. Animal-food-centered diets may (1) increase cardiovascular toll-like receptor (TLR) signaling, due to increased serum endotoxins and oxidized LDL cholesterol, (2) increase cardiovascular lipotoxicity, (3) increase renin-angiotensin system components and subsequent angiotensin II type-1 receptor (AT1R) signaling and (4) increase serum trimethylamine-N-oxide concentrations. These nutritionally mediated factors independently increase cardiovascular oxidative stress and inflammation and are all independently tied to CVD development. Public policy efforts should continue to advocate for the consumption of a mostly plant-based diet, with the minimization of animal-based foods.
Collapse
Affiliation(s)
- Rami Salim Najjar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
24
|
Richter H, Gover O, Schwartz B. Anti-Inflammatory Activity of Black Soldier Fly Oil Associated with Modulation of TLR Signaling: A Metabolomic Approach. Int J Mol Sci 2023; 24:10634. [PMID: 37445812 DOI: 10.3390/ijms241310634] [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: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Dietary intervention in the treatment of ulcerative colitis involves, among other things, modifications in fatty acid content and/or profile. For example, replacing saturated long chain fatty acids with medium chain fatty acids (MCFAs) has been reported to ameliorate inflammation. The Black Soldier Fly Larvae's (BSFL) oil is considered a sustainable dietary ingredient rich in the MCFA C12:0; however, its effect on inflammatory-related conditions has not been studied until now. Thus, the present study aimed to investigate the anti-inflammatory activity of BSFL oil in comparison to C12:0 using TLR4- or TLR2-activated THP-1 and J774A.1 cell lines and to assess its putative protective effect against dextran sulfate sodium (DSS)-induced acute colitis in mice. BSFL oil and C12:0 suppressed proinflammatory cytokines release in LPS-stimulated macrophages; however, only BSFL oil exerted anti-inflammatory activity in Pam3CSK4-stimulated macrophages. Transcriptome analysis provided insight into the possible role of BSFL oil in immunometabolism switch, involving mTOR signaling and an increase in PPAR target genes promoting fatty acid oxidation, exhibiting a discrepant mode of action compared to C12:0 treatment, which mainly affected cholesterol biosynthesis pathways. Additionally, we identified anti-inflammatory eicosanoids, oxylipins, and isoprenoids in the BSFL oil that may contribute to an orchestrated anti-inflammatory response. In vivo, a BSFL oil-enriched diet (20%) ameliorated the clinical signs of colitis, as indicated by improved body weight recovery, reduced colon shortening, reduced splenomegaly, and an earlier phase of secretory IgA response. These results indicate the novel beneficial use of BSFL oil as a modulator of inflammation.
Collapse
Affiliation(s)
- Hadas Richter
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 761001, Israel
| | - Ofer Gover
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 761001, Israel
| | - Betty Schwartz
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 761001, Israel
| |
Collapse
|
25
|
Bestepe F, Fritsche C, Lakhotiya K, Niosi CE, Ghanem GF, Martin GL, Pal-Ghosh R, Becker-Greene D, Weston J, Hollan I, Risnes I, Rynning SE, Solheim LH, Feinberg MW, Blanton RM, Icli B. Deficiency of miR-409-3p improves myocardial neovascularization and function through modulation of DNAJB9/p38 MAPK signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:995-1009. [PMID: 37332476 PMCID: PMC10276151 DOI: 10.1016/j.omtn.2023.05.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
Angiogenesis is critical for tissue repair following myocardial infarction (MI), which is exacerbated under insulin resistance or diabetes. MicroRNAs are regulators of angiogenesis. We examined the metabolic regulation of miR-409-3p in post-infarct angiogenesis. miR-409-3p was increased in patients with acute coronary syndrome (ACS) and in a mouse model of acute MI. In endothelial cells (ECs), miR-409-3p was induced by palmitate, while vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) decreased its expression. Overexpression of miR-409-3p decreased EC proliferation and migration in the presence of palmitate, whereas inhibition had the opposite effects. RNA sequencing (RNA-seq) profiling in ECs identified DNAJ homolog subfamily B member 9 (DNAJB9) as a target of miR-409-3p. Overexpression of miR-409-3p decreased DNAJB9 mRNA and protein expression by 47% and 31% respectively, while enriching DNAJB9 mRNA by 1.9-fold after Argonaute2 microribonucleoprotein immunoprecipitation. These effects were mediated through p38 mitogen-activated protein kinase (MAPK). Ischemia-reperfusion (I/R) injury in EC-specific miR-409-3p knockout (KO) mice (miR-409ECKO) fed a high-fat, high-sucrose diet increased isolectin B4 (53.3%), CD31 (56%), and DNAJB9 (41.5%). The left ventricular ejection fraction (EF) was improved by 28%, and the infarct area was decreased by 33.8% in miR-409ECKO compared with control mice. These findings support an important role of miR-409-3p in the angiogenic EC response to myocardial ischemia.
Collapse
Affiliation(s)
- Furkan Bestepe
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Colette Fritsche
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Kartik Lakhotiya
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Carolyn E. Niosi
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - George F. Ghanem
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Ruma Pal-Ghosh
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Dakota Becker-Greene
- Cardiovascular Division, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - James Weston
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Ivana Hollan
- Department of Health Sciences, Norwegian University of Science and Technology, Gjøvik, Norway
| | - Ivar Risnes
- Department of Cardiac Surgery, LHL Hospital Gardermoen, Jessheim, Norway
| | - Stein Erik Rynning
- Department of Heart Diseases, Haukeland University Hospital, Bergen, Norway
| | | | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Basak Icli
- Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| |
Collapse
|
26
|
Yang N, Wang M, Lin K, Wang M, Xu D, Han X, Zhao X, Wang Y, Wu G, Luo W, Liang G, Shan P. Dectin-1 deficiency alleviates diabetic cardiomyopathy by attenuating macrophage-mediated inflammatory response. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166710. [PMID: 37054997 DOI: 10.1016/j.bbadis.2023.166710] [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: 02/08/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Cardiovascular diseases are the primary cause of mortality in patients with diabetes and obesity. Hyperglycemia and hyperlipidemia in diabetes alters cardiac function, which is associated with broader cellular processes such as aberrant inflammatory signaling. Recent studies have shown that a pattern recognition receptor called Dectin-1, expressed on macrophages, mediates pro-inflammatory responses in innate immunity. In the present study, we examined the role of Dectin-1 in the pathogenesis of diabetic cardiomyopathy. We observed increased Dectin-1 expression in heart tissues of diabetic mice and localized the source to macrophages. We then investigated the cardiac function in Dectin-1-deficient mice with STZ-induced type 1 diabetes and high-fat-diet-induced type 2 diabetes. Our results show that Dectin-1 deficient mice are protected against diabetes-induced cardiac dysfunction, cardiomyocyte hypertrophy, tissue fibrosis, and inflammation. Mechanistically, our studies show that Dectin-1 is important for cell activation and induction of inflammatory cytokines in high-concentration glucose and palmitate acid (HG + PA)-challenged macrophages. Deficiency of Dectin-1 generate fewer paracrine inflammatory factors capable of causing cardiomyocyte hypertrophy and fibrotic responses in cardiac fibroblasts. In conclusion, this study provides evidence that Dectin-1 mediates diabetes-induced cardiomyopathy through regulating inflammation. Dectin-1 may be a potential target to combat diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Na Yang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengyang Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyun Xu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wu Luo
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Peiren Shan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
27
|
Fatty acids act on vascular endothelial cells and influence the development of cardiovascular disease. Prostaglandins Other Lipid Mediat 2023; 165:106704. [PMID: 36621562 DOI: 10.1016/j.prostaglandins.2023.106704] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Endothelial cells (ECs) maintain the health of blood vessels and prevent the development of cardiovascular disease (CVD). Free saturated fatty acids (FAs) induce EC damage and increase the risk of CVD by promoting arteriosclerosis. Conversely, polyunsaturated FAs (PUFAs), such as docosahexaenoic acid, are thought to suppress EC damage induced during the early stages of CVD. This review describes the effects of multiple dietary FAs on EC disorders involved in the development of CVD. The roles of FAs in atherosclerosis and CVD were analyzed by evaluating articles published in PubMed, Science Direct, and Web of Science. Saturated FAs were found to induce EC damage by reducing the production and action of EC-derived nitric oxide. Oxidative stress, inflammation, and the renin-angiotensin system were found to be involved in EC disorder. Furthermore, n-3 PUFAs were found to reduce EC dysfunction and prevent the development of EC disorder. These results indicate that FAs may affect EC failure induced during the early stages of CVD and reduce the risk of developing the disease.
