1
|
Dhillon J, Pandey S, Newman JW, Fiehn O, Ortiz RM. Almond consumption for 8 weeks differentially modulates metabolomic responses to an acute glucose challenge compared to crackers in young adults. Nutr Res 2025; 135:67-81. [PMID: 39965269 DOI: 10.1016/j.nutres.2025.01.003] [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: 07/08/2024] [Revised: 12/15/2024] [Accepted: 01/05/2025] [Indexed: 02/20/2025]
Abstract
This study investigated the dynamic responses to an acute glucose challenge after 8 weeks of almond or cracker consumption (clinicaltrials.gov ID: NCT03084003). Young adults (n = 73, age: 18-19 years, BMI: 18-41 kg/m2) participated in an 8-week randomized, controlled, parallel-arm intervention and were assigned to consume either almonds (2 oz/d, n = 38) or an isocaloric control snack of graham crackers (325 kcal/d, n = 35) daily. Twenty participants from each group underwent a 2-hour oral glucose tolerance test (oGTT) at the end of the intervention. Metabolite abundances in the oGTT serum samples were quantified using untargeted metabolomics, and targeted analyses for free PUFAs, total fatty acids, oxylipins, and endocannabinoids. We hypothesized that 8-week almond consumption would differentially modulate the metabolomic response to a glucose challenge compared to crackers. Multivariate, univariate, and chemical enrichment analyses were conducted to identify significant metabolic shifts. Findings exhibit a biphasic lipid response with higher levels of unsaturated triglycerides earlier in the oGTT followed by lower levels later in the almond vs cracker group (p-value <.05, chemical enrichment analyses). Almond (vs cracker) consumption was also associated with higher AUC120 min of aminomalonate, and oxylipins (P-value <.05), but lower AUC120 min of l-cystine, N-acetylmannosamine, and isoheptadecanoic acid (P-value <.05). Additionally, the Matsuda Index in the almond group correlated with AUC120 min of CE 22:6 (r = -0.46; P-value <.05) and 12,13 DiHOME (r = 0.45; P-value <.05). Almond consumption for 8 weeks leads to dynamic, differential shifts in response to an acute glucose challenge, marked by alterations in lipid and amino acid mediators involved in metabolic and physiological pathways.
Collapse
Affiliation(s)
- Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA; Department of Molecular and Cell Biology, University of California, Merced, CA, USA.
| | - Saurabh Pandey
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA; Jaypee University of Information Technology, Waknaghat, Himachal Pradesh, India
| | - John W Newman
- West Coast Metabolomics Center, University of California, Davis, CA, USA; Department of Nutrition, University of California, Davis, CA, USA; Obesity and Metabolism Research Unit, USDA-Agricultural Research Service Western Human Nutrition Research Center, University of California, Davis, CA, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Rudy M Ortiz
- Department of Molecular and Cell Biology, University of California, Merced, CA, USA
| |
Collapse
|
2
|
Zhang Y, Zhang X, Jin X, Li Z, Li L, Zhu Y, Kong F, Wang D. Structural characterization of polysaccharide isolated from Inonotus hispidus and its anti-obesity effect based on regulation of the interleukin-17-mediated inflammatory response. Int J Biol Macromol 2025; 291:138975. [PMID: 39706397 DOI: 10.1016/j.ijbiomac.2024.138975] [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: 06/25/2024] [Revised: 11/19/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
A heteropolysaccharide (IHP3) with a molecular weight of 22.0 kDa was isolated from Inonotus hispidus (Bull.: Fr.) P. Karst using column chromatography purification from water extraction. Its backbone was predominantly composed of →6)-α-D-Galp-(1→, →2,6)-α-D-Galp-(1→,→6)-α-D-O-Me-Galp-(1→, →3)-α-D-Manp-(1→, and →3,4,6) -β-D-Galp-(1→ residues, branched at C2 of partial α-D-Galp, or C3 and C4 of β-D-Galp, and terminated by α-D-Manp, and α-L-Fucp. In high-fat diet (HFD)-fed obese mice, IHP3 effectively suppressed body weight and plasma glucose gain, decreased fat accumulation, ameliorated lipid metabolism, and protected liver function from HFD-induced damage. Combining the analysis of gut microbiota metabolomics, hepatic proteomics and biochemical detection revealed, IHP3 significantly altered cecum fecal metabolite abundances, inhibited the phosphorylation of peroxisome proliferator-activated receptor gamma, and promoted the browning of white adipose tissue and the activation of brown adipose tissue. These changes collectively contributed to alleviating obesity symptoms by suppressing the interleukin (IL)-17-mediated inflammatory response in obese mice. Therefore, these findings suggest that IHP3 could be a potential candidate for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Yongfeng Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Xin Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; Shenyang Agricultural University, Shenyang 110866, China.
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhige Li
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
3
|
Malicka A, Ali A, MacCannell ADV, Roberts LD. Brown and beige adipose tissue-derived metabokine and lipokine inter-organ signalling in health and disease. Exp Physiol 2024. [PMID: 39591977 DOI: 10.1113/ep092008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Adipose tissue has an established endocrine function through the secretion of adipokines. However, a role for bioactive metabolites and lipids, termed metabokines and lipokines, is emerging in adipose tissue-mediated autocrine, paracrine and endocrine signalling and inter-organ communication. Traditionally seen as passive entities, metabolites are now recognized for their active roles in regulating cellular signalling and local and systemic metabolism. Distinct from white adipose tissue, specific endocrine functions have been attributed to thermogenic brown and beige adipose tissues. Brown and beige adipose tissues have been identified as sources of metabokines and lipokines, which influence diverse metabolic pathways, such as fatty acid β-oxidation, mitochondrial function and glucose homeostasis, across a range of tissues, including skeletal muscle, adipose tissue and heart. This review explores the intricate signalling mechanisms of brown and beige adipose tissue-derived metabokines and lipokines, emphasizing their roles in maintaining metabolic homeostasis and their potential dysregulation in metabolic diseases. Furthermore, we discuss the therapeutic potential of targeting these pathways, proposing that precise modulation of metabokine receptors and transporters could offer superior specificity and efficacy in comparison to conventional approaches, such as β-adrenergic signalling-stimulated activation of brown adipose tissue thermogenesis. Understanding the complex interactions between adipokines, metabokines and lipokines is essential for developing a systems-level approach to new interventions for metabolic disorders, underscoring the need for continued research in this rapidly evolving field.
Collapse
Affiliation(s)
- Anna Malicka
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Aysha Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
4
|
Saponaro F, Bertolini A, Baragatti R, Galfo L, Chiellini G, Saba A, D'Urso G. Myokines and Microbiota: New Perspectives in the Endocrine Muscle-Gut Axis. Nutrients 2024; 16:4032. [PMID: 39683426 DOI: 10.3390/nu16234032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
This review explores the dual role of skeletal muscle as both a mechanical and endocrine organ, highlighting its contributions to overall health and its adaptability to various inputs such as nutrition, hormones, exercise, and injuries. In addition to its role in metabolism and energy conversion, skeletal muscle secretes signalling molecules called myokines (at rest) and exerkines (during/after physical exercise), which communicate with other organs like the brain, the cardiovascular system, and the immune system. Key molecules such as interleukins, irisin, and myostatin are discussed for their roles in mediating muscle health and inter-organ communication. This work also focuses on the muscle-gut axis, emphasising the bidirectional interaction between skeletal muscle and the gut microbiota, a complex ecosystem influencing immune defence, digestion, and metabolism. Muscle activity, particularly exercise, alters the gut microbial composition, promoting beneficial species, while gut-derived metabolites like short-chain fatty acids (SCFAs) impact muscle metabolism, mitochondrial function, and insulin sensitivity. Dysbiosis, or an imbalanced microbiota, can lead to muscle atrophy, inflammation, and metabolic dysfunction. This evidence highlights emerging research into myokines and exerkines as potential therapeutic targets for managing conditions like muscle decline, ageing, and metabolic diseases through muscle-gut interactions.
