1
|
Zhang W, Tian D, Yu Y, Tong D, Zhou W, Yu Y, Lu L, Li W, Liu G, Shi W. Micro/nanoplastics impair the feeding of goldfish by disrupting the complicated peripheral and central regulation of appetite. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:174112. [PMID: 38908581 DOI: 10.1016/j.scitotenv.2024.174112] [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: 04/23/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
The ubiquitous presence of plastic particles in water bodies poses a potential threat to aquatic species. Although numerous adverse effects of microplastics (MPs) and nanoplastics (NPs) have been documented, their effects on fish feeding, one of the most important behaviors of animals, are far from being fully understood. In this study, the effects of MPs and NPs (at environmentally realistic levels) on fish food consumption and feeding behavior were assessed using goldfish (Carassius auratus) and polystyrene (PS) particles as representatives. In addition, to reveal the potential mechanisms, the effects of MPs and NPs on peripheral and central regulation of appetite were evaluated by examining appetite-regulation related intestinal, serous, and hypothalamic parameters. The results obtained indicated that the 28-day MP- and NP-exposure significantly impaired goldfish feeding by disrupting peripheral and central appetite regulation. Based on differences observed in their effects on the abovementioned behavioral, histological, and physiological parameters, MPs and NPs may interfere with appetite regulation in a size-dependent manner. Blocking the gastrointestinal tract and causing histopathological and functional damage to inner organs may be the main routes through which MPs and NPs disrupt appetite regulation. Our findings suggested that plastic particles exposure may have far-reaching effects on fish species through impaired feeding, which warrants further attention.
Collapse
Affiliation(s)
- Weixia Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dandan Tian
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihan Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Difei Tong
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weishang Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingying Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingzheng Lu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weifeng Li
- College of Marine Sciences, Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Beibu Gulf University, Qinzhou 535011, PR China
| | - Guangxu Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Wei Shi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
2
|
Carrageta DF, Pereira SC, Ferreira R, Monteiro MP, Oliveira PF, Alves MG. Signatures of metabolic diseases on spermatogenesis and testicular metabolism. Nat Rev Urol 2024; 21:477-494. [PMID: 38528255 DOI: 10.1038/s41585-024-00866-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 03/27/2024]
Abstract
Diets leading to caloric overload are linked to metabolic disorders and reproductive function impairment. Metabolic and hormonal abnormalities stand out as defining features of metabolic disorders, and substantially affect the functionality of the testis. Metabolic disorders induce testicular metabolic dysfunction, chronic inflammation and oxidative stress. The disruption of gastrointestinal, pancreatic, adipose tissue and testicular hormonal regulation induced by metabolic disorders can also contribute to a state of compromised fertility. In this Review, we will delve into the effects of high-fat diets and metabolic disorders on testicular metabolism and spermatogenesis, which are crucial elements for male reproductive function. Moreover, metabolic disorders have been shown to influence the epigenome of male gametes and might have a potential role in transmitting phenotype traits across generations. However, the existing evidence strongly underscores the unmet need to understand the mechanisms responsible for transgenerational paternal inheritance of male reproductive function impairment related to metabolic disorders. This knowledge could be useful for developing targeted interventions to prevent, counteract, and most of all break the perpetuation chain of male reproductive dysfunction associated with metabolic disorders across generations.
Collapse
Affiliation(s)
- David F Carrageta
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Sara C Pereira
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana P Monteiro
- Clinical and Experimental Endocrinology, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE & Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Marco G Alves
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Campus de Santiago Agra do Crasto, Aveiro, Portugal.
| |
Collapse
|
3
|
Xiang Y, Sun J, Ma G, Dai X, Meng Y, Fu C, Zhang Y, Zhao Q, Li J, Zhang S, Zheng Z, Li X, Fu L, Li K, Qi X. Integrating Multi-Omics Data to Identify Key Functional Variants Affecting Feed Efficiency in Large White Boars. Genes (Basel) 2024; 15:980. [PMID: 39202341 PMCID: PMC11353296 DOI: 10.3390/genes15080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Optimizing feed efficiency through the feed conversion ratio (FCR) is paramount for economic viability and sustainability. In this study, we integrated RNA-seq, ATAC-seq, and genome-wide association study (GWAS) data to investigate key functional variants associated with feed efficiency in pigs. Identification of differentially expressed genes in the duodenal and muscle tissues of low- and high-FCR pigs revealed that pathways related to digestion of dietary carbohydrate are responsible for differences in feed efficiency between individuals. Differential open chromatin regions identified by ATAC-seq were linked to genes involved in glycolytic and fatty acid processes. GWAS identified 211 significant single-nucleotide polymorphisms associated with feed efficiency traits, with candidate genes PPP1R14C, TH, and CTSD. Integration of duodenal ATAC-seq data and GWAS data identified six key functional variants, particularly in the 1500985-1509676 region on chromosome 2. In those regions, CTSD was found to be highly expressed in the duodenal tissues of pigs with a high feed conversion ratio, suggesting its role as a potential target gene. Overall, the integration of multi-omics data provided insights into the genetic basis of feed efficiency, offering valuable information for breeding more efficient pig breeds.