Collapse
|
28
|
Jacquet A. The HDM allergen orchestra and its cysteine protease maestro: Stimulators of kaleidoscopic innate immune responses. Mol Immunol 2023; 156:48-60. [PMID: 36889186 DOI: 10.1016/j.molimm.2023.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
House dust mite (HDM) encloses an explosive cocktail of allergenic proteins sensitizing hundreds of millions of people worldwide. To date, the innate cellular and molecular mechanism(s) orchestrating the HDM-induced allergic inflammation remains partially deciphered. Understanding the kaleidoscope of HDM-induced innate immune responses is hampered by (1) the large complexity of the HDM allergome with very diverse functional bioreactivities, (2) the perpetual presence of microbial compounds (at least LPS, β-glucan, chitin) promoting as well pro-Th2 innate signaling pathways and (3) multiple cross-talks involving structural, neuronal and immune cells. The present review provides an update on the innate immune properties, identified so far, of multiple HDM allergen groups. Experimental evidence highlights the importance of HDM allergens displaying protease or lipid-binding activities on the initiation of the allergic responses. Specifically, group 1 HDM cysteine proteases are considered as the key initiators of the allergic response through their capacities to impair the epithelial barrier integrity, to stimulate the release of pro-Th2 danger-associated molecular patterns (DAMPs) in epithelial cells, to produce super-active forms of IL-33 alarmin and to mature thrombin leading to Toll-like receptor 4 (TLR4) activation. Remarkably, the recently evidenced primary sensing of cysteine protease allergens by nociceptive neurons confirms the critical role of this HDM allergen group in the early events leading to Th2 differentiation.
Collapse
Affiliation(s)
- Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
29
|
Granieri MC, Rocca C, De Bartolo A, Nettore IC, Rago V, Romeo N, Ceramella J, Mariconda A, Macchia PE, Ungaro P, Sinicropi MS, Angelone T. Quercetin and Its Derivative Counteract Palmitate-Dependent Lipotoxicity by Inhibiting Oxidative Stress and Inflammation in Cardiomyocytes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3492. [PMID: 36834186 PMCID: PMC9958705 DOI: 10.3390/ijerph20043492] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Cardiac lipotoxicity plays an important role in the pathogenesis of obesity-related cardiovascular disease. The flavonoid quercetin (QUE), a nutraceutical compound that is abundant in the "Mediterranean diet", has been shown to be a potential therapeutic agent in cardiac and metabolic diseases. Here, we investigated the beneficial role of QUE and its derivative Q2, which demonstrates improved bioavailability and chemical stability, in cardiac lipotoxicity. To this end, H9c2 cardiomyocytes were pre-treated with QUE or Q2 and then exposed to palmitate (PA) to recapitulate the cardiac lipotoxicity occurring in obesity. Our results showed that both QUE and Q2 significantly attenuated PA-dependent cell death, although QUE was effective at a lower concentration (50 nM) when compared with Q2 (250 nM). QUE decreased the release of lactate dehydrogenase (LDH), an important indicator of cytotoxicity, and the accumulation of intracellular lipid droplets triggered by PA. On the other hand, QUE protected cardiomyocytes from PA-induced oxidative stress by counteracting the formation of malondialdehyde (MDA) and protein carbonyl groups (which are indicators of lipid peroxidation and protein oxidation, respectively) and intracellular ROS generation, and by improving the enzymatic activities of catalase and superoxide dismutase (SOD). Pre-treatment with QUE also significantly attenuated the inflammatory response induced by PA by reducing the release of key proinflammatory cytokines (IL-1β and TNF-α). Similar to QUE, Q2 (250 nM) also significantly counteracted the PA-provoked increase in intracellular lipid droplets, LDH, and MDA, improving SOD activity and decreasing the release of IL-1β and TNF-α. These results suggest that QUE and Q2 could be considered potential therapeutics for the treatment of the cardiac lipotoxicity that occurs in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Concetta Granieri
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Naomi Romeo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “Gaetano Salvatore”, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
- National Institute of Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
30
|
Qian J, Liang S, Wang Q, Xu J, Huang W, Wu G, Liang G. Toll-like receptor-2 in cardiomyocytes and macrophages mediates isoproterenol-induced cardiac inflammation and remodeling. FASEB J 2023; 37:e22740. [PMID: 36583707 DOI: 10.1096/fj.202201345r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
Heart failure (HF) is the leading cause of morbidity and mortality worldwide. Activation of the innate immune system initiates an inflammatory response during cardiac remodeling induced by isoproterenol (ISO). Here, we investigated whether Toll-like receptor-2 (TLR2) mediates ISO-induced inflammation, hypertrophy, and fibrosis. TLR2 was found to be increased in the heart tissues of mouse with HF under ISO challenge. Further, cardiomyocytes and macrophages were identified as the main cellular sources of the increased TLR2 levels in the model under ISO stimulation. The effect of TLR2 deficiency on ISO-induced cardiac remodeling was determined using TLR2 knockout mice and bone marrow transplantation models. In vitro studies involving ISO-treated cultured cardiomyocytes and macrophages showed that TLR2 knockdown significantly decreased ISO-induced cell inflammation and remodeling via MAPKs/NF-κB signaling. Mechanistically, ISO significantly increased the TLR2-MyD88 interaction in the above cells in a TLR1-dependent manner. Finally, DAMPs, such as HSP70 and fibronectin 1 (FN1), were found to be released from the cells under ISO stimulation, which further activated TLR1/2-Myd88 signaling and subsequently activated pro-inflammatory cytokine expression and cardiac remodeling. In summary, our findings suggest that TLR2 may be a target for the alleviation of chronic adrenergic stimulation-associated HF. In addition, this paper points out the possibility of TLR2 as a new target for heart failure under ISO stimulation.