Collapse
Affiliation(s)
- Federica Saponaro
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Andrea Bertolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Riccardo Baragatti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Leonardo Galfo
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Grazia Chiellini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Giuseppina D'Urso
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| |
Collapse
|
5
|
Ağagündüz D, Yeşildemir Ö, Koçyiğit E, Koçak T, Özen Ünaldı B, Ayakdaş G, Budán F. Oxylipins Derived from PUFAs in Cardiometabolic Diseases: Mechanism of Actions and Possible Nutritional Interactions. Nutrients 2024; 16:3812. [PMID: 39599599 PMCID: PMC11597274 DOI: 10.3390/nu16223812] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Oxylipins are oxidized fatty acids, both saturated and unsaturated, formed through pathways that involve singlet oxygen or dioxygen-mediated oxygenation reactions and are primarily produced by enzyme families such as cyclooxygenases, lipoxygenases, and cytochrome P450. These lipid-based complex bioactive molecules are pivotal signal mediators, acting in a hormone-like manner in the pathophysiology of numerous diseases, especially cardiometabolic diseases via modulating plenty of mechanisms. It has been reported that omega-6 and omega-3 oxylipins are important novel biomarkers of cardiometabolic diseases. Moreover, collected literature has noted that diet and dietary components, especially fatty acids, can modulate these oxygenated lipid products since they are mainly derived from dietary omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) or linoleic acid and α-linolenic by elongation and desaturation pathways. This comprehensive review aims to examine their correlations to cardiometabolic diseases and how diets modulate oxylipins. Also, some aspects of developing new biomarkers and therapeutical utilization are detailed in this review.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Türkiye
| | - Özge Yeşildemir
- Department of Nutrition and Dietetics, Bursa Uludag University, Görükle Campus, 16059 Bursa, Türkiye;
| | - Emine Koçyiğit
- Department of Nutrition and Dietetics, Ordu University, Cumhuriyet Yerleşkesi, 52200 Ordu, Türkiye;
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Gümüşhane University, Gümüşhanevî Kampüsü, 29100 Gümüşhane, Türkiye;
| | - Buket Özen Ünaldı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye;
| | - Gamze Ayakdaş
- Department of Nutrition and Dietetics, Acıbadem University, Kerem Aydınlar Campus, 34752 İstanbul, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
6
|
Huang Y, Liang M, Liao Y, Ji Z, Lin W, Pu X, Wang L, Wang W. Investigating the Mechanisms of 15-PGDH Inhibitor SW033291 in Improving Type 2 Diabetes Mellitus: Insights from Metabolomics and Transcriptomics. Metabolites 2024; 14:509. [PMID: 39330516 PMCID: PMC11434390 DOI: 10.3390/metabo14090509] [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: 08/02/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
This study focused on exploring the effects of SW033291, an inhibitor of 15-hydroxyprostaglandin dehydrogenase, on type 2 diabetes mellitus (T2DM) mice from a comprehensive perspective. Studies have demonstrated that SW033291 benefits tissue repair, organ function, and muscle mass in elderly mice. Our recent investigation initially reported the beneficial effect of SW033291 on T2DM progression. Herein, we used a T2DM mouse model induced by a high-fat diet and streptozotocin injection. Then, serum and liver metabolomics, as well as liver transcriptomic analyses, were performed to provide a systematic perspective of the SW033291-ameliorated T2DM. The results indicate SW033291 improved T2DM by regulating steroid hormone biosynthesis and linoleic/arachidonic acid metabolism. Furthermore, integrated transcriptomic and metabolomic analyses suggested that key genes and metabolites such as Cyp2c55, Cyp3a11, Cyp21a1, Myc, Gstm1, Gstm3, 9,10-dihydroxyoctadecenoic acid, 11-dehydrocorticosterone, and 12,13-dihydroxy-9Z-octadecenoic acid played crucial roles in these pathways. qPCR analysis validated the significant decreases in the hepatic gene expressions of Cyp2c55, Cyp3a11, Myc, Gstm1, and Gstm3 in the T2DM mice, which were reversed following SW033291 treatment. Meanwhile, the elevated mRNA level of Cyp21a1 in T2DM mice was decreased after SW033291 administration. Taken together, our findings suggest that SW033291 has promising potential in alleviating T2DM and could be a novel therapeutic candidate.
Collapse
Affiliation(s)
- Yuanfeng Huang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Mingjie Liang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
- Guangdong Nephrotic Drug Engineering Technology Research Center, Guangdong Consun Pharmaceutical Group, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangzhou 510700, China
| | - Yiwen Liao
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Zirui Ji
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Wanfen Lin
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Xiangjin Pu
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Lexun Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| | - Weixuan Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, No. 280, Waihuan East Road, University Town, Guangzhou 510006, China; (Y.H.); (M.L.); (Y.L.); (Z.J.); (W.L.); (X.P.); (L.W.)
- Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou 510006, China
| |
Collapse
|
7
|
Chen L, Liu L. Adipose thermogenic mechanisms by cold, exercise and intermittent fasting: Similarities, disparities and the application in treatment. Clin Nutr 2024; 43:2043-2056. [PMID: 39088961 DOI: 10.1016/j.clnu.2024.07.024] [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: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Given its nonnegligible role in metabolic homeostasis, adipose tissue has been the target for treating metabolic disorders such as obesity, diabetes and cardiovascular diseases. Besides its lipolytic function, adipose thermogenesis has gained increased interest due to the irreplaceable contribution to dissipating energy to restore equilibrium, and its therapeutic effects have been testified in various animal models. In this review, we will brief about the canonical cold-stimulated adipose thermogenic mechanisms, elucidate on the exercise- and intermittent fasting-induced adipose thermogenic mechanisms, with a focus on the similarities and disparities among these signaling pathways, in an effort to uncover the overlapped and specific targets that may yield potent therapeutic efficacy synergistically in improving metabolic health.
Collapse
Affiliation(s)
- Linshan Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China.
| |
Collapse
|
8
|
Tang X, Wang J, Ouyang X, Chen Q, Dong R, Luo Y, Zhong J, Huang Z, Peng L, Xie X, Zhu J, Zheng Z, Li S. Coronary Sinus Metabolite 12,13-diHOME Is a Novel Biomarker for Left Atrial Remodeling in Patients With Atrial Fibrillation. Circ Arrhythm Electrophysiol 2024; 17:e012486. [PMID: 38690652 DOI: 10.1161/circep.123.012486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/22/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) has shown potential in protecting against heart disease, but its relationship with atrial fibrillation (AF) remains unknown. METHODS Coronary sinus (CS) and femoral vein blood samplings were synchronously collected from AF and non-AF subjects (paroxysmal supraventricular tachycardia or idiopathic premature ventricular complexes) who underwent catheter ablation. First, untargeted metabolomic profiling was performed in a discovery cohort (including 12 AF and 12 non-AF subjects) to identify the most promising CS or femoral vein metabolite. Then, the selected metabolite was further measured in a validation cohort (including 119 AF and 103 non-AF subjects) to confirm its relationship with left atrium remodeling and 1-year postablation recurrence of AF. Finally, the biological function of the selected metabolite was validated in a rapid-paced cultured HL-1 atrial cardiomyocytes model. RESULTS Metabolomic analysis identified CS 12,13-diHOME as the most pronounced change metabolite correlated with left atrium remodeling in the discovery cohort. In the validation cohort, CS 12,13-diHOME was significantly lower in patients with AF than non-AF controls (84.32±20.13 versus 96.24±23.56 pg/mL; P<0.01), and associated with worse structural, functional, and electrical remodeling of left atrium. Multivariable regression analyses further demonstrated that decreased CS 12,13-diHOME was an independent predictor of 1-year postablation recurrence of AF (odds ratio, 0.754 [95% CI, 0.648-0.920]; P=0.005). Biological function validations showed that 12,13-diHOME treatment significantly protect the cell viability, improved the expression of MHC (myosin heavy chain) and Cav1.2 (L-type calcium channel α1c), and attenuated mitochondrial damage in the rapid-paced cultured HL-1 cardiomyocytes model. CONCLUSIONS CS metabolite 12,13-diHOME is decreased in patients with AF and can serve as a novel biomarker for left atrium remodeling.
Collapse
Affiliation(s)
- Xixiang Tang
- VIP Medical Service Center (X.T.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiafu Wang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaolan Ouyang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Chen
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruimin Dong
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanting Luo
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junlin Zhong
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuoshan Huang
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long Peng
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xujing Xie
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieming Zhu
- Department of Ultrasonography (J.Z.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhenda Zheng
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suhua Li
- Department of Cardiovascular Medicine (J.W., X.O., Q.C., R.D., Y.L., Z.H., L.P., X.X., J.Z., Z.Z., S.L.), the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Zhang J, Chen F. Integrated transcriptome and metabolome study reveal the therapeutic effects of nicotinamide riboside and nicotinamide mononucleotide on nonalcoholic fatty liver disease. Biomed Pharmacother 2024; 175:116701. [PMID: 38729053 DOI: 10.1016/j.biopha.2024.116701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR) have received considerable attention as anti-aging and anti-metabolic disease nutraceuticals. However, few studies have focused on their role in ameliorating hepatic metabolic disturbances. In the present study, the effects of NMN and NR on the liver of mice with nonalcoholic fatty liver disease (NAFLD) were investigated via transcriptome and metabolome analyses. NMN and NR reduced body weight gain, improved glucose homeostasis, regulated plasma lipid levels, and ameliorated liver injury, oxidative stress, and lipid accumulation in mice with HFD-induced NAFLD. Integrated transcriptome and metabolome analyses indicated that NMN and NR altered the biosynthesis of unsaturated fatty acids, arachidonic acid metabolism, and linoleic acid metabolism pathways, increased saturated fatty acid (palmitic acid, stearate, and arachidic acid) content, and increased polyunsaturated fatty acid (linoleic acid and eicosapentaenoic acid) content. Quantitative reverse transcription PCR (qRT-PCR) showed that NMN and NR primarily promoted arachidonic acid and linoleic acid catabolism via cytochrome P450 (CYP450) enzymes. This study established a theoretical foundation for the potential use of NMN and NR in future clinical settings.
Collapse
Affiliation(s)
- Jingting Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China; College of Management, Liaoning Economy Vocational and Technical College, Shenyang, Liaoning 110122, China.
| | - Fu Chen
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China.
| |
Collapse
|
10
|
Dhillon J, Pandey S, Newman JW, Fiehn O, Ortiz RM. Metabolic Responses to an Acute Glucose Challenge: The Differential Effects of Eight Weeks of Almond vs. Cracker Consumption in Young Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.19.24307571. [PMID: 38826341 PMCID: PMC11142291 DOI: 10.1101/2024.05.19.24307571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study investigated the dynamic responses to an acute glucose challenge following chronic almond versus cracker consumption for 8 weeks (clinicaltrials.gov ID: NCT03084003). Seventy-three young adults (age: 18-19 years, BMI: 18-41 kg/m2) participated in an 8-week randomized, controlled, parallel-arm intervention and were randomly assigned to consume either almonds (2 oz/d, n=38) or an isocaloric control snack of graham crackers (325 kcal/d, n=35) daily for 8 weeks. Twenty participants from each group underwent a 2-hour oral glucose tolerance test (oGTT) at the end of the 8-week intervention. Metabolite abundances in the oGTT serum samples were quantified using untargeted metabolomics, and targeted analyses for free PUFAs, total fatty acids, oxylipins, and endocannabinoids. Multivariate, univariate, and chemical enrichment analyses were conducted to identify significant metabolic shifts. Findings exhibit a biphasic lipid response distinguished by higher levels of unsaturated triglycerides in the earlier periods of the oGTT followed by lower levels in the latter period in the almond versus cracker group (p-value<0.05, chemical enrichment analyses). Almond (vs. cracker) consumption was also associated with higher AUC120 min of aminomalonate, and oxylipins (p-value<0.05), but lower AUC120 min of L-cystine, N-acetylmannosamine, and isoheptadecanoic acid (p-value<0.05). Additionally, the Matsuda Index in the almond group correlated with AUC120 min of CE 22:6 (r=-0.46; p-value<0.05) and 12,13 DiHOME (r=0.45; p-value<0.05). Almond consumption for 8 weeks leads to dynamic, differential shifts in response to an acute glucose challenge, marked by alterations in lipid and amino acid mediators involved in metabolic and physiological pathways.