Collapse
Affiliation(s)
- Yue Xiang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Jiahui Sun
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Guojian Ma
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Xueting Dai
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Yuan Meng
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Chong Fu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Yan Zhang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Qiulin Zhao
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Jingjin Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Saixian Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Zhuqing Zheng
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Xinyun Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Liangliang Fu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (J.S.); (G.M.); (X.D.); (C.F.); (Y.Z.); (Q.Z.); (Z.Z.); (X.L.); (L.F.)
| | - Kui Li
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| | - Xiaolong Qi
- Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China; (Y.X.); (Y.M.); (J.L.); (S.Z.); (K.L.)
| |
Collapse
|
4
|
Cheong KL, Liu K, Chen W, Zhong S, Tan K. Recent progress in Porphyra haitanensis polysaccharides: Extraction, purification, structural insights, and their impact on gastrointestinal health and oxidative stress management. Food Chem X 2024; 22:101414. [PMID: 38711774 PMCID: PMC11070828 DOI: 10.1016/j.fochx.2024.101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024] Open
Abstract
Porphyra haitanensis, a red seaweed species, represents a bountiful and sustainable marine resource. P. haitanensis polysaccharide (PHP), has garnered considerable attention for its numerous health benefits. However, the comprehensive utilization of PHP on an industrial scale has been limited by the lack of comprehensive information. In this review, we endeavor to discuss and summarize recent advancements in PHP extraction, purification, and characterization. We emphasize the multifaceted mechanisms through which PHP promotes gastrointestinal health. Furthermore, we present a summary of compelling evidence supporting PHP's protective role against oxidative stress. This includes its demonstrated potent antioxidant properties, its ability to neutralize free radicals, and its capacity to enhance the activity of antioxidant enzymes. The information presented here also lays the theoretical groundwork for future research into the structural and functional aspects of PHP, as well as its potential applications in functional foods.
Collapse
Affiliation(s)
- Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Keying Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Wenting Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Karsoon Tan
- Guangxi Key Laboratory of Beibu Gulf Biodiversity Conservation, Beibu Gulf University, Qinzhou, Guangxi, China
| |
Collapse
|
5
|
Saneyasu T. Recent Research on Mechanisms of Feeding Regulation in Chicks. J Poult Sci 2024; 61:2024012. [PMID: 38681189 PMCID: PMC11039390 DOI: 10.2141/jpsa.2024012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Food intake affects poultry productivity. A complete understanding of these regulatory mechanisms provides new strategies to improve productivity. Food intake is regulated by complex mechanisms involving many factors, including the central nervous system, gastrointestinal tract, hormones, and nutrients. Although several studies have been conducted to elucidate regulatory mechanisms in chickens, the mechanisms remain unclear. To update the current knowledge on feeding regulation in chickens, this review focuses on recent findings that have not been summarized in previous reviews, including spexins, adipokines, neurosecretory proteins GL and GM, and central intracellular signaling factors.
Collapse
Affiliation(s)
- Takaoki Saneyasu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501,
Japan
| |
Collapse
|
6
|
Shao R, Liao X, Wang W, Lan Y, Zhang H, Du Q, Jiao L, Yin Z, Ai Q, Mai K, Wan M. Vitamin D regulates glucose metabolism in zebrafish (Danio rerio) by maintaining intestinal homeostasis. J Nutr Biochem 2024; 123:109473. [PMID: 37844767 DOI: 10.1016/j.jnutbio.2023.109473] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/22/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023]
Abstract
Vitamin D (VD) is a steroid hormone that is widely known to play an important role in maintaining mineral homeostasis, and regulating various physiological functions. Our previous results demonstrated that the interruption of VD metabolism caused hyperglycemia in zebrafish. In the present study we further explored the mechanism that VD regulates glucose metabolism by maintaining intestinal homeostasis in zebrafish. Our results showed that the expression of several peptide hormones including gastric inhibitory peptide, peptide YY, and fibroblast growth factor 19 in the intestine decreased, while the expression of sodium glucose cotransporter-1 and gcg was increased in the intestine of the zebrafish fed with the VD3-deficient diet. Consistently, similar results were obtained in cyp2r1-/- zebrafish, in which endogenous VD metabolism is blocked. Furthermore, the results obtained from germ-free zebrafish exhibited that VD-regulated glucose metabolism was partly dependent on the microbiota in zebrafish. Importantly, the transplantation of gut microbiota collected from cyp2r1-/- zebrafish to germ-free zebrafish led to hyperglycemic symptoms in the fish, which were associated with the altered structure and functions of the microbiota in cyp2r1-/- zebrafish. Interestingly, the treatments with acetate or Cetobacterium somerae, a potent acetate producer, lowered the glucose contents whereas augmented insulin expression in zebrafish larvae. Notably, acetate supplementation alleviated hyperglycemia in cyp2r1-/- zebrafish and other diabetic zebrafish. In conclusion, our study has demonstrated that VD modulates the gut microbiota-SCFAs-gastrointestinal hormone axis, contributing to the maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Rui Shao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Xinmeng Liao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Wentao Wang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Yawen Lan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Hui Zhang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Qingyang Du
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Lin Jiao
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China
| | - Min Wan
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture & Key Laboratory of Mariculture, Ministry of Education, College of Fisheries, Ocean University of China, Qingdao, China.
| |
Collapse
|
7
|
Ferreira-Hermosillo A, de Miguel Ibañez R, Pérez-Dionisio EK, Villalobos-Mata KA. Obesity as a Neuroendocrine Disorder. Arch Med Res 2023; 54:102896. [PMID: 37945442 DOI: 10.1016/j.arcmed.2023.102896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Obesity is one of the most prevalent diseases in the world. Based on hundreds of clinical and basic investigations, its etiopathogenesis goes beyond the simple imbalance between energy intake and expenditure. The center of the regulation of appetite and satiety lies in the nuclei of the hypothalamus where peripheral signals derived from adipose tissue (e.g., leptin), the gastrointestinal tract, the pancreas, and other brain structures, arrive. These signals are part of the homeostatic control system (eating to survive). Additionally, a hedonic or reward system (eating for pleasure) is integrated into the regulation of appetite. This reward system consists of a dopaminergic circuit that affects eating-related behaviors influencing food preferences, food desires, gratification when eating, and impulse control to avoid compulsions. These systems are not separate. Indeed, many of the hormones that participate in the homeostatic system also participate in the regulation of the hedonic system. In addition, factors such as genetic and epigenetic changes, certain environmental and sociocultural elements, the microbiota, and neuronal proinflammatory effects of high-energy diets also contribute to the development of obesity. Therefore, obesity can be considered a complex neuroendocrine disease, and all of the aforementioned components should be considered for the management of obesity.