Collapse
Affiliation(s)
- Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shiqi Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qinyan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiachen Xu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weijian Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
31
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
32
|
Wu Q, Chen Z, Wu C, Zhang L, Wu Y, Liu X, Wang Y, Zhang Z. MD2 Inhibits Choroidal Neovascularization via Antagonizing TLR4/MD2 Mediated Signaling Pathway. Curr Eye Res 2023; 48:474-484. [PMID: 36591949 DOI: 10.1080/02713683.2022.2164780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE To explore the pathological mechanism of Toll-like receptor 4 (TLR4) mediating neovascular age-related macular degeneration (nAMD) and the potential role of the TLR4 coreceptor myeloid differentiation protein 2 (MD2). METHODS In the study, we inhibited MD2 with the chalcone derivative L2H17 and we utilized a laser-induced choroidal neovascularization (CNV) mouse model and Tert-butyl hydroperoxide (TBHP)-challenged rhesus choroid-retinal endothelial (RF/6A) cells to assess the effect of MD2 blockade on CNV. RESULTS Inhibiting MD2 with L2H17 reduced angiogenesis in CNV mice, and significantly protected against retinal dysfunction. In retina and choroid/retinal pigment epithelium (RPE) tissues, L2H17 reduced phospho-ERK, phospho-P65 but not phospho-P38, phospho-JNK, and reduced the transcriptional levels of IL-6, TNF-α, ICAM-1 but not VCAM-1. L2H17 could protect RF/6A against TBHP-induced inflammation, oxidative stress, and apoptosis, via inhibiting the TLR4/MD2 signaling pathway and the following downstream mitogen-activated protein kinase (MAPK) and nuclear transcription factor-κB (NF-κB) activation. CONCLUSIONS Inhibiting MD2 with L2H17 significantly reduced CNV, suppressed inflammation, and oxidative stress by antagonizing TLR4/MD2 pathway in an MD2-dependent manner. MD2 may be a potential therapeutic target and L2H17 may offer an alternative treatment strategy for nAMD.
Collapse
Affiliation(s)
- Qi Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health P. R. China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, China
| | - Zhang Chen
- The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenxin Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health P. R. China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, China
| | - Lingxi Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuyang Wu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health P. R. China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, China
| | - Xiyuan Liu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health P. R. China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zongduan Zhang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health P. R. China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, China
| |
Collapse
|
33
|
Role of Myeloid Cell-Specific TLR9 in Mitochondrial DNA-Induced Lung Inflammation in Mice. Int J Mol Sci 2023; 24:ijms24020939. [PMID: 36674451 PMCID: PMC9864555 DOI: 10.3390/ijms24020939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/17/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
Mitochondrial dysfunction is common in various pathological conditions including obesity. Release of mitochondrial DNA (mtDNA) during mitochondrial dysfunction has been shown to play a role in driving the pro-inflammatory response in leukocytes including macrophages. However, the mechanisms by which mtDNA induces leukocyte inflammatory responses in vivo are still unclear. Moreover, how mtDNA is released in an obese setting has not been well understood. By using a mouse model of TLR9 deficiency in myeloid cells (e.g., macrophages), we found that TLR9 signaling in myeloid cells was critical to mtDNA-mediated pro-inflammatory responses such as neutrophil influx and chemokine production. mtDNA release by lung macrophages was enhanced by exposure to palmitic acid (PA), a major saturated fatty acid related to obesity. Moreover, TLR9 contributed to PA-mediated mtDNA release and inflammatory responses. Pathway analysis of RNA-sequencing data in TLR9-sufficient lung macrophages revealed the up-regulation of axon guidance molecule genes and down-regulation of metabolic pathway genes by PA. However, in TLR9-deficient lung macrophages, PA down-regulated axon guidance molecule genes, but up-regulated metabolic pathway genes. Our results suggest that mtDNA utilizes TLR9 signaling in leukocytes to promote lung inflammatory responses in hosts with increased PA. Moreover, TLR9 signaling is involved in the regulation of axon guidance and metabolic pathways in lung macrophages exposed to PA.
Collapse
|
34
|
Cardioprotective Effects of Aconite in Isoproterenol-Induced Myocardial Infarction in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1090893. [PMID: 36600948 PMCID: PMC9807305 DOI: 10.1155/2022/1090893] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/27/2022]
Abstract
Background Myocardial infarction (MI) is a severe clinical condition caused by decreased or complete cessation of blood flow to a portion of the myocardium. Aconite, the lateral roots of Aconitum carmichaelii Debx., is a well-known Chinese medicine for treatment of heart failure and related cardiac diseases. The present study is aimed at investigating the cardioprotective effect of aconite on isoproterenol- (ISO)- induced MI. Methods The qualitative analysis of aqueous extracts from brained aconite (AEBA) was conducted by HPLC. A rat model of MI induced by ISO was established to examine the effects of AEBA. The cardiac function was assessed by echocardiography. The serum levels of SOD, CK-MB, cTnT, and cTnI were detected to estimate myocardial injury. The pathological changes of heart tissue were evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (HE) staining, and Masson's trichrome staining. The expressions of abnormal vascular remodeling and hypoxia-related components and the levels of inflammation-associated genes and proteins were detected by RT-qPCR, western blotting, and immunofluorescence. Results The contents of benzoylaconine, benzoylmesaconine, benzoylhypacoitine, and hypaconitine in AEBA were 1.35 μg/g, 37.35 μg/g, 57.10 μg/g, and 2.46 μg/g, respectively. AEBA obviously improved heart function through promoting echocardiographic parameters, radial strain, and circumferential strain. The data of TTC staining, HE staining, and Masson's trichrome staining disclosed that AEBA could significantly reduce infarct size, inhibit inflammatory cell infiltration, and decrease the myocardial fibrosis. Moreover, AEBA distinctly suppressed the serum levels of SOD, MDA, CK-MB, cTnT, and cTnI in ISO-induced rats. The results of RT-qPCR indicated that AEBA inhibited the expressions of hypoxia- and inflammation-related genes, including VEGF, PKM2, GLUT-1, LDHA, TNF-α, IL-1β, IL-6, and COX2. In addition, the western blotting and immunofluorescence analyses further confirmed the results of RT-qPCR. Conclusion In summary, our results indicate that the AEBA could improve ISO-induced myocardial infarction by promoting cardiac function, alleviating myocardial hypoxia, and inhibiting inflammatory response and fibrosis in heart tissue.