Collapse
Affiliation(s)
- Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Department of Molecular and Cell Biology, University of California, Merced
| | - Saurabh Pandey
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Jaypee University of Information Technology, Waknaghat, India
| | - John W. Newman
- West Coast Metabolomics Center, University of California, Davis
- Department of Nutrition, University of California, Davis
- Obesity and Metabolism Research Unit, USDA Agricultural Research Service Western Human Nutrition Research Center, University of California, Davis
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis
| | - Rudy M. Ortiz
- Department of Molecular and Cell Biology, University of California, Merced
| |
Collapse
|
11
|
Suthar H, Manea T, Pak D, Woodbury M, Eick SM, Cathey A, Watkins DJ, Strakovsky RS, Ryva BA, Pennathur S, Zeng L, Weller D, Park JS, Smith S, DeMicco E, Padula A, Fry RC, Mukherjee B, Aguiar A, Geiger SD, Ng S, Huerta-Montanez G, Vélez-Vega C, Rosario Z, Cordero JF, Zimmerman E, Woodruff TJ, Morello-Frosch R, Schantz SL, Meeker JD, Alshawabkeh AN, Aung MT. Cross-Sectional Associations between Prenatal Per- and Poly-Fluoroalkyl Substances and Bioactive Lipids in Three Environmental Influences on Child Health Outcomes (ECHO) Cohorts. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:8264-8277. [PMID: 38691655 PMCID: PMC11097396 DOI: 10.1021/acs.est.4c00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
Prenatal per- and poly-fluoroalkyl substances (PFAS) exposure may influence gestational outcomes through bioactive lipids─metabolic and inflammation pathway indicators. We estimated associations between prenatal PFAS exposure and bioactive lipids, measuring 12 serum PFAS and 50 plasma bioactive lipids in 414 pregnant women (median 17.4 weeks' gestation) from three Environmental influences on Child Health Outcomes Program cohorts. Pairwise association estimates across cohorts were obtained through linear mixed models and meta-analysis, adjusting the former for false discovery rates. Associations between the PFAS mixture and bioactive lipids were estimated using quantile g-computation. Pairwise analyses revealed bioactive lipid levels associated with PFDeA, PFNA, PFOA, and PFUdA (p < 0.05) across three enzymatic pathways (cyclooxygenase, cytochrome p450, lipoxygenase) in at least one combined cohort analysis, and PFOA and PFUdA (q < 0.2) in one linear mixed model. The strongest signature revealed doubling in PFOA corresponding with PGD2 (cyclooxygenase pathway; +24.3%, 95% CI: 7.3-43.9%) in the combined cohort. Mixture analysis revealed nine positive associations across all pathways with the PFAS mixture, the strongest signature indicating a quartile increase in the PFAS mixture associated with PGD2 (+34%, 95% CI: 8-66%), primarily driven by PFOS. Bioactive lipids emerged as prenatal PFAS exposure biomarkers, deepening insights into PFAS' influence on pregnancy outcomes.
Collapse
Affiliation(s)
- Himal Suthar
- Department
of Population and Public Health Sciences, University of Southern California, Los Angeles, California 90032, United States
| | - Tomás Manea
- Department
of Population and Public Health Sciences, University of Southern California, Los Angeles, California 90032, United States
| | - Dominic Pak
- Department
of Population and Public Health Sciences, University of Southern California, Los Angeles, California 90032, United States
| | - Megan Woodbury
- Department
of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Stephanie M. Eick
- Gangarosa
Department of Environmental Health, Emory
University Rollins School of Public Health, Atlanta, Georgia 30322, United States
| | - Amber Cathey
- Department
of Environmental Health Sciences, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Deborah J. Watkins
- Department
of Environmental Health Sciences, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Rita S. Strakovsky
- Institute
for Integrative Toxicology, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Food Sciences and Human Nutrition, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Brad A. Ryva
- Institute
for Integrative Toxicology, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
- College
of Osteopathic Medicine, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Subramaniam Pennathur
- Department
of Internal Medicine-Nephrology, University
of Michigan, Ann Arbor, Michigan 48824, United States
- Department
of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lixia Zeng
- Department
of Internal Medicine-Nephrology, University
of Michigan, Ann Arbor, Michigan 48824, United States
| | - David Weller
- NSF International, Ann Arbor, Michigan 48105, United States
| | - June-Soo Park
- Environmental Chemistry Laboratory, Department of Toxic
Substances
Control, California Environmental Protection
Agency, Berkeley, California 94710, United States
| | - Sabrina Smith
- Environmental Chemistry Laboratory, Department of Toxic
Substances
Control, California Environmental Protection
Agency, Berkeley, California 94710, United States
| | - Erin DeMicco
- Program on Reproductive
Health and the Environment, University of
California, San Francisco, San
Francisco, California 94143, United States
| | - Amy Padula
- Program on Reproductive
Health and the Environment, University of
California, San Francisco, San
Francisco, California 94143, United States
| | - Rebecca C. Fry
- Department
of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, Gillings
School of Global Public Health, Chapel Hill, North Carolina 27599, United States
| | - Bhramar Mukherjee
- Department of Biostatistics, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Andrea Aguiar
- Beckman
Institute for Advanced Science and Technology, University of Illinois Urbana−Champaign, Champaign, Illinois 61801, United States
- Department of Comparative Biosciences, University of Illinois Urbana−Champaign, Champaign, Illinois 61802, United States
| | - Sarah Dee Geiger
- Department of Comparative Biosciences, University of Illinois Urbana−Champaign, Champaign, Illinois 61802, United States
- Department of Kinesiology and Community Health, University of Illinois at Urbana−Champaign, Champaign, Illinois 61801, United States
| | - Shukhan Ng
- Department of Comparative Biosciences, University of Illinois Urbana−Champaign, Champaign, Illinois 61802, United States
| | - Gredia Huerta-Montanez
- Department
of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Carmen Vélez-Vega
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia 30606, United States
| | - Zaira Rosario
- University of Puerto Rico Graduate School of Public Health, San Juan, Puerto Rico 00935, United States
| | - Jose F. Cordero
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia 30606, United States
| | - Emily Zimmerman
- Department of Communication Sciences and Disorders, Northeastern University, Boston, Massachusetts 02115, United States
| | - Tracey J. Woodruff
- Program on Reproductive
Health and the Environment, University of
California, San Francisco, San
Francisco, California 94143, United States
| | - Rachel Morello-Frosch
- Program on Reproductive
Health and the Environment, University of
California, San Francisco, San
Francisco, California 94143, United States
- Department of Environmental Science, Policy and Management
and School of Public Health, University
of California, Berkeley, Berkeley, California 94720, United States
| | - Susan L. Schantz
- Beckman
Institute for Advanced Science and Technology, University of Illinois Urbana−Champaign, Champaign, Illinois 61801, United States
- Department of Comparative Biosciences, University of Illinois Urbana−Champaign, Champaign, Illinois 61802, United States
| | - John D. Meeker
- Department
of Environmental Health Sciences, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Akram N. Alshawabkeh
- Department
of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Max T. Aung
- Department
of Population and Public Health Sciences, University of Southern California, Los Angeles, California 90032, United States
| | - on behalf of Program Collaborators
for Environmental Influences on Child Health Outcomes
- Department
of Population and Public Health Sciences, University of Southern California, Los Angeles, California 90032, United States
- Department
of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts 02115, United States
- Gangarosa
Department of Environmental Health, Emory
University Rollins School of Public Health, Atlanta, Georgia 30322, United States
- Department
of Environmental Health Sciences, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
- Institute
for Integrative Toxicology, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Food Sciences and Human Nutrition, Michigan
State University, East Lansing, Michigan 48824, United States
- Department
of Pharmacology and Toxicology, Michigan
State University, East Lansing, Michigan 48824, United States
- College
of Osteopathic Medicine, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Internal Medicine-Nephrology, University
of Michigan, Ann Arbor, Michigan 48824, United States
- Department
of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, United States
- NSF International, Ann Arbor, Michigan 48105, United States
- Environmental Chemistry Laboratory, Department of Toxic
Substances
Control, California Environmental Protection
Agency, Berkeley, California 94710, United States
- Program on Reproductive
Health and the Environment, University of
California, San Francisco, San
Francisco, California 94143, United States
- Department
of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, Gillings
School of Global Public Health, Chapel Hill, North Carolina 27599, United States
- Department of Biostatistics, University
of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
- Beckman
Institute for Advanced Science and Technology, University of Illinois Urbana−Champaign, Champaign, Illinois 61801, United States
- Department of Comparative Biosciences, University of Illinois Urbana−Champaign, Champaign, Illinois 61802, United States
- Department of Kinesiology and Community Health, University of Illinois at Urbana−Champaign, Champaign, Illinois 61801, United States
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia 30606, United States
- University of Puerto Rico Graduate School of Public Health, San Juan, Puerto Rico 00935, United States
- Department of Communication Sciences and Disorders, Northeastern University, Boston, Massachusetts 02115, United States
- Department of Environmental Science, Policy and Management
and School of Public Health, University
of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
12
|
Li H, Zou L, Zheng J, Yang T. 12,13-diHOME attenuates high glucose-induced calcification of vascular smooth muscle cells through repressing CPT1A-mediated HMGB1 succinylation. Exp Cell Res 2024; 438:114031. [PMID: 38616032 DOI: 10.1016/j.yexcr.2024.114031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024]
Abstract
Diabetes is closely associated with vascular calcification (VC). Exorbitant glucose concentration activates pro-calcific effects in vascular smooth muscle cells (VSMCs). This study enrolled 159 elderly patients with type 2 diabetes and divided them into three groups, T1, T2 and T3, according to brachial-ankle pulse wave velocity(BaPWV). There were statistically significant differences in the waist circumference, waist hip ratio, systolic blood pressure, 12,13-diHOME (a lipokin) concentration among T1, T2 and T3. 12,13-diHOME levels were positively correlated to high density lipoprotein cholesterol and total cholesterol, but negatively correlated to with waist circumference, waist hip ratio, systolic blood pressure and baPWV. Studies in vitro showed that 12,13-diHOME effectively inhibits calcification in VSMCs under high glucose conditions. Notably, 12,13-diHOME suppressed the up-regulation of carnitine O-palmitoyltransferase 1 (CPT1A) and CPT1A-induced succinylation of HMGB1. The succinylation of HMGB1 at the K90 promoted the protein stability and induced the enrichment of HMGB1 in cytoplasm, which induced the calcification in VSMCs. Together, 12,13-diHOME attenuates high glucose-induced calcification in VSMCs through repressing CPT1A-mediated HMGB1 succinylation.