Collapse
Affiliation(s)
- Aldo Ferreira-Hermosillo
- Endocrine Research Unit, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| | - Regina de Miguel Ibañez
- Endocrinology Service, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Enid Karina Pérez-Dionisio
- Endocrinology Service, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Karen Alexandra Villalobos-Mata
- Endocrinology Service, Hospital de Especialidades del Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
8
|
Chaiyasoot K, Khumkhana N, Deekum W, Chaichana C, Taweerutchana V, Srisuworanan N, Pramyothin P. Alteration of BDNF, SPARC, FGF-21, and GDF-15 circulating levels after 1 year of anti-obesity treatments and their association with 1-year weight loss. Endocrine 2023; 82:57-68. [PMID: 37436597 PMCID: PMC10462550 DOI: 10.1007/s12020-023-03435-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/16/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE Emerging evidence revealed that brain-derived neurotrophic factor (BDNF), secreted protein acidic and rich in cysteine (SPARC), fibroblast growth factor 21(FGF-21) and growth differentiation factor 15 (GDF-15) are involved in energy metabolism and body weight regulation. Our study aimed at examining their association with BMI, their alterations after anti-obesity treatments, and their association with 1-year weight loss. METHODS A prospective observational study of 171 participants with overweight and obesity and 46 lean controls was established. All participants received lifestyle educational intervention (LEI) with or without anti-obesity treatments (LEI + bariatric/metabolic surgery, n = 41; LEI + topiramate, n = 46; LEI + liraglutide, n = 31; LEI + orlistat, n = 12; and LEI alone, n = 41). Anthropometric and metabolic parameters, insulin sensitivity, C-reactive protein (CRP), fasting plasma levels of BDNF, SPARC, GDF-15, and FGF-21 were measured at baseline and 1 year. RESULTS Multiple linear regression showed that fasting levels of SPARC, FGF-21, and GDF-15 were significantly associated with baseline BMI after adjustment for age and sex. At 1 year, the average weight loss was 4.8% in the entire cohort with a significant improvement in glycemia, insulin sensitivity, and CRP. Multiple linear regression adjusted for age, sex, baseline BMI, type of treatment, and presence of T2DM revealed that the decrease in log10FGF-21 and log10GDF-15 at 1 year from baseline was significantly associated with a greater percentage of weight loss at 1 year. CONCLUSIONS This study highlights the association of SPARC, FGF-21, and GDF-15 levels with BMI. Decreased circulating levels of GDF-15 and FGF-21 were associated with greater weight loss at 1 year, regardless of the types of anti-obesity modalities.
Collapse
Affiliation(s)
- Kusuma Chaiyasoot
- Division of Nutrition, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Nanta Khumkhana
- Division of Nutrition, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Wanjan Deekum
- Division of Nutrition, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Chartchai Chaichana
- Siriraj Center of Research Excellence for Diabetes and Obesity, Mahidol University, Bangkok, Thailand
| | - Voraboot Taweerutchana
- Division of Minimal Invasive Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nicha Srisuworanan
- Division of Minimal Invasive Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornpoj Pramyothin
- Division of Nutrition, Department of Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
9
|
Lee J, Kim WK. Applications of Enteroendocrine Cells (EECs) Hormone: Applicability on Feed Intake and Nutrient Absorption in Chickens. Animals (Basel) 2023; 13:2975. [PMID: 37760373 PMCID: PMC10525316 DOI: 10.3390/ani13182975] [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: 08/14/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
This review focuses on the role of hormones derived from enteroendocrine cells (EECs) on appetite and nutrient absorption in chickens. In response to nutrient intake, EECs release hormones that act on many organs and body systems, including the brain, gallbladder, and pancreas. Gut hormones released from EECs play a critical role in the regulation of feed intake and the absorption of nutrients such as glucose, protein, and fat following feed ingestion. We could hypothesize that EECs are essential for the regulation of appetite and nutrient absorption because the malfunction of EECs causes severe diarrhea and digestion problems. The importance of EEC hormones has been recognized, and many studies have been carried out to elucidate their mechanisms for many years in other species. However, there is a lack of research on the regulation of appetite and nutrient absorption by EEC hormones in chickens. This review suggests the potential significance of EEC hormones on growth and health in chickens under stress conditions induced by diseases and high temperature, etc., by providing in-depth knowledge of EEC hormones and mechanisms on how these hormones regulate appetite and nutrient absorption in other species.
Collapse
Affiliation(s)
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
10
|
Wei R, Li D, Jia S, Chen Y, Wang J. MC4R in Central and Peripheral Systems. Adv Biol (Weinh) 2023; 7:e2300035. [PMID: 37043700 DOI: 10.1002/adbi.202300035] [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: 01/21/2023] [Revised: 02/25/2023] [Indexed: 04/14/2023]
Abstract
Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
11
|
Chaukimath P, Frankel G, Visweswariah SS. The metabolic impact of bacterial infection in the gut. FEBS J 2023; 290:3928-3945. [PMID: 35731686 DOI: 10.1111/febs.16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 06/02/2022] [Accepted: 06/21/2022] [Indexed: 08/17/2023]
Abstract
Bacterial infections of the gut are one of the major causes of morbidity and mortality worldwide. The interplay between the pathogen and the host is finely balanced, with the bacteria evolving to proliferate and establish infection. In contrast, the host mounts a response to first restrict and then eliminate the infection. The intestine is a rapidly proliferating tissue, and metabolism is tuned to cater to the demands of proliferation and differentiation along the crypt-villus axis (CVA) in the gut. As bacterial pathogens encounter the intestinal epithelium, they elicit changes in the host cell, and core metabolic pathways such as the tricarboxylic acid (TCA) cycle, lipid metabolism and glycolysis are affected. This review highlights the mechanisms utilized by diverse gut bacterial pathogens to subvert host metabolism and describes host responses to the infection.