Collapse
|
35
|
Zhou J, Lin Y, Yang X, Shen B, Hao J, Wang J, Wang J. Metabolic disorders sensitise endometrial carcinoma through endoplasmic reticulum stress. Cell Mol Biol Lett 2022; 27:110. [PMID: 36526973 PMCID: PMC9756454 DOI: 10.1186/s11658-022-00412-x] [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: 10/09/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Metabolic disorder is considered a well-established risk factor for endometrial carcinoma (EC). However, the mechanism remains unclear. Insulin resistance and excessive flux of free fatty acids serve as fundamental pathogenic factors in metabolic disorders, including obesity and type 2 diabetes. The aim of this study was to test the correlation between insulin resistance and dyslipidaemia in EC and to determine the effect of insulin and saturated fatty acids on EC cells. METHODS A retrospective study on the medical records of patients with EC and RNA-seq from the TCGA database analysed with edgR and Gene Ontology (GO) were used to assess the correlation of dyslipidaemia and diabetes as well as obesity. Crystal violet assays and CCK-8 assays were used to detect the proliferation of EC cells, and Annexin V-PI was used to examine apoptosis. Transient changes in mitochondrial Ca2+ and reactive oxygen species (ROS) were monitored via confocal microscopy. DNA damage was assessed by comet assays. Changes in signalling pathways were detected via phospho-kinase array. western blotting was used to assess the molecular changes in endoplasmic reticulum (ER) stress and DNA damage. RESULTS We found that glucose metabolism disorders accompanied dyslipidaemia in patients with EC. As a key regulator of glucose metabolism disorders, insulin promoted DNA damage, ROS and Ca2+ homoeostasis imbalance in a panel of established EC cell lines. Interestingly, excessive insulin boosted saturated fatty acid-induced pro-apoptotic effects in EC cells. Furthermore, our data showed that insulin synergised with saturated fatty acids to activate the mechanistic target of rapamycin kinase/70 kDa ribosomal protein S6 kinase (mTOR/p70S6K) pathway and ER stress, resulting in Ca2+ release from ER and unfolded protein response (UPR) activation, which contributed to combined insulin and saturated fatty acid treatment-induced apoptosis and tumour progression. CONCLUSIONS Our data are the first to illustrate that impaired glucose metabolism accelerates dyslipidaemia-promoted EC progression, which is attributed to hyperinsulinaemia and saturated fatty acid-induced Ca2+ dyshomoeostasis and UPR activation in EC cells via ER stress.
Collapse
Affiliation(s)
- Jingyi Zhou
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China
| | - Yanying Lin
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China ,grid.256112.30000 0004 1797 9307Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China ,grid.459516.aFujian Key Laboratory of Women and Children’s Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Xiao Yang
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China
| | - Boqiang Shen
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China
| | - Juan Hao
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China
| | - Jiaqi Wang
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China
| | - Jianliu Wang
- grid.411634.50000 0004 0632 4559Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, 100044 China ,Beijing Key Laboratory of Female Pelvic Floor Disorders Disease, Beijing, 100044 China
| |
Collapse
|
36
|
Luo W, Wu G, Chen X, Zhang Q, Zou C, Wang J, Liu J, Chattipakorn N, Wang Y, Liang G. Blockage of MyD88 in cardiomyocytes alleviates cardiac inflammation and cardiomyopathy in experimental diabetic mice. Biochem Pharmacol 2022; 206:115292. [DOI: 10.1016/j.bcp.2022.115292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022]
|
37
|
Huang D, Wang P, Chen J, Li Y, Zhu M, Tang Y, Zhou W. Selective targeting of MD2 attenuates intestinal inflammation and prevents neonatal necrotizing enterocolitis by suppressing TLR4 signaling. Front Immunol 2022; 13:995791. [PMID: 36389716 PMCID: PMC9663461 DOI: 10.3389/fimmu.2022.995791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/14/2022] [Indexed: 10/17/2023] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) is an inflammatory disease that occurs in premature infants and has a high mortality rate; however, the mechanisms behind this disease remain unclear. The TLR4 signaling pathway in intestinal epithelial cells, mediated by TLR4, is important for the activation of the inflammatory storm in NEC infants. Myeloid differentiation protein 2 (MD2) is a key auxiliary component of the TLR4 signaling pathway. In this study, MD2 was found to be significantly increased in intestinal tissues of NEC patients at the acute stage. We further confirmed that MD2 was upregulated in NEC rats. MD2 inhibitor (MI) pretreatment reduced the occurrence and severity of NEC in neonatal rats, inhibited the activation of NF-κB and the release of inflammatory molecules (TNF-α and IL-6), and reduced the severity of intestinal injury. MI pretreatment significantly reduced enterocyte apoptosis while also maintaining tight junction proteins, including occludin and claudin-1, and protecting intestinal mucosal permeability in NEC rats. In addition, an NEC in vitro model was established by stimulating IEC-6 enterocytes with LPS. MD2 overexpression in IEC-6 enterocytes significantly activated NF-κB. Further, both MD2 silencing and MI pretreatment inhibited the inflammatory response. Overexpression of MD2 increased damage to the IEC-6 monolayer cell barrier, while both MD2 silencing and MI pretreatment played a protective role. In conclusion, MD2 triggers an inflammatory response through the TLR4 signaling pathway, leading to intestinal mucosal injury in NEC. In addition, MI alleviates inflammation and reduces intestinal mucosal injury caused by the inflammatory response by blocking the TLR4-MD2/NF-κB signaling axis. These results suggest that inhibiting MD2 may be an important way to prevent NEC.
Collapse
Affiliation(s)
- Dabin Huang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Juncao Chen
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanbin Li
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Tang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Chen J, Peng H, Chen C, Wang Y, Sang T, Cai Z, Zhao Q, Chen S, Lin X, Eling T, Wang X. NAG-1/GDF15 inhibits diabetic nephropathy via inhibiting AGE/RAGE-mediated inflammation signaling pathways in C57BL/6 mice and HK-2 cells. Life Sci 2022; 311:121142. [DOI: 10.1016/j.lfs.2022.121142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022]
|
39
|
Yang B, Luo W, Wang M, Tang Y, Zhu W, Jin L, Wang M, Wang Y, Zhang Y, Zuo W, Huang LJ, Zhao Y, Liang G. Macrophage-specific MyD88 deletion and pharmacological inhibition prevents liver damage in non-alcoholic fatty liver disease via reducing inflammatory response. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166480. [PMID: 35811033 DOI: 10.1016/j.bbadis.2022.166480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/14/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023]
Abstract
Activation of the innate immune system through toll-like receptors (TLRs) has been repeatedly demonstrated in non-alcoholic fatty liver disease (NAFLD) and several TLRs have been shown to contribute. Myeloid differentiation primary response 88 (MyD88) is as an adapter protein for the activation of TLRs and bridges TLRs to NF-κB-mediated inflammation in macrophages. However, whether myeloid cell MyD88 contributes to NAFLD are largely unknown. To test this approach, we generated macrophage-specific MyD88 knockout mice and show that these mice are protected against high-fat diet (HFD)-induced hepatic injury, lipid accumulation, and fibrosis. These protective effects were associated with reduced macrophage numbers in liver tissues and surpassed inflammatory responses. In cultured macrophages, saturated fatty acid palmitate utilizes MyD88 to activate NF-κB and induce inflammatory and fibrogenic factors. In hepatocytes, these factors may cause lipid accumulation and a further elaboration of inflammatory cytokines. In hepatic stellate cells, macrophage-derived factors, especially TGF-β, cause activation and hepatic fibrosis. We further show that pharmacological inhibition of MyD88 is also able to reduce NAFLD injury in HFD-fed mice. Therefore, our study has provided empirical evidence that macrophage MyD88 participates in HFD-induced NAFLD and could be targeted to prevent the development and progression of NAFLD/NASH.