Collapse
MESH Headings
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Carnitine O-Palmitoyltransferase/metabolism
- Carnitine O-Palmitoyltransferase/genetics
- HMGB1 Protein/metabolism
- Glucose/metabolism
- Glucose/pharmacology
- Male
- Aged
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Female
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Cells, Cultured
Collapse
Affiliation(s)
- Huahua Li
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Geriatric, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Lingling Zou
- Department of Geriatric, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jin Zheng
- Department of Geriatric, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Gómez-Hernández A, de las Heras N, Gálvez BG, Fernández-Marcelo T, Fernández-Millán E, Escribano Ó. New Mediators in the Crosstalk between Different Adipose Tissues. Int J Mol Sci 2024; 25:4659. [PMID: 38731880 PMCID: PMC11083914 DOI: 10.3390/ijms25094659] [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: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Adipose tissue is a multifunctional organ that regulates many physiological processes such as energy homeostasis, nutrition, the regulation of insulin sensitivity, body temperature, and immune response. In this review, we highlight the relevance of the different mediators that control adipose tissue activity through a systematic review of the main players present in white and brown adipose tissues. Among them, inflammatory mediators secreted by the adipose tissue, such as classical adipokines and more recent ones, elements of the immune system infiltrated into the adipose tissue (certain cell types and interleukins), as well as the role of intestinal microbiota and derived metabolites, have been reviewed. Furthermore, anti-obesity mediators that promote the activation of beige adipose tissue, e.g., myokines, thyroid hormones, amino acids, and both long and micro RNAs, are exhaustively examined. Finally, we also analyze therapeutic strategies based on those mediators that have been described to date. In conclusion, novel regulators of obesity, such as microRNAs or microbiota, are being characterized and are promising tools to treat obesity in the future.
Collapse
Affiliation(s)
- Almudena Gómez-Hernández
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Natalia de las Heras
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain;
| | - Beatriz G. Gálvez
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Tamara Fernández-Marcelo
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
| | - Elisa Fernández-Millán
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Óscar Escribano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, s/n, 28040 Madrid, Spain; (A.G.-H.); (B.G.G.); (T.F.-M.); (E.F.-M.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
14
|
Liu Y, Qian SW, Tang Y, Tang QQ. The secretory function of adipose tissues in metabolic regulation. LIFE METABOLISM 2024; 3:loae003. [PMID: 39872218 PMCID: PMC11748999 DOI: 10.1093/lifemeta/loae003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2025]
Abstract
In addition to their pivotal roles in energy storage and expenditure, adipose tissues play a crucial part in the secretion of bioactive molecules, including peptides, lipids, metabolites, and extracellular vesicles, in response to physiological stimulation and metabolic stress. These secretory factors, through autocrine and paracrine mechanisms, regulate various processes within adipose tissues. These processes include adipogenesis, glucose and lipid metabolism, inflammation, and adaptive thermogenesis, all of which are essential for the maintenance of the balance and functionality of the adipose tissue micro-environment. A subset of these adipose-derived secretory factors can enter the circulation and target the distant tissues to regulate appetite, cognitive function, energy expenditure, insulin secretion and sensitivity, gluconeogenesis, cardiovascular remodeling, and exercise capacity. In this review, we highlight the role of adipose-derived secretory factors and their signaling pathways in modulating metabolic homeostasis. Furthermore, we delve into the alterations in both the content and secretion processes of these factors under various physiological and pathological conditions, shedding light on potential pharmacological treatment strategies for related diseases.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Martins FF, Martins BC, Teixeira AVS, Ajackson M, Souza-Mello V, Daleprane JB. Brown Adipose Tissue, Batokines, and Bioactive Compounds in Foods: An Update. Mol Nutr Food Res 2024; 68:e2300634. [PMID: 38402434 DOI: 10.1002/mnfr.202300634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/20/2023] [Indexed: 02/26/2024]
Abstract
The discovery of metabolically active brown adipose tissue (BAT) in human adults and the worldwide increase in obesity and obesity-related chronic noncommunicable diseases (NCDs) has made BAT a therapeutic target in the last two decades. The potential of BAT to oxidize fatty acids rapidly and increase energy expenditure inversely correlates with adiposity, insulin and glucose resistance, and cardiovascular and metabolic diseases. Currently, BAT is recognized by a new molecular signature; several BAT-derived molecules that act positively on target tissues have been identified and collectively called batokines. Bioactive compounds present in foods are endowed with thermogenic properties that increase BAT activation signaling. Understanding the mechanisms that lead to BAT activation and the batokines secreted by it within the thermogenic state is fundamental for its recruitment and management of obesity and NCDs. This review contributes to recent updates on the morphophysiology of BAT, its endocrine role in obesity, and the main bioactive compounds present in foods involved in classical and nonclassical thermogenic pathways activation.
Collapse
Affiliation(s)
- Fabiane Ferreira Martins
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, São Francisco Xavier 524, Rio de Janeiro, 20550900, Brazil
- Department of Morphology, Federal University of Rio Grande do Norte, Rio Grande do Norte, 59078-970, Brazil
| | - Bruna Cadete Martins
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, São Francisco Xavier 524, Rio de Janeiro, 20550900, Brazil
| | - Ananda Vitoria Silva Teixeira
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, São Francisco Xavier 524, Rio de Janeiro, 20550900, Brazil
| | - Matheus Ajackson
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, São Francisco Xavier 524, Rio de Janeiro, 20550900, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, 205521031, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, São Francisco Xavier 524, Rio de Janeiro, 20550900, Brazil
| |
Collapse
|
16
|
Lobbes H, Dalle C, Pereira B, Ruivard M, Mazur A, Gladine C. Eicosanoids and Oxylipin Signature in Hereditary Hemochromatosis Patients Are Similar to Dysmetabolic Iron Overload Syndrome Patients but Are Impacted by Dietary Iron Absorption. ANNALS OF NUTRITION & METABOLISM 2024; 80:117-127. [PMID: 38354712 DOI: 10.1159/000536657] [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: 06/20/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Oxylipins are mediators of oxidative stress. To characterize the underlying inflammatory processes and phenotype effect of iron metabolism disorders, we investigated the oxylipin profile in hereditary hemochromatosis (HH) and dysmetabolic iron overload syndrome (DIOS) patients. METHODS An LC-MS/MS-based method was performed to quantify plasma oxylipins in 20 HH and 20 DIOS patients in fasting conditions and 3 h after an iron-rich meal in HH patients. RESULTS Principal component analysis showed no separation between HH and DIOS, suggesting that the clinical phenotype has no direct impact on oxylipin metabolism. 20-HETE was higher in DIOS and correlated with hypertension (p = 0.03). Different oxylipin signatures were observed in HH before and after the iron-rich meal. Discriminant oxylipins include epoxy fatty acids derived from docosahexaenoic acid and arachidonic acid as well as 13-HODE and 9-HODE. Mediation analysis found no major contribution of dietary iron absorption for 16/22 oxylipins significantly affected by the meal. DISCUSSION The oxylipin profiles of HH and DIOS seemed similar except for 20-HETE, possibly reflecting different hypertension prevalence between the two groups. Oxylipins were significantly affected by the iron-rich meal, but the specific contribution of iron was not clear. Although iron may contribute to oxidative stress and inflammation in HH and DIOS, this does not seem to directly affect oxylipin metabolism.
Collapse
Affiliation(s)
- Hervé Lobbes
- Médecine Interne, Hôpital Estaing, 1 Place Lucie et Raymond Aubrac, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France,
- Institut Pascal, UMR 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France,
| | - Céline Dalle
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| | - Bruno Pereira
- Unité de biostatistiques, Direction de la Recherche Clinique et de l'Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Marc Ruivard
- Médecine Interne, Hôpital Estaing, 1 Place Lucie et Raymond Aubrac, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Institut Pascal, UMR 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Andrzej Mazur
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| | - Cécile Gladine
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| |
Collapse
|
17
|
Siegel L, Rooney J, Marjoram L, Mason L, Bowles E, van Keulen TV, Helander C, Rayo V, Hong MY, Liu C, Hooshmand S, Kern M, Witard OC. Chronic almond nut snacking alleviates perceived muscle soreness following downhill running but does not improve indices of cardiometabolic health in mildly overweight, middle-aged, adults. Front Nutr 2024; 10:1298868. [PMID: 38260074 PMCID: PMC10800814 DOI: 10.3389/fnut.2023.1298868] [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: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction As a popular food snack rich in protein, fiber, unsaturated fatty acids, antioxidants and phytonutrients, almond nut consumption is widely associated with improvements in cardiometabolic health. However, limited data exists regarding the role of almond consumption in improving exercise recovery. Accordingly, we aimed to investigate the impact of chronic almond snacking on muscle damage and cardiometabolic health outcomes during acute eccentric exercise recovery in mildly overweight, middle-aged, adults. Methods Using a randomized cross-over design, 25 mildly overweight (BMI: 25.8 ± 3.6 kg/m2), middle-aged (35.1 ± 4.7 y) males (n = 11) and females (n = 14) performed a 30-min downhill treadmill run after 8-weeks of consuming either 57 g/day of whole almonds (ALMOND) or an isocaloric amount (86 g/day) of unsalted pretzels (CONTROL). Muscle soreness (visual analogue scale), muscle function (vertical jump and maximal isokinetic torque) and blood markers of muscle damage (creatine kinase (CK) concentration) and inflammation (c-reactive protein concentration) were measured pre and post (24, 48, and 72 h) exercise. Blood biomarkers of cardiometabolic health (total cholesterol, triglycerides, HDL cholesterol, and LDL cholesterol), body composition and psycho-social assessments of mood (POMS-2 inventory), appetite and well-being were measured pre and post intervention. Results Downhill running successfully elicited muscle damage, as evidenced by a significant increase in plasma CK concentration, increased perception of muscle soreness, and impaired vertical jump performance (all p < 0.05) during acute recovery. No effect of trial order was observed for any outcome measurement. However, expressed as AUC over the cumulative 72 h recovery period, muscle soreness measured during a physical task (vertical jump) was reduced by ~24% in ALMOND vs. CONTROL (p < 0.05) and translated to an improved maintenance of vertical jump performance (p < 0.05). However, ALMOND did not ameliorate the CK response to exercise or isokinetic torque during leg extension and leg flexion (p > 0.05). No pre-post intervention changes in assessments of cardiometabolic health, body composition, mood state or appetite were observed in ALMOND or CONTROL (all p > 0.05). Conclusion Chronic almond supplementation alleviates task-specific perceived feelings of muscle soreness during acute recovery from muscle damaging exercise, resulting in the better maintenance of muscle functional capacity. These data suggest that almonds represent a functional food snack to improve exercise tolerance in mildly overweight, middle-aged adults.