Collapse
Affiliation(s)
- Pooja Chaukimath
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Gad Frankel
- Centre for Molecular Bacteriology and Infection and Department of Life Sciences, Imperial College, London, UK
| | - Sandhya S Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
12
|
Kuckuck S, van der Valk ES, Scheurink AJW, van der Voorn B, Iyer AM, Visser JA, Delhanty PJD, van den Berg SAA, van Rossum EFC. Glucocorticoids, stress and eating: The mediating role of appetite-regulating hormones. Obes Rev 2023; 24:e13539. [PMID: 36480471 PMCID: PMC10077914 DOI: 10.1111/obr.13539] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
Disrupted hormonal appetite signaling plays a crucial role in obesity as it may lead to uncontrolled reward-related eating. Such disturbances can be induced not only by weight gain itself but also by glucocorticoid overexposure, for example, due to chronic stress, disease, or medication use. However, the exact pathways are just starting to be understood. Here, we present a conceptual framework of how glucocorticoid excess may impair hormonal appetite signaling and, consequently, eating control in the context of obesity. The evidence we present suggests that counteracting glucocorticoid excess can lead to improvements in appetite signaling and may therefore pose a crucial target for obesity prevention and treatment. In turn, targeting hormonal appetite signals may not only improve weight management and eating behavior but may also decrease detrimental effects of glucocorticoid excess on cardio-metabolic outcomes and mood. We conclude that gaining a better understanding of the relationship between glucocorticoid excess and circulating appetite signals will contribute greatly to improvements in personalized obesity prevention and treatment.
Collapse
Affiliation(s)
- Susanne Kuckuck
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Eline S van der Valk
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anton J W Scheurink
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Bibian van der Voorn
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Anand M Iyer
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| | - Jenny A Visser
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Patric J D Delhanty
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands
| | - Sjoerd A A van den Berg
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Department of Clinical Chemistry, Erasmus MC, Rotterdam, Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, Netherlands.,Obesity Center CGG, Erasmus MC, Room Rg528, P.O. Box 2040, Rotterdam, 3000 CA, Netherlands
| |
Collapse
|
13
|
Kotronarou K, Charalambous A, Evangelou A, Georgiou O, Demetriou A, Apidianakis Y. Dietary Stimuli, Intestinal Bacteria and Peptide Hormones Regulate Female Drosophila Defecation Rate. Metabolites 2023; 13:metabo13020264. [PMID: 36837883 PMCID: PMC9965912 DOI: 10.3390/metabo13020264] [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: 01/20/2023] [Revised: 02/09/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Peptide hormones control Drosophila gut motility, but the intestinal stimuli and the gene networks coordinating this trait remain poorly defined. Here, we customized an assay to quantify female Drosophila defecation rate as a proxy of intestinal motility. We found that bacterial infection with the human opportunistic bacterial pathogen Pseudomonas aeruginosa (strain PA14) increases defecation rate in wild-type female flies, and we identified specific bacteria of the fly microbiota able to increase defecation rate. In contrast, dietary stress, imposed by either water-only feeding or high ethanol consumption, decreased defecation rate and the expression of enteroendocrine-produced hormones in the fly midgut, such as Diuretic hormone 31 (Dh31). The decrease in defecation due to dietary stress was proportional to the impact of each stressor on fly survival. Furthermore, we exploited the Drosophila Genetic Reference Panel wild type strain collection and identified strains displaying high and low defecation rates. We calculated the narrow-sense heritability of defecation rate to be 91%, indicating that the genetic variance observed using our assay is mostly additive and polygenic in nature. Accordingly, we performed a genome-wide association (GWA) analysis revealing 17 candidate genes linked to defecation rate. Downregulation of four of them (Pmp70, CG11307, meso18E and mub) in either the midgut enteroendocrine cells or in neurons reduced defecation rate and altered the midgut expression of Dh31, that in turn regulates defecation rate via signaling to the visceral muscle. Hence, microbial and dietary stimuli, and Dh31-controlling genes, regulate defecation rate involving signaling within and among neuronal, enteroendocrine, and visceral muscle cells.