Collapse
Affiliation(s)
- Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yelin Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Weiwei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Leiming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Meihong Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Wei Zuo
- Affiliated Xiangshan Hospital of Wenzhou Medial University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, Zhejiang 315799, China
| | - Li-Jiang Huang
- Affiliated Xiangshan Hospital of Wenzhou Medial University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, Zhejiang 315799, China
| | - Yunjie Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
40
|
Lin K, Yang N, Luo W, Qian JF, Zhu WW, Ye SJ, Yuan CX, Xu DY, Liang G, Huang WJ, Shan PR. Direct cardio-protection of Dapagliflozin against obesity-related cardiomyopathy via NHE1/MAPK signaling. Acta Pharmacol Sin 2022; 43:2624-2635. [PMID: 35217813 PMCID: PMC9525284 DOI: 10.1038/s41401-022-00885-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/06/2022] [Indexed: 12/22/2022] Open
Abstract
Obesity is an important independent risk factor for cardiovascular diseases, remaining an important health concern worldwide. Evidence shows that saturated fatty acid-induced inflammation in cardiomyocytes contributes to obesity-related cardiomyopathy. Dapagliflozin (Dapa), a selective SGLT2 inhibitor, exerts a favorable preventive activity in heart failure. In this study, we investigated the protective effect of Dapa against cardiomyopathy caused by high fat diet-induced obesity in vitro and in vivo. Cultured rat cardiomyocyte H9c2 cells were pretreated with Dapa (1, 2.5 μM) for 1.5 h, followed by treatment with palmitic acid (PA, 200 μM) for 24 h. We showed that Dapa pretreatment concentration-dependently attenuated PA-induced cell hypertrophy, fibrosis and apoptosis. Transcriptome analysis revealed that inhibition of PA-activated MAPK/AP-1 pathway contributed to the protective effect of Dapa in H9c2 cells, and this was confirmed by anti-p-cJUN fluorescence staining assay. Using surface plasmon resonance analysis we found the direct binding of Dapa with NHE1. Gain and loss of function experiments further demonstrated the role of NHE1 in the protection of Dapa. In vivo experiments were conducted in mice fed a high fat diet for 5 months. The mice were administered Dapa (1 mg·kg-1·d-1, i.g.) in the last 2 months. Dapa administration significantly reduced the body weight and improved the serum lipid profiles. Dapa administration also alleviated HFD-induced cardiac dysfunction and cardiac aberrant remodeling via inhibiting MAPK/AP-1 pathway and ameliorating cardiac inflammation. In conclusion, Dapa exerts a direct protective effect against saturated fatty acid-induced cardiomyocyte injury in addition to the lowering effect on serum lipids. The protective effect results from negative regulating MAPK/AP-1 pathway in a NHE1-dependent way. The current study highlights the potential of clinical use of Dapa in the prevention of obesity-related cardiac dysfunction.
Collapse
Affiliation(s)
- Ke Lin
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Na Yang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jin-Fu Qian
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Wei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shi-Ju Ye
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen-Xin Yuan
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Di-Yun Xu
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei-Jian Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Pei-Ren Shan
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
41
|
Lin J, Lin K, Huang L, Jiang Y, Ding X, Luo W, Samorodov AV, Pavlov VN, Liang G, Qian J, Wang Y. Heme induces inflammatory injury by directly binding to the complex of myeloid differentiation protein 2 and toll-like receptor 4. Toxicol Lett 2022; 370:15-23. [PMID: 36115635 DOI: 10.1016/j.toxlet.2022.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/20/2022] [Accepted: 09/12/2022] [Indexed: 10/31/2022]
Abstract
Heme, as an essential component of hemoproteins, is a prosthetic co-factor found in many cells, which is essential for physiologically vital oxygen transport. However, extracellular or circulatory heme is cytotoxic and triggers inflammation. Although the proinflammatory role of heme has been reported to be associated with Toll-like receptor 4 (TLR4) signaling, the exact mechanism remains unknown. Here, we show that heme promotes TLR4 signaling and inflammation via directly physically interacting with TLR4 and its adaptor protein myeloid differentiation protein 2 (MD2). Genetic loss of MD2 ameliorates heme-induced inflammation and inflammatory cytokine production in the spleen of MD2 knockout (MD2-/-) mice. Using mouse macrophage RAW 264.7 cell line, we show that heme induces TLR4 dimerization and MD2/TLR4/MyD88 activation by physically interacting with TLR4 and MD2 in vitro. Genetic loss of MD2 inhibits heme-induced inflammation and MAPK/NF-κB pathway in mouse primary macrophages extracted from MD2-/- mice. Furthermore, pharmacological inhibition of MD2 by L6H9 ameliorates heme-induced inflammation in macrophages. These findings demonstrate that heme causes inflammation by directly binding to MD2/TLR4 complex, leading to activation of TLR4/MAPK/NF-κB signaling pathway and production of downstream effectors of inflammation.
Collapse
Affiliation(s)
- Jianjun Lin
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lijiang Huang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Yongsheng Jiang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Xiaoxiao Ding
- Department of pharmacy, the People' s Hospital of Beilun District , Ningbo, Zhejiang 315807, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Aleksandr V Samorodov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Valentin N Pavlov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yi Wang
- The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
42
|
Gan AM, Tracz-Gaszewska Z, Ellert-Miklaszewska A, Navrulin VO, Ntambi JM, Dobrzyn P. Stearoyl-CoA Desaturase Regulates Angiogenesis and Energy Metabolism in Ischemic Cardiomyocytes. Int J Mol Sci 2022; 23:ijms231810459. [PMID: 36142371 PMCID: PMC9499489 DOI: 10.3390/ijms231810459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
New blood vessel formation is a key component of the cardiac repair process after myocardial infarction (MI). Hypoxia following MI is a major driver of angiogenesis in the myocardium. Hypoxia-inducible factor 1α (HIF1α) is the key regulator of proangiogenic signaling. The present study found that stearoyl-CoA desaturase (SCD) significantly contributed to the induction of angiogenesis in the hypoxic myocardium independently of HIF1α expression. The pharmacological inhibition of SCD activity in HL-1 cardiomyocytes and SCD knockout in an animal model disturbed the expression and secretion of proangiogenic factors including vascular endothelial growth factor-A, proinflammatory cytokines (interleukin-1β, interleukin-6, tumor necrosis factor α, monocyte chemoattractant protein-1, and Rantes), metalloproteinase-9, and platelet-derived growth factor in ischemic cardiomyocytes. These disturbances affected the proangiogenic potential of ischemic cardiomyocytes after SCD depletion. Together with the most abundant SCD1 isoform, the heart-specific SCD4 isoform emerged as an important regulator of new blood vessel formation in the murine post-MI myocardium. We also provide evidence that SCD shapes energy metabolism of the ischemic heart by maintaining the shift from fatty acids to glucose as the substrate that is used for adenosine triphosphate production. Furthermore, we propose that the regulation of the proangiogenic properties of hypoxic cardiomyocytes by key modulators of metabolic signaling such as adenosine monophosphate kinase, protein kinase B (AKT), and peroxisome-proliferator-activated receptor-γ coactivator 1α/peroxisome proliferator-activated receptor α depends on SCD to some extent. Thus, our results reveal a novel mechanism that links SCD to cardiac repair processes after MI.