Collapse
Affiliation(s)
- Leah Siegel
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Jessica Rooney
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Lindsey Marjoram
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Lauren Mason
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Elena Bowles
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Thomas Valente van Keulen
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Carina Helander
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Vernon Rayo
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Mee Young Hong
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Shirin Hooshmand
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Mark Kern
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States
| | - Oliver C. Witard
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
18
|
Zhu S, Zhao Y, Liu L, Xu Y, Zhu J, Li W, Liu Y, Xia M. High Plant Protein Diet Ameliorated Hepatic Lipid Accumulation Through the Modulation of Gut Microbiota. Mol Nutr Food Res 2023; 67:e2300515. [PMID: 37876152 DOI: 10.1002/mnfr.202300515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Indexed: 10/26/2023]
Abstract
SCOPE Substituting plant protein for animal protein has emerged as a promising strategy for managing atherogenic lipids. However, the impact of long-term intake of a high plant protein diet (HPD) on hepatic lipid disorder remains unclear. METHODS AND RESULTS Eight-week-old apolipoprotein E deficient (apoE-/- ) mice are fed with either a normal protein diet (NCD) or HPD for 12 weeks. HPD intervention results in decreased body weight accompanied by increased energy expenditure, with no significant effect on glycemic control. Long-term intake of HPD improves the serum and hepatic lipid and cholesterol accumulation by suppressing hepatic squalene epoxidase (SQLE) expression, a key enzyme in cholesterol biosynthesis. Integrated analysis of 16S rDNA sequencing and metabolomics profiling reveals that HPD intervention increases the abundance of the Lachnospiraece family and serum levels of 12,13-DiHOME. Furthermore, in vivo studies demonstrate that 12,13-DiHOME significantly inhibits lipid accumulation, as well as SQLE expression induced by oleic acid in HepG2 cells. CONCLUSION Diet rich in plant protein diet alleviates hyperlipidemia via increased microbial production of 12,13-DiHOME.
Collapse
Affiliation(s)
- Shanshan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Yawen Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Ludi Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
- Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Yingxi Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
- Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Jiangyuan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Wenkang Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, P.R. China
| |
Collapse
|
19
|
Ling Q, Han Y, Ma Y, Wang X, Zhu Z, Wang J, Cao J, Lin X, Wang J, Wang B. Alterations in the Gut Microbiome in Liver Recipients with Post-Transplant Diabetes Mellitus. ENGINEERING 2023; 31:98-111. [DOI: 10.1016/j.eng.2023.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
|
20
|
Lau ES, Roshandelpoor A, Zarbafian S, Wang D, Guseh JS, Allen N, Varadarajan V, Nayor M, Shah RV, Lima JAC, Shah SJ, Yu B, Alotaibi M, Cheng S, Jain M, Lewis GD, Ho JE. Eicosanoid and eicosanoid-related inflammatory mediators and exercise intolerance in heart failure with preserved ejection fraction. Nat Commun 2023; 14:7557. [PMID: 37985769 PMCID: PMC10662264 DOI: 10.1038/s41467-023-43363-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Systemic inflammation has been implicated in the pathobiology of heart failure with preserved ejection fraction (HFpEF). Here, we examine the association of upstream mediators of inflammation as ascertained by fatty-acid derived eicosanoid and eicosanoid-related metabolites with HFpEF status and exercise manifestations of HFpEF. Among 510 participants with chronic dyspnea and preserved LVEF who underwent invasive cardiopulmonary exercise testing, we find that 70 of 890 eicosanoid and related metabolites are associated with HFpEF status, including 17 named and 53 putative eicosanoids (FDR q-value < 0.1). Prostaglandin (15R-PGF2α, 11ß-dhk-PGF2α) and linoleic acid derivatives (12,13 EpOME) are associated with greater odds of HFpEF, while epoxides (8(9)-EpETE), docosanoids (13,14-DiHDPA), and oxylipins (12-OPDA) are associated with lower odds of HFpEF. Among 70 metabolites, 18 are associated with future development of heart failure in the community. Pro- and anti-inflammatory eicosanoid and related metabolites may contribute to the pathogenesis of HFpEF and serve as potential targets for intervention.
Collapse
Affiliation(s)
- Emily S Lau
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts, 02114, USA
- Cardiovascular Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Athar Roshandelpoor
- CardioVascular Institute, Division of Cardiology, Department of Medicine, 330 Brookline Avenue, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Shahrooz Zarbafian
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts, 02114, USA
- Google LLC, 1600 Amphitheatre Parkway, Mountain View, CA, 94043, USA
| | - Dongyu Wang
- CardioVascular Institute, Division of Cardiology, Department of Medicine, 330 Brookline Avenue, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Department of Biostatistics, Boston University School of Public Health, 715 Albany Street, Boston, MA, 02118, USA
| | - James S Guseh
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts, 02114, USA
- Cardiovascular Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Norrina Allen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 420 East Superior Street, Chicago, IL, 60611, USA
| | - Vinithra Varadarajan
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Matthew Nayor
- Cardiology Division, Boston University School of Medicine, 715 Albany Street, Boston, MA, 02118, USA
| | - Ravi V Shah
- Vanderbilt Clinical and Translational Research Center (VTRACC), Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN, 37232, USA
| | - Joao A C Lima
- Division of Cardiology, Department of Medicine Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD, 21205, USA
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, 420 East Superior Street, Chicago, IL, 60611, USA
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 420 East Superior Street, Chicago, IL, 60611, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health School of Public Health, 1200 Pressler Street, Houston, TX, 77030, USA
| | - Mona Alotaibi
- Division of Pulmonary and Critical Care and Sleep Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South San Vincente Pavilion, Los Angeles, CA, 90048, USA
| | - Mohit Jain
- Department of Medicine and Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Gregory D Lewis
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts, 02114, USA
- Cardiovascular Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Jennifer E Ho
- Cardiovascular Research Center, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
21
|
Suthar H, Manea T, Pak D, Woodbury M, Eick SM, Cathey A, Watkins DJ, Strakovsky RS, Ryva BA, Pennathur S, Zeng L, Weller D, Park JS, Smith S, DeMicco E, Padula A, Fry RC, Mukherjee B, Aguiar A, Dee Geiger S, Ng S, Huerta-Montanez G, Vélez-Vega C, Rosario Z, Cordero JF, Zimmerman E, Woodruff TJ, Morello-Frosch R, Schantz SL, Meeker JD, Alshawabkeh A, Aung MT. Cross-sectional associations between prenatal maternal per- and poly-fluoroalkyl substances and bioactive lipids in three Environmental influences on Child Health Outcomes (ECHO) cohorts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297930. [PMID: 37961525 PMCID: PMC10635258 DOI: 10.1101/2023.11.03.23297930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Per- and poly-fluoroalkyl substances (PFAS) exposure can occur through ingestion of contaminated food and water, and inhalation of indoor air contaminated with these chemicals from consumer and industrial products. Prenatal PFAS exposures may confer risk for pregnancy-related outcomes such as hypertensive and metabolic disorders, preterm birth, and impaired fetal development through intermediate metabolic and inflammation pathways. Objective Estimate associations between maternal pregnancy PFAS exposure (individually and as a mixture) and bioactive lipids. Methods Our study included pregnant women in the Environmental influences on Child Health Outcomes Program: Chemicals in our Bodies cohort (CiOB, n=73), Illinois Kids Developmental Study (IKIDS, n=287), and the ECHO-PROTECT cohort (n=54). We measured twelve PFAS in serum and 50 plasma bioactive lipids (parent fatty acids and eicosanoids derived from cytochrome p450, lipoxygenase, and cyclooxygenase) during pregnancy (median 17 gestational weeks). Pairwise associations across cohorts were estimated using linear mixed models and meta-analysis. Associations between the PFAS mixture and individual bioactive lipids were estimated using quantile g-computation. Results PFDeA, PFOA, and PFUdA were associated (p<0.05) with changes in bioactive lipid levels in all three enzymatic pathways (cyclooxygenase [n=6 signatures]; cytochrome p450 [n=5 signatures]; lipoxygenase [n=7 signatures]) in at least one combined cohort analysis. The strongest signature indicated that a doubling in PFOA corresponded with a 24.3% increase (95% CI [7.3%, 43.9%]) in PGD2 (cyclooxygenase pathway) in the combined cohort. In the mixtures analysis, we observed nine positive signals across all pathways associated with the PFAS mixture. The strongest signature indicated that a quartile increase in the PFAS mixture was associated with a 34% increase in PGD2 (95% CI [8%, 66%]), with PFOS contributing most to the increase. Conclusions Bioactive lipids were revealed as biomarkers of PFAS exposure and could provide mechanistic insights into PFAS' influence on pregnancy outcomes, informing more precise risk estimation and prevention strategies.