Collapse
|
14
|
Cai X, Bao D, Hua R, Cai B, Wang L, Dong R, Hua L. A Comparative Study on the Distribution Pattern of Endocrine Cells in the Gastrointestinal Tract of Two Small Alpine Mammals, Plateau Zokor ( Eospalax baileyi) and Plateau Pika ( Ochotona curzoniae). Animals (Basel) 2023; 13:ani13040640. [PMID: 36830427 PMCID: PMC9951659 DOI: 10.3390/ani13040640] [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: 12/06/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Endocrine cells can secrete a variety of gastrointestinal hormones that regulate gastrointestinal digestion and absorption, which, in turn, play an important role in animal growth, metabolism, and acclimation. The small alpine mammals, plateau zokor (Eospalax baileyi) and plateau pika (Ochotona curzoniae), live in a unique ecotope with cold, hypoxic environments and short plant-growing seasons, resulting in differential adaptive digestive strategies for foods. Studying the distribution pattern of endocrine cells in the gastrointestinal tract (GIT) of these two animals can lead to a better understanding of the survival strategies of animals in an alpine environment. In this study, we used histochemical and immunohistochemical methods to compare the distribution pattern of argyrophilic cells and the expression of 5-HT cells, Gas cells, and Glu cells in the GIT of the plateau zokor with those of the plateau pika. The results showed that these endocrine cells we studied were widely distributed in the gastrointestinal organs of both these small mammals, and their morphology and distribution location in the GIT were almost the same. However, there were significant differences in the distribution density of argyrophilic cells between different organs in the GIT. The distribution density of argyrophilic cells in the duodenum, jejunum, ileum, and rectum of plateau zokor was significantly lower than that of plateau pika (p < 0.05) and, in the cecum of plateau zokor, was significantly higher than that of plateau pika (p < 0.001). The positive expression of 5-HT cells in the corpus I, corpus II, and pylorus of the stomach, duodenum, ileum, and rectum of plateau zokor was significantly higher than that of plateau pika (p < 0.01). In addition, the positive expression of Glu cells in the cecum was significantly higher (p < 0.01) and in the duodenum and colon was significantly lower (p < 0.05) in the plateau zokor than in the plateau pika. We conclude that the distribution pattern of endocrine cells in the GIT is consistent with the respective animals' diets, with the plateau zokor feeding on high-fiber roots and plateau pika preferring to intake the aboveground parts of plants with lower fibers.
Collapse
|
15
|
Rolls ET. The orbitofrontal cortex, food reward, body weight and obesity. Soc Cogn Affect Neurosci 2023; 18:nsab044. [PMID: 33830272 PMCID: PMC9997078 DOI: 10.1093/scan/nsab044] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/17/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
In primates including humans, the orbitofrontal cortex is the key brain region representing the reward value and subjective pleasantness of the sight, smell, taste and texture of food. At stages of processing before this, in the insular taste cortex and inferior temporal visual cortex, the identity of the food is represented, but not its affective value. In rodents, the whole organisation of reward systems appears to be different, with reward value reflected earlier in processing systems. In primates and humans, the amygdala is overshadowed by the great development of the orbitofrontal cortex. Social and cognitive factors exert a top-down influence on the orbitofrontal cortex, to modulate the reward value of food that is represented in the orbitofrontal cortex. Recent evidence shows that even in the resting state, with no food present as a stimulus, the liking for food, and probably as a consequence of that body mass index, is correlated with the functional connectivity of the orbitofrontal cortex and ventromedial prefrontal cortex. This suggests that individual differences in these orbitofrontal cortex reward systems contribute to individual differences in food pleasantness and obesity. Implications of how these reward systems in the brain operate for understanding, preventing and treating obesity are described.
Collapse
Affiliation(s)
- Edmund T Rolls
- Oxford Centre for Computational Neuroscience, Oxford, UK
- Department of Computer Science, University of Warwick, Coventry, UK
| |
Collapse
|
16
|
The Bidirectional Relationship of NPY and Mitochondria in Energy Balance Regulation. Biomedicines 2023; 11:biomedicines11020446. [PMID: 36830982 PMCID: PMC9953676 DOI: 10.3390/biomedicines11020446] [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: 01/12/2023] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Energy balance is regulated by several hormones and peptides, and neuropeptide Y is one of the most crucial in feeding and energy expenditure control. NPY is regulated by a series of peripheral nervous and humoral signals that are responsive to nutrient sensing, but its role in the energy balance is also intricately related to the energetic status, namely mitochondrial function. During fasting, mitochondrial dynamics and activity are activated in orexigenic neurons, increasing the levels of neuropeptide Y. By acting on the sympathetic nervous system, neuropeptide Y modulates thermogenesis and lipolysis, while in the peripheral sites, it triggers adipogenesis and lipogenesis instead. Moreover, both central and peripheral neuropeptide Y reduces mitochondrial activity by decreasing oxidative phosphorylation proteins and other mediators important to the uptake of fatty acids into the mitochondrial matrix, inhibiting lipid oxidation and energy expenditure. Dysregulation of the neuropeptide Y system, as occurs in metabolic diseases like obesity, may lead to mitochondrial dysfunction and, consequently, to oxidative stress and to the white adipose tissue inflammatory environment, contributing to the development of a metabolically unhealthy profile. This review focuses on the interconnection between mitochondrial function and dynamics with central and peripheral neuropeptide Y actions and discusses possible therapeutical modulations of the neuropeptide Y system as an anti-obesity tool.
Collapse
|
17
|
Male Sex Hormones, Metabolic Syndrome, and Aquaporins: A Triad of Players in Male (in)Fertility. Int J Mol Sci 2023; 24:ijms24031960. [PMID: 36768282 PMCID: PMC9915845 DOI: 10.3390/ijms24031960] [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: 12/30/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Infertility is becoming a chronic and emerging problem in the world. There is a resistant stigma that this health condition is mostly due to the female, although the literature supports that the responsibility for the onset of infertility is equally shared between both sexes in more or less equal proportions. Nevertheless, male sex hormones, particularly testosterone (T), are key players in male-related infertility. Indeed, hypogonadism, which is also characterized by changes in T levels, is one of the most common causes of male infertility and its incidence has been interconnected to the increased prevalence of metabolic diseases. Recent data also highlight the role of aquaporin (AQP)-mediated water and solute diffusion and the metabolic homeostasis in testicular cells suggesting a strong correlation between AQPs function, metabolism of testicular cells, and infertility. Indeed, recent studies showed that both metabolic and sexual hormone concentrations can change the expression pattern and function of AQPs. Herein, we review up-to-date information on the involvement of AQP-mediated function and permeability in men with metabolic syndrome and testosterone deficit, highlighting the putative mechanisms that show an interaction between sex hormones, AQPs, and metabolic syndrome that may contribute to male infertility.