Collapse
Affiliation(s)
- Ana-Maria Gan
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Zuzanna Tracz-Gaszewska
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Aleksandra Ellert-Miklaszewska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Viktor O. Navrulin
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - James M. Ntambi
- Departments of Biochemistry and Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
- Correspondence:
| |
Collapse
|
43
|
Xue C, Xie Q, Zhang C, Hu Y, Song X, Jia Y, Shi X, Chen Y, Liu Y, Zhao L, Huang F, Yuan H. Vertical transmission of the gut microbiota influences glucose metabolism in offspring of mice with hyperglycaemia in pregnancy. MICROBIOME 2022; 10:122. [PMID: 35941695 PMCID: PMC9361546 DOI: 10.1186/s40168-022-01318-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/30/2022] [Indexed: 05/21/2023]
Abstract
BACKGROUND Hyperglycaemia in pregnancy (HIP) is a common metabolic disorder that not only poses risks to maternal health but also associates with an increased risk of diabetes among offspring. Vertical transmission of microbiota may influence the offspring microbiome and subsequent glucose metabolism. However, the mechanism by which maternal gut microbiota may influence glucose metabolism of the offspring remains unclear and whether intervening microbiota vertical transmission could be used as a strategy to prevent diabetes in the offspring of mothers with HIP has not been investigated. So we blocked vertical transmission to investigate its effect on glucose metabolism in the offspring. RESULTS We established a murine HIP model with a high-fat diet (HFD) and investigated the importance of vertical transmission of gut microbiota on the glucose metabolism of offspring via birth and nursing by blocking these events through caesarean section (C-section) and cross-fostering. After weaning, all offspring were fed a normal diet. Based on multi-omics analysis, biochemical and transcriptional assays, we found that the glucometabolic deficits in the mothers were subsequently 'transmitted' to the offspring. Meanwhile, the partial change in mothers' gut microbial community induced by HIP could be transmitted to offspring, supported by the closed clustering of the microbial structure and composition between the offspring and their mothers. Further study showed that the microbiota vertical transmission was blocked by C-section and cross-fostering, which resulted in improved insulin sensitivity and islet function of the offspring of the mothers with HIP. These effects were correlated with changes in the relative abundances of specific bacteria and their metabolites, such as increased relative abundances of Bifidobacterium and short-chain fatty acids. In particular, gut microbial communities of offspring were closely related to those of their foster mothers but not their biological mothers, and the effect of cross-fostering on the offspring's gut microbiota was more profound than that of C-section. CONCLUSION Our study demonstrates that the gut microbiota transmitted via birth and nursing are important contributors to the glucose metabolism phenotype in offspring. Video Abstract.
Collapse
Affiliation(s)
- Cunxi Xue
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qinyuan Xie
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yimeng Hu
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoting Song
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yifan Jia
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyang Shi
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiqi Chen
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yalei Liu
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingyun Zhao
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fenglian Huang
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huijuan Yuan
- Department of Endocrinology of Henan Provincial People's Hospital, Henan Provincial Key Laboratory of Intestinal Microecology and Diabetes Control, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
44
|
Zhou X, Li S, Zhou Y, Zhang H, Yan B, Wang H, Xiao Y. A metabolomics study of the intervention effect of Tartary buckwheat on hyperlipidemia mice. J Food Biochem 2022; 46:e14359. [DOI: 10.1111/jfbc.14359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/06/2022] [Accepted: 07/18/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Xiaoli Zhou
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
- Institute of Beautiful China and Ecological Civilization University Think Tank of Shanghai Municipality Shanghai China
| | - Senjie Li
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
| | - Yiming Zhou
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
- Department of Food Science and Engineering Shanghai Institute of Technology Shanghai P. R. China
| | - Huan Zhang
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
| | - Beibei Yan
- Institute of Beautiful China and Ecological Civilization University Think Tank of Shanghai Municipality Shanghai China
| | - Hong Wang
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
| | - Ying Xiao
- School of Perfume and Aroma Technology Shanghai Institute of Technology Shanghai China
| |
Collapse
|
45
|
Romić S, Tepavčević S, Popović T, Zec M, Stojiljković M, Ćulafić T, Bošković M, Korićanac G. Consumption of walnuts suppresses the conversion of palmitic to palmitoleic acid and enhances omega-3 fatty acid metabolism in the heart of fructose-fed rats. Int J Food Sci Nutr 2022; 73:940-953. [PMID: 35918845 DOI: 10.1080/09637486.2022.2107186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Walnut consumption mostly has a positive implication for cardiovascular health. Walnut diet effects on the cardiac fatty acid (FA) metabolism of healthy rats and those with fructose diet-induced metabolic burden were analysed. Both walnuts and fructose increased CD36 transporter level and the nuclear content of some/all of Lipin 1/PPARα/PGC-1 complex partners, as well as cytosolic and nuclear FOXO1. However, fructose, independently of walnuts, increased the content of palmitic (PA), oleic, and vaccenic acid (VA), while in walnut-fed rats failed to increase palmitoleic acid (POA) level and the POA/PA ratio, as well as total MUFA content. In opposite, walnuts reduced the level of PA and VA and increased alpha-linolenic, eicosapentaenoic and docosapentaenoic acid level, regardless of fructose. In conclusion, both fructose and walnuts stimulated the uptake and oxidation of FA in the heart, but the walnuts, opposite to fructose, favourably altered cardiac FA profile in healthy and metabolically compromised rats.