Collapse
Affiliation(s)
- Himal Suthar
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Tomás Manea
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Dominic Pak
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Megan Woodbury
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, USA
| | - Stephanie M. Eick
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Amber Cathey
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Deborah J. Watkins
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Rita S. Strakovsky
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Food Sciences and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Brad A. Ryva
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Subramaniam Pennathur
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lixia Zeng
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, MI, USA
| | | | - June-Soo Park
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, USA
| | - Sabrina Smith
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, USA
| | - Erin DeMicco
- Program on Reproductive Health and the Environment, University of California, San Francisco, San Francisco, CA, USA
| | - Amy Padula
- Program on Reproductive Health and the Environment, University of California, San Francisco, San Francisco, CA, USA
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Andrea Aguiar
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Illinois, USA
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, IL, USA
| | - Sarah Dee Geiger
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, IL, USA
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Shukhan Ng
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, IL, USA
| | - Gredia Huerta-Montanez
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, USA
| | - Carmen Vélez-Vega
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, USA
| | - Zaira Rosario
- University of Puerto Rico Graduate School of Public Health, San Juan, PR, USA
| | - Jose F. Cordero
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, Georgia, USA
| | - Emily Zimmerman
- Department of Communication Sciences and Disorders, Northeastern University, Boston, MA, USA
| | - Tracey J. Woodruff
- Program on Reproductive Health and the Environment, University of California, San Francisco, San Francisco, CA, USA
| | - Rachel Morello-Frosch
- Program on Reproductive Health and the Environment, University of California, San Francisco, San Francisco, CA, USA
- Department of Environmental Science, Policy and Management and School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Susan L. Schantz
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Illinois, USA
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, IL, USA
| | - John D. Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Akram Alshawabkeh
- Department of Civil and Environmental Engineering, Northeastern University, Boston, MA, USA
| | - Max T. Aung
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Moens de Hase E, Petitfils C, Alhouayek M, Depommier C, Le Faouder P, Delzenne NM, Van Hul M, Muccioli GG, Cenac N, Cani PD. Dysosmobacter welbionis effects on glucose, lipid, and energy metabolism are associated with specific bioactive lipids. J Lipid Res 2023; 64:100437. [PMID: 37648213 PMCID: PMC10542644 DOI: 10.1016/j.jlr.2023.100437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
The newly identified bacterium Dysosmobacter welbionis J115T improves host metabolism in high-fat diet (HFD)-fed mice. To investigate mechanisms, we used targeted lipidomics to identify and quantify bioactive lipids produced by the bacterium in the culture medium, the colon, the brown adipose tissue (BAT), and the blood of mice. In vitro, we compared the bioactive lipids produced by D. welbionis J115T versus the probiotic strain Escherichia coli Nissle 1917. D. welbionis J115T administration reduced body weight, fat mass gain, and improved glucose tolerance and insulin resistance in HFD-fed mice. In vitro, 19 bioactive lipids were highly produced by D. welbionis J115T as compared to Escherichia coli Nissle 1917. In the plasma, 13 lipids were significantly changed by the bacteria. C18-3OH was highly present at the level of the bacteria, but decreased by HFD treatment in the plasma and normalized in D. welbionis J115T-treated mice. The metabolic effects were associated with a lower whitening of the BAT. In the BAT, HFD decreased the 15-deoxy-Δ12,14-prostaglandin J2, a peroxisome proliferator-activated receptor (PPAR-γ) agonist increased by 700% in treated mice as compared to HFD-fed mice. Several genes controlled by PPAR-γ were upregulated in the BAT. In the colon, HFD-fed mice had a 60% decrease of resolvin D5, whereas D. welbionis J115T-treated mice exhibited a 660% increase as compared to HFD-fed mice. In a preliminary experiment, we found that D. welbionis J115T improves colitis. In conclusion, D. welbionis J115T influences host metabolism together with several bioactive lipids known as PPAR-γ agonists.
Collapse
Affiliation(s)
- Emilie Moens de Hase
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium; WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Camille Petitfils
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Clara Depommier
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium; WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
| | | | - Nathalie M Delzenne
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium; WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group (BPBL), Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Cenac
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Patrice D Cani
- Metabolism and Nutrition Research group, Louvain Drug Research Institute (LDRI), UCLouvain, Université catholique de Louvain, Brussels, Belgium; WELBIO-Walloon Excellence in Life Sciences and Biotechnology, WELBIO department, WEL Research Institute, Wavre, Belgium; Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
23
|
Edwards-Glenn JM, Fontes MT, Waigi EW, Costa TJ, Maiseyeu A, Webb RC, McCarthy CG, Wenceslau CF. Specialized Pro-resolving Mediator Improves Vascular Relaxation via Formyl Peptide Receptor-2. Am J Hypertens 2023; 36:542-550. [PMID: 37439351 PMCID: PMC10502783 DOI: 10.1093/ajh/hpad062] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/29/2023] [Accepted: 07/11/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The resolution of inflammation is an active phenomenon important for switching off inflammatory processes once the harmful stimuli are removed and facilitate the return to homeostasis. Specialized pro-resolving mediators (SPMs), such as lipoxin A4, resolvin D1, and resolvin E1, derived from ω-3 or ω-6 polyunsaturated fatty acids, are crucial for the resolution of inflammation. We hypothesized that SPMs are decreased in hypertension which contributes to the acetylcholine-induced contraction in resistance arteries, which are well known to be mediated by leukotrienes and prostaglandins. Moreover, treatment with SPMs will decrease this contraction via formyl peptide receptor-2 (FPR-2) in resistance arteries from spontaneously hypertensive rats (SHR). METHODS AND RESULTS We performed a comprehensive eicosanoid lipid panel analysis, and our data showed for the first time that precursors of SPMs are decreased in SHR, limiting the production of SPMs and resolution of inflammation in vivo. This phenomenon was associated with an increase in lipid peroxidation in resistance arteries. Although SPMs did not abolish acetylcholine-induced contraction, these lipid mediators improved endothelial function in arteries from SHR via FPR-2 activation at nanomolar concentrations. SPMs also buffered TNF-α-induced reactive oxygen species generation in endothelial cells from C57Bl/6 mice. CONCLUSIONS We suggest that FPR-2 and SPMs could be revealed as a new target or therapeutic agent to improve vascular function in arteries from hypertensive rats.
Collapse
Affiliation(s)
- Jonnelle M Edwards-Glenn
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Milene T Fontes
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Emily W Waigi
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Tiago J Costa
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Andrei Maiseyeu
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Camilla F Wenceslau
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Biomedical Engineering Program, College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| |
Collapse
|
24
|
Liang S, Jiang Y, Zhang M. Integrative analysis of the effects of organic and conventional farming methods on peanut based on transcriptome and metabolomics. Food Res Int 2023; 171:113065. [PMID: 37330858 DOI: 10.1016/j.foodres.2023.113065] [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: 04/30/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/19/2023]
Abstract
To elucidate the nutritional quality of peanut under different farming methods, we selected two cultivars, "jihua13" and "jihua4", to grow in organic and conventional environments, respectively. After harvest, we measured physiological parameters and differential metabolites. Metabolomics showed that most of the amino acids, carbohydrates, and secondary metabolites in organically grown jihua4 were downregulated, which was completely the opposite in jihua13. Fatty acids associated with heart disease and hypertension are reduced in organically grown peanuts. In particular, the highly statistically significant tryptophan betaine seems to be used as a reference to distinguish between organic and conventional cultivation. Mechanisms leading to differences in crop chemical composition are explained by transcriptome analysis. The results of the transcriptome analysis indicated that organic cultivation largely affects the synthesis of amino acids and carbohydrates in jihua13. Combined analysis of transcriptome and metabolomics found that variety jihua13 is more sensitive to farming methods and produces more unsaturated fatty acids than jihua4.
Collapse
Affiliation(s)
- Shan Liang
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Yangjie Jiang
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Min Zhang
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
25
|
McReynolds C, Hammock B, Morisseau C. Regulatory lipid vicinal diols counteract the biological activity of epoxy fatty acids and can act as biomarkers and mechanisms for disease progression. Pharmacol Ther 2023; 248:108454. [PMID: 37268114 PMCID: PMC10529401 DOI: 10.1016/j.pharmthera.2023.108454] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) are essential fatty acids required for human health and are obtained primarily from food or synthesized in the body by highly regulated processes. The metabolites of these lipids, formed largely through the action of cyclooxygenase, lipoxygenase, or cytochrome P450 (CYP450) enzymes, are responsible for multiple biological functions including inflammation, tissue repair, cell proliferation, blood vessel permeability, and immune cell behavior. The role of these regulatory lipids in disease has been well studied since their discovery as druggable targets; however, the metabolites generated downstream of these pathways have only recently gained attention for regulating biology. Specifically, the biological activity of lipid vicinal diols formed from the metabolism of CYP450-generated epoxy fatty acids (EpFA) by epoxide hydrolases were previously thought to have little biological activity but increasingly are recognized as promoting inflammation and brown fat adipogenesis, and exciting neurons through the regulation of ion channel activity at low concentrations. These metabolites also appear to balance the action of the EpFA precursor. For example, EpFA demonstrate the ability to resolve inflammation and reduce pain, while some lipid diols, through opposing mechanisms, promote inflammation and pain. This review describes recent studies that highlight the role of regulatory lipids, focusing on the balance between EpFA and their diol metabolites in promoting or resolving disease.