Collapse
|
18
|
Zhu W, Oteiza PI. Proanthocyanidins at the gastrointestinal tract: mechanisms involved in their capacity to mitigate obesity-associated metabolic disorders. Crit Rev Food Sci Nutr 2022; 64:220-240. [PMID: 35943169 DOI: 10.1080/10408398.2022.2105802] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The prevalence of overweight and obesity is continually increasing worldwide. Obesity is a major public health concern given the multiple associated comorbidities. Finding dietary approaches to prevent/mitigate these conditions is of critical relevance. Proanthocyanidins (PACs), oligomers or polymers of flavan-3-ols that are extensively distributed in nature, represent a major part of total dietary polyphenols. Although current evidence supports the capacity of PACs to mitigate obesity-associated comorbidities, the underlying mechanisms remain speculative due to the complexity of PACs' structure. Given their limited bioavailability, the major site of the biological actions of intact PACs is the gastrointestinal (GI) tract. This review discusses the actions of PACs at the GI tract which could underlie their anti-obesity effects. These mechanisms include: i) inhibition of digestive enzymes at the GI lumen, including pancreatic lipase, α-amylase, α-glucosidase; ii) modification of gut microbiota composition; iii) modulation of inflammation- and oxidative stress-triggered signaling pathways, e.g. NF-κB and MAPKs; iv) protection of the GI barrier integrity. Further understanding of the mechanisms and biological activities of PACs at the GI tract can contribute to develop nutritional and pharmacological strategies oriented to mitigate the serious comorbidities of obesity.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Nutrition, University of California, Davis, California, USA
- Department of Environmental Toxicology, University of California, Davis, California, USA
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, California, USA
- Department of Environmental Toxicology, University of California, Davis, California, USA
| |
Collapse
|
19
|
Damasceno ROS, Soares PMG, Barbosa ALDR, Nicolau LAD, Medeiros JVR, Souza MHLP. Modulatory Role of Carbon Monoxide on the Inflammatory Response and Oxidative Stress Linked to Gastrointestinal Disorders. Antioxid Redox Signal 2022; 37:98-114. [PMID: 34806398 DOI: 10.1089/ars.2020.8223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Carbon monoxide (CO) is an endogenous gaseous mediator that plays an important role in maintaining gastrointestinal (GI) tract homeostasis, acting in mucosal defense, and providing negative modulation of pathophysiological markers of clinical conditions. Recent Advances: Preclinical studies using animal models and/or cell culture show that CO can modulate the inflammatory response and oxidative stress in GI mucosal injuries and pathological conditions, reducing proinflammatory cytokines and reactive oxygen species, while increasing antioxidant defense mechanisms. Critical Issues: CO has potent anti-inflammatory and antioxidant effects. The defense mechanisms of the GI tract are subject to aggression by different chemical agents (e.g., drugs and ethanol) as well as complex and multifactorial diseases, with inflammation and oxidative stress as strong triggers for the deleterious effects. Thus, it is possible that CO acts on a variety of molecules involved in the inflammatory and oxidative signaling cascades, as well as reinforcing several defense mechanisms that maintain GI homeostasis. Future Directions: CO-based therapies are promising tools for the treatment of GI disorders, such as gastric and intestinal injuries, inflammatory bowel disease, and pancreatitis. Therefore, it is necessary to develop safe and selective CO-releasing agents and/or donor drugs to facilitate effective treatments and methods for analysis of CO levels that are simple and inexpensive. Antioxid. Redox Signal. 37, 98-114.
Collapse
Affiliation(s)
| | | | | | | | - Jand-Venes Rolim Medeiros
- Biotechnology and Biodiversity Center Research, Federal University of the Parnaíba Delta, Parnaíba, Brazil
| | | |
Collapse
|
20
|
Angelidi AM, Belanger MJ, Kokkinos A, Koliaki CC, Mantzoros CS. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr Rev 2022; 43:507-557. [PMID: 35552683 PMCID: PMC9113190 DOI: 10.1210/endrev/bnab034] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 02/08/2023]
Abstract
Recent insights into the pathophysiologic underlying mechanisms of obesity have led to the discovery of several promising drug targets and novel therapeutic strategies to address the global obesity epidemic and its comorbidities. Current pharmacologic options for obesity management are largely limited in number and of modest efficacy/safety profile. Therefore, the need for safe and more efficacious new agents is urgent. Drugs that are currently under investigation modulate targets across a broad range of systems and tissues, including the central nervous system, gastrointestinal hormones, adipose tissue, kidney, liver, and skeletal muscle. Beyond pharmacotherapeutics, other potential antiobesity strategies are being explored, including novel drug delivery systems, vaccines, modulation of the gut microbiome, and gene therapy. The present review summarizes the pathophysiology of energy homeostasis and highlights pathways being explored in the effort to develop novel antiobesity medications and interventions but does not cover devices and bariatric methods. Emerging pharmacologic agents and alternative approaches targeting these pathways and relevant research in both animals and humans are presented in detail. Special emphasis is given to treatment options at the end of the development pipeline and closer to the clinic (ie, compounds that have a higher chance to be added to our therapeutic armamentarium in the near future). Ultimately, advancements in our understanding of the pathophysiology and interindividual variation of obesity may lead to multimodal and personalized approaches to obesity treatment that will result in safe, effective, and sustainable weight loss until the root causes of the problem are identified and addressed.