Collapse
Affiliation(s)
- Snježana Romić
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Snežana Tepavčević
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Tamara Popović
- Centre of Excellence in Nutrition and Metabolism Research, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Manja Zec
- Centre of Excellence in Nutrition and Metabolism Research, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Serbia.,School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA
| | - Mojca Stojiljković
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Tijana Ćulafić
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Maja Bošković
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| | - Goran Korićanac
- Laboratory for Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Serbia
| |
Collapse
|
46
|
Najjar RS, Knapp D, Wanders D, Feresin RG. Raspberry and blackberry act in a synergistic manner to improve cardiac redox proteins and reduce NF-κB and SAPK/JNK in mice fed a high-fat, high-sucrose diet. Nutr Metab Cardiovasc Dis 2022; 32:1784-1796. [PMID: 35487829 DOI: 10.1016/j.numecd.2022.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS Increased cardiac inflammation and oxidative stress are common features in obesity, and toll-like receptor (TLR)4 signaling is a key inflammatory pathway in this deleterious process. This study aimed to investigate whether berries could attenuate the detrimental effects of a high-fat, high-sucrose (HFHS) diet on the myocardium at the molecular level. METHODS AND RESULTS Eight-week-old male C57BL/6 mice consumed a low-fat, low-sucrose (LFLS) diet alone or supplemented with 10% blackberry (BL), 10% raspberry (RB) or 10% blackberry + raspberry (BL + RB) for four weeks. Animals were then switched to a HFHS diet for 24 weeks with or without berry supplementation or maintained on a LFLS control diet without berry supplementation. Left ventricles of the heart were isolated for protein and mRNA analysis. Berry consumption, particularly BL + RB reduced NADPH-oxidase (NOX)1 and NOX2 and increased catalase (CAT) and superoxide dismutase (SOD)2, expression while BL and RB supplementation alone was less efficacious. Downstream TLR4 signaling was attenuated mostly by both RB and BL + RB supplementation, while NF-κB pathway was attenuated by BL + RB supplementation. Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) was also attenuated by BL + RB supplementation, and reduced TNF-α transcription and protein expression was observed only with BL + RB supplementation. CONCLUSION The synergistic effects of BL + RB may reduce obesity-induced cardiac inflammation and oxidative stress to a greater extent than BL or RB alone.
Collapse
Affiliation(s)
- Rami S Najjar
- Department of Nutrition, Georgia State University, Atlanta, GA, USA
| | - Denise Knapp
- Department of Nutrition, Georgia State University, Atlanta, GA, USA; Department of Exercise Science and Sport Management, Kennesaw State University, Kennesaw, GA, USA
| | - Desiree Wanders
- Department of Nutrition, Georgia State University, Atlanta, GA, USA
| | - Rafaela G Feresin
- Department of Nutrition, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
47
|
Han XD, Jiang XG, Yang M, Chen WJ, Li LG. miRNA‑124 regulates palmitic acid‑induced epithelial‑mesenchymal transition and cell migration in human retinal pigment epithelial cells by targeting LIN7C. Exp Ther Med 2022; 24:481. [PMID: 35761801 PMCID: PMC9214593 DOI: 10.3892/etm.2022.11408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/25/2022] [Indexed: 12/02/2022] Open
Abstract
The present study revealed that palmitic acid (PA) treatment induced epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells, which are involved in the progression of proliferative vitreoretinopathy (PVR). ARPE-19 cells were treated with PA followed by miRNA screening and EMT marker detection using qRT-PCR. Then, miR-124 mimic or inhibitor was transfected into ARPE-19 cells to explore the role of miR-124 on the EMT of ARPE-19 cells using transwell assay. The underlying mechanism of miRNA were predicted by bioinformatics method and confirmed by luciferase activity reporter assay. Furthermore, gain-of-function strategy was also used to explore the role of LIN7C in the EMT of ARPE-19 cells. The expression of miRNA or mRNA expression was determined by qRT-PCR and the protein expression was determined using western blot assay. The result presented that PA reduced the expression of E-cadherin/ZO-1 whilst increasing the expression of fibronectin/α-SMA. In addition, PA treatment enhanced the expression of microRNA (miR)-124 in ARPE-19 cells. Overexpression of miR-124 enhanced PA-induced upregulation of E-cadherin and ZO-1 expression and downregulation of fibronectin and α-SMA. Moreover, miR-124 mimic also enhanced the migration of ARPE-19 cells induced by PA treatment. Inversely, miR-124 inhibitor presented opposite effect on PA-induced EMT and cell migration in ARPE-19 cells. Luciferase activity reporter assay confirmed that Lin-7 homolog C (LIN7C) was a direct target of miR-124 in ARPE-19 cells. Overexpression of LIN7C was found to suppress the migration ability and expression of fibronectin and α-SMA, while increasing expression of E-cadherin and ZO-1; miR-124 mimic abrogated the inhibitive effect of LIN7C on the EMT of ARPE-19 cells and PA further enhanced this abolishment. Collectively, these findings suggest that miR-124/LIN7C can modulate EMT and cell migration in RPE cells, which may have therapeutic implications in the management of PVR diseases.
Collapse
Affiliation(s)
- Xiao-Dong Han
- Department of Ocular Fundus Diseases, Xi'an Aier Ancient City Eye Hospital, Xi'an, Shaanxi 710082, P.R. China
| | - Xu-Guang Jiang
- Department of Ocular Fundus Diseases, Xi'an Aier Ancient City Eye Hospital, Xi'an, Shaanxi 710082, P.R. China
| | - Min Yang
- Department of Ocular Fundus Diseases, Xi'an Aier Ancient City Eye Hospital, Xi'an, Shaanxi 710082, P.R. China
| | - Wen-Jun Chen
- Department of Ocular Fundus Diseases, Xi'an Aier Ancient City Eye Hospital, Xi'an, Shaanxi 710082, P.R. China
| | - Li-Gang Li
- Department of Cataracts, Xi'an Aier Ancient City Eye Hospital, Xi'an, Shaanxi 710082, P.R. China
| |
Collapse
|
48
|
Dietary Fatty Acid Regulation of the NLRP3 Inflammasome via the TLR4/NF-κB Signaling Pathway Affects Chondrocyte Pyroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3711371. [PMID: 35571243 PMCID: PMC9095358 DOI: 10.1155/2022/3711371] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Dietary fatty acid (FA) content and type have different effects on obesity-associated osteoarthritis (OA), but the mechanisms underlying these differences are not fully understood. Inflammation activated by toll-like receptor 4 (TLR4)/nuclear factor- (NF-) κB signaling and pyroptosis induced by the NLRP3/caspase-1/gasdermin D (GSDMD) signaling pathway play important roles in OA development. Our aim in this study was to observe the effects of dietary FAs on the articular cartilage of obese post-traumatic OA model mice and on chondrocytes stimulated by lipopolysaccharide (LPS) and to determine whether the underlying mechanisms involve TLR4/NF-κB and NLRP3/caspase-1/GSDMD signaling pathways. Mice were fed high-fat diets rich in various FAs and underwent surgical destabilization of the medial meniscus to establish the obesity-related post-traumatic OA model. LPS-induced SW1353 chondrosarcoma cells were used to mimic OA status in vitro, and TLR4 inhibitors or TLR4 overexpressing lentivirus was administered. Analysis using weight-matched mice and multiple regression models revealed that OA was associated with dietary FA content and serum inflammatory factor levels, but not body weight. Diets rich in n-3 polyunsaturated fatty acids (PUFAs) attenuated OA and inhibited the TLR4/NF-κB and NLRP3/caspase-1/GSDMD signaling pathways, whereas diets rich in saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs), or n-6 PUFAs increased OA severity and activated these pathways. In vitro results for SFAs, n-6 PUFAs, and n-3 PUFAs were consistent with the animal experiments. However, those for MUFAs were not. FA effects on the NLRP3/caspase-1/GSDMD pathway were associated with the inhibition or activation of the TLR4 signaling pathway. In conclusion, diets rich in SFAs or n-6 PUFAs can exacerbate obesity-associated OA, whereas those rich in n-3 PUFAs have protective effects against this disease, due to their respective pro-/anti-inflammatory and pyroptotic effects. Further research on dietary FA supplements as a potential therapeutic approach for OA is needed.