Collapse
Affiliation(s)
| | - Bruce Hammock
- EicOsis, Davis, CA, United States of America; University of California, Davis, CA, United States of America
| | | |
Collapse
|
26
|
Li J, Liu X, Shi Y, Xie Y, Yang J, Du Y, Zhang A, Wu J. Differentiation in TCM patterns of chronic obstructive pulmonary disease by comprehensive metabolomic and lipidomic characterization. Front Immunol 2023; 14:1208480. [PMID: 37492573 PMCID: PMC10363632 DOI: 10.3389/fimmu.2023.1208480] [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: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 07/27/2023] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a complex disease involving inflammation, cell senescence, and autoimmunity. Dialectical treatment for COPD with traditional Chinese medicine (TCM) has the advantage of fewer side effects, more effective suppression of inflammation, and improved immune function. However, the biological base of TCM pattern differentiation in COPD remains unclear. Methods Liquid Chromatography-Quadrupole-Orbitrap mass spectrometry (LC-Q-Orbitrap MS/MS) based metabolomics and lipidomics were used to analyze the serum samples from COPD patients of three TCM patterns in Lung Qi Deficiency (n=65), Lung-Kidney Qi Deficiency (n=54), Lung-Spleen Qi Deficiency (n=52), and healthy subjects (n=41). Three cross-comparisons were performed to characterize metabolic markers for different TCM patterns of COPD vs healthy subjects. Results We identified 28, 8, and 16 metabolites with differential abundance between three TCM patterns of COPD vs healthy subjects, respectively, the metabolic markers included cortisol, hypoxanthine, fatty acids, alkyl-/alkenyl-substituted phosphatidylethanolamine, and phosphatidylcholine, etc. Three panels of metabolic biomarkers specific to the above three TCM patterns yielded areas under the receiver operating characteristic curve of 0.992, 0.881, and 0.928, respectively, with sensitivity of 97.1%, 88.6%, and 91.4%, respectively, and specificity of 96.4%, 81.8%, and 83.9%, respectively. Discussion Combining metabolomics and lipidomics can more comprehensively and accurately trace metabolic markers. As a result, the differences in metabolism were proven to underlie different TCM patterns of COPD, which provided evidence to aid our understanding of the biological basis of dialectical treatment, and can also serve as biomarkers for more accurate diagnosis.
Collapse
Affiliation(s)
- Jiansheng Li
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xinguang Liu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanmin Shi
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Xie
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jianya Yang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yan Du
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ang Zhang
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinyan Wu
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
27
|
Fu X, Wang Y, Zhao F, Cui R, Xie W, Liu Q, Yang W. Shared biological mechanisms of depression and obesity: focus on adipokines and lipokines. Aging (Albany NY) 2023; 15:5917-5950. [PMID: 37387537 PMCID: PMC10333059 DOI: 10.18632/aging.204847] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Depression and obesity are both common disorders currently affecting public health, frequently occurring simultaneously within individuals, and the relationship between these disorders is bidirectional. The association between obesity and depression is highly co-morbid and tends to significantly exacerbate metabolic and related depressive symptoms. However, the neural mechanism under the mutual control of obesity and depression is largely inscrutable. This review focuses particularly on alterations in systems that may mechanistically explain the in vivo homeostatic regulation of the obesity and depression link, such as immune-inflammatory activation, gut microbiota, neuroplasticity, HPA axis dysregulation as well as neuroendocrine regulators of energy metabolism including adipocytokines and lipokines. In addition, the review summarizes potential and future treatments for obesity and depression and raises several questions that need to be answered in future research. This review will provide a comprehensive description and localization of the biological connection between obesity and depression to better understand the co-morbidity of obesity and depression.
Collapse
Affiliation(s)
- Xiying Fu
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yicun Wang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Xie
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qianqian Liu
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Yang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
28
|
Zhang L, Wang L, Huang J, Jin Z, Guan J, Yu H, Zhang M, Yu M, Jiang H, Qiao Z. Effects of Aeromonas hydrophila infection on the intestinal microbiota, transcriptome, and metabolomic of common carp (Cyprinus carpio). FISH & SHELLFISH IMMUNOLOGY 2023:108876. [PMID: 37271325 DOI: 10.1016/j.fsi.2023.108876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Aeromonas hydrophila frequently has harmful effects on aquatic organisms. The intestine is an important defense against stress. In this study, we investigated the intestinal microbiota and transcriptomic and metabolomic responses of Cyprinus carpio subjected to A. hydrophila infection. The results showed that obvious variation in the intestinal microbiota was observed after infection, with increased levels of Firmicutes and Bacteroidetes and decreased levels of Proteobacteria. Several genera of putatively beneficial microbiota (Cetobacterium, Bacteroides, and Lactobacillus) were abundant, while Demequina, Roseomonas, Rhodobacter, Pseudoxanthomonas, and Cellvibrio were decreased; pathogenic bacteria of the genus Vibrio were increased after microbiota infection. The intestinal transcriptome revealed several immune-related differentially expressed genes associated with the cytokines and oxidative stress. The metabolomic analysis showed that microbiota infection disturbed the metabolic processes of the carp, particularly amino acid metabolism. This study provides insight into the underlying mechanisms associated with the intestinal microbiota, immunity, and metabolism of carp response to A. hydrophila infection; eleven stress-related metabolite markers were identified, including N-acetylglutamic acid, capsidiol, sedoheptulose 7-phosphate, prostaglandin B1, 8,9-DiHETrE, 12,13-DHOME, ADP, cellobiose, 1H-Indole-3-carboxaldehyde, sinapic acid and 5,7-dihydroxyflavone.
Collapse
Affiliation(s)
- Lan Zhang
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Lei Wang
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China.
| | - Jintai Huang
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Zhan Jin
- College of Fisheries, Henan Normal University, Xinxiang, China
| | - Junxiang Guan
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Hang Yu
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Meng Zhang
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Miao Yu
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Hongxia Jiang
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| | - Zhigang Qiao
- College of Fisheries, Henan Normal University, Xinxiang, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang, China; Engineering Lab of Henan Province for Aquatic Animal Disease Control, Henan Normal University, Xinxiang, China
| |
Collapse
|
29
|
Navarro-Perez J, Vidal-Puig A, Carobbio S. Recent developments in adipose tissue-secreted factors and their target organs. Curr Opin Genet Dev 2023; 80:102046. [PMID: 37099831 DOI: 10.1016/j.gde.2023.102046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/12/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023]
Abstract
The white adipose tissue's primary roles are to store and mobilise energy, which is very different from the brown adipose tissue's function of using fuel to generate heat and maintain the body temperature. The adipose tissues (ATs), co-ordinately with the other organs, sense energetic demands and inform of their reserves before embarking on energetically demanding physiological functions. It is not surprising that ATs exhibit highly integrated regulatory mechanisms mediated by a diversified secretome, including adipokines, lipokines, metabolites and a repertoire of extracellular miRNAs that contribute to integrating the function of the AT niche and connect the AT through paracrine and endocrine effects with the whole organism. Characterising the adipose secretome, its changes in health and disease, regulation by ageing and gender and their contribution to energy homoeostasis is necessary to optimise its use for personalised strategies to prevent or reverse metabolic diseases.
Collapse
Affiliation(s)
- Jaime Navarro-Perez
- Centro de Investigacion Principe Felipe, Valencia, Spain. https://twitter.com/@JaimeNavarroPr1
| | - Antonio Vidal-Puig
- Centro de Investigacion Principe Felipe, Valencia, Spain; Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
30
|
Lipo-nutritional quality of pork: the lipid composition, regulation, and molecular mechanisms of fatty acid deposition. ANIMAL NUTRITION 2023; 13:373-385. [DOI: 10.1016/j.aninu.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/13/2022] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
|
31
|
Enhanced Wound Healing Potential of Spirulina platensis Nanophytosomes: Metabolomic Profiling, Molecular Networking, and Modulation of HMGB-1 in an Excisional Wound Rat Model. Mar Drugs 2023; 21:md21030149. [PMID: 36976198 PMCID: PMC10051127 DOI: 10.3390/md21030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Excisional wounds are considered one of the most common physical injuries. This study aims to test the effect of a nanophytosomal formulation loaded with a dried hydroalcoholic extract of S. platensis on promoting excisional wound healing. The Spirulina platensis nanophytosomal formulation (SPNP) containing 100 mg PC and 50 mg CH exhibited optimum physicochemical characteristics regarding particle size (598.40 ± 9.68 nm), zeta potential (−19.8 ± 0.49 mV), entrapment efficiency (62.76 ± 1.75%), and Q6h (74.00 ± 1.90%). It was selected to prepare an HPMC gel (SPNP-gel). Through metabolomic profiling of the algal extract, thirteen compounds were identified. Molecular docking of the identified compounds on the active site of the HMGB-1 protein revealed that 12,13-DiHome had the highest docking score of −7.130 kcal/mol. SPNP-gel showed higher wound closure potential and enhanced histopathological alterations as compared to standard (MEBO® ointment) and S. platensis gel in wounded Sprague-Dawley rats. Collectively, NPS promoted the wound healing process by enhancing the autophagy process (LC3B/Beclin-1) and the NRF-2/HO-1antioxidant pathway and halting the inflammatory (TNF-, NF-κB, TlR-4 and VEGF), apoptotic processes (AIF, Caspase-3), and the downregulation of HGMB-1 protein expression. The present study’s findings suggest that the topical application of SPNP-gel possesses a potential therapeutic effect in excisional wound healing, chiefly by downregulating HGMB-1 protein expression.
Collapse
|
32
|
Gu X, Wang L, Liu S, Shan T. Adipose tissue adipokines and lipokines: Functions and regulatory mechanism in skeletal muscle development and homeostasis. Metabolism 2023; 139:155379. [PMID: 36538987 DOI: 10.1016/j.metabol.2022.155379] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/29/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Skeletal muscle plays important roles in normal biological activities and whole-body energy homeostasis in humans. The growth and development of skeletal muscle also directly influence meat production and meat quality in animal production. Therefore, regulating the development and homeostasis of skeletal muscle is crucial for human health and animal production. Adipose tissue, which includes white adipose tissue (WAT) and brown adipose tissue (BAT), not only functions as an energy reserve but also has attracted substantial attention because of its role as an endocrine organ. The novel signalling molecules known as "adipokines" and "lipokines" that are secreted by adipose tissue were identified through the secretomic technique, which broadened our understanding of the previously unknown crosstalk between adipose tissue and skeletal muscle. In this review, we summarize and discuss the secretory role of adipose tissues, both WAT and BAT, as well as the regulatory roles of various adipokines and lipokines in skeletal muscle development and homeostasis. We suggest that adipokines and lipokines have potential as drug candidates for the treatment of skeletal muscle dysfunction and related metabolic diseases and as promising nutrients for improving animal production.