Collapse
Affiliation(s)
- Angeliki M Angelidi
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew J Belanger
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Chrysi C Koliaki
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Frühbeck G, Becerril S, Martín M, Ramírez B, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Silva C, Burrell MA, Escalada J, Rodríguez A. High plasma and lingual uroguanylin as potential contributors to changes in food preference after sleeve gastrectomy. Metabolism 2022; 128:155119. [PMID: 34990711 DOI: 10.1016/j.metabol.2021.155119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/25/2021] [Accepted: 12/27/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND The biological mediators supporting long-term weight loss and changes in dietary choice behaviour after sleeve gastrectomy remain unclear. Guanylin and uroguanylin are gut hormones involved in the regulation of satiety, food preference and adiposity. Thus, we sought to analyze whether the guanylin system is involved in changes in food preference after sleeve gastrectomy in obesity. METHODS Proguanylin (GUCA2A) and prouroguanylin (GUCA2B) were determined in patients with severe obesity (n = 41) as well as in rats with diet-induced obesity (n = 48), monogenic obesity (Zucker fa/fa) (n = 18) or in a food choice paradigm (normal diet vs high-fat diet) (n = 16) submitted to sleeve gastrectomy. Lingual distribution and expression of guanylins (GUCA2A and GUCA2B) and their receptor GUCY2C as well as the fatty acid receptor CD36 were evaluated in the preclinical models. RESULTS Circulating concentrations of GUCA2A and GUCA2B were increased after sleeve gastrectomy in patients with severe obesity as well as in rats with diet-induced and monogenic (fa/fa) obesity. Interestingly, the lower dietary fat preference observed in obese rats under the food choice paradigm as well as in patients with obesity after sleeve gastrectomy were negatively associated with post-surgical GUCA2B levels. Moreover, sleeve gastrectomy upregulated the low expression of GUCA2A and GUCA2B in taste bud cells of tongues from rats with diet-induced and monogenic (fa/fa) obesity in parallel to a downregulation of the lingual lipid sensor CD36. CONCLUSIONS The increased circulating and lingual GUCA2B after sleeve gastrectomy suggest an association between the uroguanylin-GUCY2C endocrine axis and food preference through the regulation of gustatory responses.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marina Martín
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - María A Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
22
|
Martínez-Herrero S, Martínez A. Adrenomedullin: Not Just Another Gastrointestinal Peptide. Biomolecules 2022; 12:biom12020156. [PMID: 35204657 PMCID: PMC8961556 DOI: 10.3390/biom12020156] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 12/11/2022] Open
Abstract
Adrenomedullin (AM) and proadrenomedullin N-terminal 20 peptide (PAMP) are two bioactive peptides derived from the same precursor with several biological functions including vasodilation, angiogenesis, or anti-inflammation, among others. AM and PAMP are widely expressed throughout the gastrointestinal (GI) tract where they behave as GI hormones, regulating numerous physiological processes such as gastric emptying, gastric acid release, insulin secretion, bowel movements, or intestinal barrier function. Furthermore, it has been recently demonstrated that AM/PAMP have an impact on gut microbiome composition, inhibiting the growth of bacteria related with disease and increasing the number of beneficial bacteria such as Lactobacillus or Bifidobacterium. Due to their wide functions in the GI tract, AM and PAMP are involved in several digestive pathologies such as peptic ulcer, diabetes, colon cancer, or inflammatory bowel disease (IBD). AM is a key protective factor in IBD onset and development, as it regulates cytokine production in the intestinal mucosa, improves vascular and lymphatic regeneration and function and mucosal epithelial repair, and promotes a beneficial gut microbiome composition. AM and PAMP are relevant GI hormones that can be targeted to develop novel therapeutic agents for IBD, other GI disorders, or microbiome-related pathologies.
Collapse
|
23
|
Prasad H, Mathew JKK, Visweswariah SS. Receptor Guanylyl Cyclase C and Cyclic GMP in Health and Disease: Perspectives and Therapeutic Opportunities. Front Endocrinol (Lausanne) 2022; 13:911459. [PMID: 35846281 PMCID: PMC9276936 DOI: 10.3389/fendo.2022.911459] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Receptor Guanylyl Cyclase C (GC-C) was initially characterized as an important regulator of intestinal fluid and ion homeostasis. Recent findings demonstrate that GC-C is also causally linked to intestinal inflammation, dysbiosis, and tumorigenesis. These advances have been fueled in part by identifying mutations or changes in gene expression in GC-C or its ligands, that disrupt the delicate balance of intracellular cGMP levels and are associated with a wide range of clinical phenotypes. In this review, we highlight aspects of the current knowledge of the GC-C signaling pathway in homeostasis and disease, emphasizing recent advances in the field. The review summarizes extra gastrointestinal functions for GC-C signaling, such as appetite control, energy expenditure, visceral nociception, and behavioral processes. Recent research has expanded the homeostatic role of GC-C and implicated it in regulating the ion-microbiome-immune axis, which acts as a mechanistic driver in inflammatory bowel disease. The development of transgenic and knockout mouse models allowed for in-depth studies of GC-C and its relationship to whole-animal physiology. A deeper understanding of the various aspects of GC-C biology and their relationships with pathologies such as inflammatory bowel disease, colorectal cancer, and obesity can be leveraged to devise novel therapeutics.