Collapse
|
49
|
Guan B, Tong J, Hao H, Yang Z, Chen K, Xu H, Wang A. Bile acid coordinates microbiota homeostasis and systemic immunometabolism in cardiometabolic diseases. Acta Pharm Sin B 2022; 12:2129-2149. [PMID: 35646540 PMCID: PMC9136572 DOI: 10.1016/j.apsb.2021.12.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 02/08/2023] Open
Abstract
Cardiometabolic disease (CMD), characterized with metabolic disorder triggered cardiovascular events, is a leading cause of death and disability. Metabolic disorders trigger chronic low-grade inflammation, and actually, a new concept of metaflammation has been proposed to define the state of metabolism connected with immunological adaptations. Amongst the continuously increased list of systemic metabolites in regulation of immune system, bile acids (BAs) represent a distinct class of metabolites implicated in the whole process of CMD development because of its multifaceted roles in shaping systemic immunometabolism. BAs can directly modulate the immune system by either boosting or inhibiting inflammatory responses via diverse mechanisms. Moreover, BAs are key determinants in maintaining the dynamic communication between the host and microbiota. Importantly, BAs via targeting Farnesoid X receptor (FXR) and diverse other nuclear receptors play key roles in regulating metabolic homeostasis of lipids, glucose, and amino acids. Moreover, BAs axis per se is susceptible to inflammatory and metabolic intervention, and thereby BAs axis may constitute a reciprocal regulatory loop in metaflammation. We thus propose that BAs axis represents a core coordinator in integrating systemic immunometabolism implicated in the process of CMD. We provide an updated summary and an intensive discussion about how BAs shape both the innate and adaptive immune system, and how BAs axis function as a core coordinator in integrating metabolic disorder to chronic inflammation in conditions of CMD.
Collapse
Key Words
- AS, atherosclerosis
- ASBT, apical sodium-dependent bile salt transporter
- BAs, bile acids
- BSEP, bile salt export pump
- BSH, bile salt hydrolases
- Bile acid
- CA, cholic acid
- CAR, constitutive androstane receptor
- CCs, cholesterol crystals
- CDCA, chenodeoxycholic acid
- CMD, cardiometabolic disease
- CVDs, cardiovascular diseases
- CYP7A1, cholesterol 7 alpha-hydroxylase
- CYP8B1, sterol 12α-hydroxylase
- Cardiometabolic diseases
- DAMPs, danger-associated molecular patterns
- DCA, deoxycholic acid
- DCs, dendritic cells
- ERK, extracellular signal-regulated kinase
- FA, fatty acids
- FFAs, free fatty acids
- FGF, fibroblast growth factor
- FMO3, flavin-containing monooxygenase 3
- FXR, farnesoid X receptor
- GLP-1, glucagon-like peptide 1
- HCA, hyocholic acid
- HDL, high-density lipoprotein
- HFD, high fat diet
- HNF, hepatocyte nuclear receptor
- IL, interleukin
- IR, insulin resistance
- JNK, c-Jun N-terminal protein kinase
- LCA, lithocholic acid
- LDL, low-density lipoprotein
- LDLR, low-density lipoprotein receptor
- LPS, lipopolysaccharide
- NAFLD, non-alcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-κB
- NLRP3, NLR family pyrin domain containing 3
- Nuclear receptors
- OCA, obeticholic acid
- PKA, protein kinase A
- PPARα, peroxisome proliferator-activated receptor alpha
- PXR, pregnane X receptor
- RCT, reverses cholesterol transportation
- ROR, retinoid-related orphan receptor
- S1PR2, sphingosine-1-phosphate receptor 2
- SCFAs, short-chain fatty acids
- SHP, small heterodimer partner
- Systemic immunometabolism
- TG, triglyceride
- TGR5, takeda G-protein receptor 5
- TLR, toll-like receptor
- TMAO, trimethylamine N-oxide
- Therapeutic opportunities
- UDCA, ursodeoxycholic acid
- VDR, vitamin D receptor
- cAMP, cyclic adenosine monophosphate
- mTOR, mammalian target of rapamycin
- ox-LDL, oxidated low-density lipoprotein
Collapse
Affiliation(s)
- Baoyi Guan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Jinlin Tong
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhixu Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Keji Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Hao Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| | - Anlu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing 100091, China
| |
Collapse
|
50
|
Xin X, Duan L, Yang H, Yu H, Bao Y, Jia D, Wu N, Qiao Y. miR-141-3p regulates saturated fatty acid-induced cardiomyocyte apoptosis through Notch1/PTEN/AKT pathway via targeting PSEN1. ENVIRONMENTAL TOXICOLOGY 2022; 37:741-753. [PMID: 34897970 DOI: 10.1002/tox.23439] [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: 05/04/2021] [Revised: 11/30/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
It has been reported that miR-141-3p levels are markedly upregulated in the cardiomyocytes of obese rats induced by a high-fat diet. However, the role of miR-141-3p in myocardial lipotoxicity remains elusive. In the present study, the role of miR-141-3p in lipotoxic injury of H9c2 cells induced by palmitic acid (PA) and its possible mechanisms were assessed. The results indicated that miR-141-3p was significantly upregulated in PA-induced cardiomyocytes. miR-141-3p inhibitor enhanced the cell viability, reduced the release of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and troponin I (CTN-I), decreased cell apoptosis rate, and repressed the activation of mitochondrial apoptosis pathway in PA-treated H9c2, whereas treatment with miR-141-3p mimics resulted in the opposite effects. Mechanistically, it was further revealed that miR-141-3p could specifically bind to presenilin 1 (PSEN1) 3'UTR, and upregulating miR-141-3p levels reduced the expression of PSEN1, thereby inhibiting the activation of the Notch1/PTEN/AKT pathway. Additionally, inhibition of Notch1/AKT signaling pathway by its inhibitor could abrogate the effect of miR-141-3p on mitochondrial-mediated apoptosis induced by PA. In conclusion, the present study demonstrates that miR-141-3p regulates saturated fatty acid-induced cardiomyocyte apoptosis through Notch1/PTEN/AKT pathway via targeting PSEN1, which gains a new insight into the mechanisms of myocardial lipotoxic injury.
Collapse
Affiliation(s)
- Xin Xin
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Lian Duan
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Huimin Yang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Hang Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yandong Bao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Dalin Jia
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Nan Wu
- The Central Laboratory, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Ying Qiao
- The Central Laboratory, The First Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|