Collapse
Affiliation(s)
- Xin Gu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Shiqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, China; Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
33
|
Nieman DC, Omar AM, Kay CD, Kasote DM, Sakaguchi CA, Lkhagva A, Weldemariam MM, Zhang Q. Almond intake alters the acute plasma dihydroxy-octadecenoic acid (DiHOME) response to eccentric exercise. Front Nutr 2023; 9:1042719. [PMID: 36698469 PMCID: PMC9868138 DOI: 10.3389/fnut.2022.1042719] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction This investigation determined if 4-weeks ingestion of nutrient-dense almonds mitigated post-exercise inflammation and muscle soreness and damage. Methods An acute 90-min of eccentric exercise (90-EE) was used to induce muscle damage in 64 non-obese adults not engaging in regular resistance training (ages 30-65 years, BMI < 30 kg/m2). Using a parallel group design, participants were randomized to almond (AL) (57 g/d) or cereal bar (CB) (calorie matched) treatment groups for a 4-week period prior to the 90-EE (17 exercises). Blood and 24-h urine samples were collected before and after supplementation, with additional blood samples collected immediately post-90-EE, and then daily during 4 additional days of recovery. Changes in plasma oxylipins, urinary gut-derived phenolics, plasma cytokines, muscle damage biomarkers, mood states, and exercise performance were assessed. Results The 90-EE protocol induced significant muscle damage, delayed onset of muscle soreness (DOMS), inflammation, reduced strength and power performance, and mood disturbance. Interaction effects (2 group × 7 time points) supported that AL vs. CB was associated with reduced post-exercise fatigue and tension (p = 0.051, 0.033, respectively) and higher levels of leg-back strength (p = 0.029). No group differences were found for post-90-EE increases in DOMS and six cytokines. AL was associated with lower levels of serum creatine kinase immediately- and 1-day post-exercise (p = 0.034 and 0.013, respectively). The 90-EE bout increased plasma levels immediately post-exercise for 13 oxylipins. Interaction effects revealed significantly higher levels for AL vs. CB for 12,13-DiHOME (p < 0.001) and lower levels for 9,10-DiHOME (p < 0.001). Urine levels increased in AL vs. CB for seven gut-derived phenolics including 5-(3',4'-dihydroxyphenyl)-γ-valerolactone that was inversely related to changes in plasma 9,10-DiHOME (r = -0.029, p = 0.021). Discussion These data support some positive effects of almond intake in improving mood state, retaining strength, decreasing muscle damage, increasing the generation of gut-derived phenolic metabolites, and altering the plasma oxylipin DiHOME response to unaccustomed eccentric exercise in untrained adults. The elevated post-exercise plasma levels of 12,13-DiHOME with almond intake support positive metabolic outcomes for adults engaging in unaccustomed eccentric exercise bouts.
Collapse
Affiliation(s)
- David C. Nieman
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus, Kannapolis, NC, United States,*Correspondence: David C. Nieman,
| | - Ashraf M. Omar
- UNCG Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| | - Colin D. Kay
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, North Carolina Research Campus, Kannapolis, NC, United States
| | - Deepak M. Kasote
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, North Carolina Research Campus, Kannapolis, NC, United States
| | - Camila A. Sakaguchi
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus, Kannapolis, NC, United States
| | - Ankhbayar Lkhagva
- UNCG Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| | - Mehari Muuz Weldemariam
- UNCG Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| | - Qibin Zhang
- UNCG Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| |
Collapse
|
34
|
Chen Y, Hu Q, Wang C, Wang T. The crosstalk between BAT thermogenesis and skeletal muscle dysfunction. Front Physiol 2023; 14:1132830. [PMID: 37153220 PMCID: PMC10160478 DOI: 10.3389/fphys.2023.1132830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Metabolic defects increase the risk of skeletal muscle diseases, and muscle impairment might worsen metabolic disruption, leading to a vicious cycle. Both brown adipose tissue (BAT) and skeletal muscle play important roles in non-shivering thermogenesis to regulate energy homeostasis. BAT regulates body temperature, systemic metabolism, and seretion of batokines that have positive or negative impacts on skeletal muscle. Conversely, muscle can secrete myokines that regulate BAT function. This review explained the crosstalk between BAT and skeletal muscle, and then discussed the batokines and highlighted their impact on skeletal muscle under physiological conditions. BAT is now considered a potential therapeutic target for obesity and diabetes treatment. Moreover, manipulation of BAT may be an attractive approach for the treatment of muscle weakness by correcting metabolic deficits. Therefore, exploring BAT as a potential treatment for sarcopenia could be a promising avenue for future research.
Collapse
Affiliation(s)
- Yao Chen
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Hu
- Health Management Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Changyi Wang, ; Tiantian Wang,
| |
Collapse
|
35
|
Quaranta A, Revol-Cavalier J, Wheelock CE. The octadecanoids: an emerging class of lipid mediators. Biochem Soc Trans 2022; 50:1569-1582. [PMID: 36454542 PMCID: PMC9788390 DOI: 10.1042/bst20210644] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 10/27/2023]
Abstract
Oxylipins are enzymatic and non-enzymatic metabolites of mono- or polyunsaturated fatty acids that encompass potent lipid mediators including the eicosanoids and docosanoids. Previously considered of low interest and often dismissed as 'just fat', octadecanoid oxylipins have only recently begun to be recognized as lipid mediators in humans. In the last few years, these compounds have been found to be involved in the mediation of multiple biological processes related to nociception, tissue modulation, cell proliferation, metabolic regulation, inflammation, and immune regulation. At the same time, the study of octadecanoids is hampered by a lack of standardization in the field, a paucity of analytical standards, and a lack of domain expertise. These issues have collectively limited the investigation of the biosynthesis and bioactivity of octadecanoids. Here, we present an overview of the primary enzymatic pathways for the oxidative metabolism of 18-carbon fatty acids in humans and of the current knowledge of the major biological activity of the resulting octadecanoids. We also propose a systematic nomenclature system based upon that used for the eicosanoids in order to avoid ambiguities and resolve multiple designations for the same octadecanoid. The aim of this review is to provide an initial framework for the field and to assist in its standardization as well as to increase awareness of this class of compounds in order to stimulate research into this interesting group of lipid mediators.
Collapse
Affiliation(s)
- Alessandro Quaranta
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Johanna Revol-Cavalier
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Larodan Research Laboratory, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Craig E. Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
36
|
Nikolic M, Novakovic J, Ramenskaya G, Kokorekin V, Jeremic N, Jakovljevic V. Cooling down with Entresto. Can sacubitril/valsartan combination enhance browning more than coldness? Diabetol Metab Syndr 2022; 14:175. [PMID: 36419097 PMCID: PMC9686067 DOI: 10.1186/s13098-022-00944-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND It is a growing importance to induce a new treatment approach to encourage weight loss but also to improve maintenance of lost weight. It has been shown that promotion of brown adipose tissue (BAT) function or acquisition of BAT characteristics in white adipose tissue (terms referred as "browning") can be protective against obesity. MAIN TEXT Amongst numerous established environmental influences on BAT activity, cold exposure is the best interested technique due to its not only effects on of BAT depots in proliferation process but also de novo differentiation of precursor cells via β-adrenergic receptor activation. A novel combination drug, sacubitril/valsartan, has been shown to be more efficient in reducing cardiovascular events and heart failure readmission compared to conventional therapy. Also, this combination of drugs increases the postprandial lipid oxidation contributing to energy expenditure, promotes lipolysis in adipocytes and reduces body weight. To date, there is no research examining potential of combined sacubitril/valsartan use to promote browning or mechanisms in the basis of this thermogenic process. CONCLUSION Due to the pronounced effects of cold and sacubitril/valsartan treatment on function and metabolism of BAT, the primary goal of further research should focused on investigation of the synergistic effects of the sacubitril/valsartan treatment at low temperature environmental conditions.
Collapse
Affiliation(s)
- Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | | | - Nevena Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
- First Moscow State Medical University IM Sechenov, Moscow, Russia.
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University IM Sechenov, Moscow, Russia
| |
Collapse
|
37
|
Time to Consider the “Exposome Hypothesis” in the Development of the Obesity Pandemic. Nutrients 2022; 14:nu14081597. [PMID: 35458158 PMCID: PMC9032727 DOI: 10.3390/nu14081597] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
The obesity epidemic shows no signs of abatement. Genetics and overnutrition together with a dramatic decline in physical activity are the alleged main causes for this pandemic. While they undoubtedly represent the main contributors to the obesity problem, they are not able to fully explain all cases and current trends. In this context, a body of knowledge related to exposure to as yet underappreciated obesogenic factors, which can be referred to as the “exposome”, merits detailed analysis. Contrarily to the genome, the “exposome” is subject to a great dynamism and variability, which unfolds throughout the individual’s lifetime. The development of precise ways of capturing the full exposure spectrum of a person is extraordinarily demanding. Data derived from epidemiological studies linking excess weight with elevated ambient temperatures, in utero, and intergenerational effects as well as epigenetics, microorganisms, microbiota, sleep curtailment, and endocrine disruptors, among others, suggests the possibility that they may work alone or synergistically as several alternative putative contributors to this global epidemic. This narrative review reports the available evidence on as yet underappreciated drivers of the obesity epidemic. Broadly based interventions are needed to better identify these drivers at the same time as stimulating reflection on the potential relevance of the “exposome” in the development and perpetuation of the obesity epidemic.
Collapse
|