Collapse
Affiliation(s)
- Hari Prasad
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
| | | | - Sandhya S. Visweswariah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bengaluru, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, India
- *Correspondence: Sandhya S. Visweswariah,
| |
Collapse
|
24
|
Telese A, Sehgal V, Magee CG, Naik S, Alqahtani S, Lovat L, Haidry RJ. Bariatric and Metabolic Endoscopy: A New Paradigm. Clin Transl Gastroenterol 2021; 12:e00364. [PMID: 34142665 PMCID: PMC8216681 DOI: 10.14309/ctg.0000000000000364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/14/2021] [Indexed: 12/26/2022] Open
Abstract
The prevalence of obesity, type 2 diabetes mellitus, and metabolic syndromes is increasing globally. Minimally invasive metabobariatric (MB) endoscopic therapies are adjunct treatments that can potentially bridge the gap between surgical interventions and medical therapy. A growing number of MB techniques are becoming available, allowing for more personalized and patient-targeted treatment options for specific disease states. MB techniques are less invasive than surgery and can precisely target different parts of the gastrointestinal tract that may be responsible for the pathophysiology of obesity and metabolic syndromes such as type 2 diabetes mellitus. These alternatives should be selected on an individualized patient basis to balance the expected clinical outcomes and desired anatomical targets with the level of invasiveness and degree of acceptable risk. Each MB intervention presents great flexibility allowing for a tailored intervention and different levels of patient engagement. Patient awareness and motivation are essential to avoid therapy withdrawal and failure. Differences between MB procedures in terms of weight loss and metabolic benefit will be discussed in this review, along with the insights on clinical decision-making processes to evaluate the potential of further evolution and growth of bariatric and metabolic endoscopy.
Collapse
Affiliation(s)
- Andrea Telese
- Department of Gastrointestinal Services, University College London Hospital, London, UK;
| | - Vinay Sehgal
- Department of Gastrointestinal Services, University College London Hospital, London, UK;
| | - Cormac G. Magee
- Department of Gastrointestinal Services, University College London Hospital, London, UK;
- Centre for Obesity research, University College London, London, UK;
| | - S. Naik
- Centre for Obesity research, University College London, London, UK;
| | - S.A. Alqahtani
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, Maryland, USA;
- Liver Transplantation Unit, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - L.B. Lovat
- Department of Gastrointestinal Services, University College London Hospital, London, UK;
- Division of Surgery and Interventional Science, University College London, London, UK.
| | - Rehan J. Haidry
- Department of Gastrointestinal Services, University College London Hospital, London, UK;
| |
Collapse
|
25
|
Saneyasu T, Ueno M, Nagata K, Kewan A, Honda K, Kamisoyama H. Central administration of insulin and refeeding lead to Akt and ERK phosphorylation in the chicken medulla. Neurosci Lett 2021; 758:136008. [PMID: 34098027 DOI: 10.1016/j.neulet.2021.136008] [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: 01/13/2021] [Revised: 05/18/2021] [Accepted: 05/29/2021] [Indexed: 10/21/2022]
Abstract
The purpose of this study was to investigate whether medullary cellular signaling pathways contribute to feeding regulation in chickens. Fasting inhibited the phosphorylated protein and its rates of ERK but not Akt in the chicken medulla, while refeeding promoted Akt and ERK. Intraperitoneal administration of sulfate cholecystokinin 8 did not affect medullary Akt and ERK phosphorylation in chickens. Intracerebroventricular administration of insulin significantly induced the phosphorylation of Akt and ERK in the chicken medulla. These findings suggest that the medullary Akt and ERK pathways are involved in the appetite-suppressive pathway of insulin in chickens.
Collapse
Affiliation(s)
- Takaoki Saneyasu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | - Mizuki Ueno
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kanami Nagata
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Ahmed Kewan
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuhisa Honda
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Hiroshi Kamisoyama
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
26
|
Artificial intelligence in disease diagnostics: A critical review and classification on the current state of research guiding future direction. HEALTH AND TECHNOLOGY 2021. [DOI: 10.1007/s12553-021-00555-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AbstractThe diagnosis of diseases is decisive for planning proper treatment and ensuring the well-being of patients. Human error hinders accurate diagnostics, as interpreting medical information is a complex and cognitively challenging task. The application of artificial intelligence (AI) can improve the level of diagnostic accuracy and efficiency. While the current literature has examined various approaches to diagnosing various diseases, an overview of fields in which AI has been applied, including their performance aiming to identify emergent digitalized healthcare services, has not yet been adequately realized in extant research. By conducting a critical review, we portray the AI landscape in diagnostics and provide a snapshot to guide future research. This paper extends academia by proposing a research agenda. Practitioners understand the extent to which AI improves diagnostics and how healthcare benefits from it. However, several issues need to be addressed before successful application of AI in disease diagnostics can be achieved.
Collapse
|
27
|
Lv L, Jiang H, Chen Y, Gu S, Xia J, Zhang H, Lu Y, Yan R, Li L. The faecal metabolome in COVID-19 patients is altered and associated with clinical features and gut microbes. Anal Chim Acta 2021; 1152:338267. [PMID: 33648648 PMCID: PMC7847702 DOI: 10.1016/j.aca.2021.338267] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
Although SARS-CoV-2 can invade the intestine, though its effect on digestion and absorption is not fully understood. In the present study, 56 COVID-19 patients and 47 age- and sex-matched healthy subjects were divided into a discovery cohort and a validation cohort. Blood, faeces and clinical information were collected from the patients in the hospital and at discharge. The faecal metabolome was analysed using gas chromatography-mass spectrometry, and Spearman's correlation analyses of clinical features, the serum metabolome, and the faecal micro- and mycobiota were conducted. The results showed that, the faeces of COVID-19 patients were enriched with important nutrients that should be metabolized or absorbed, such as sucrose and 2-palmitoyl-glycerol; diet-related components that cannot be synthesized by humans, such as 1,5-anhydroglucitol and D-pinitol; and harmful metabolites, such as oxalate, were also detected. In contrast, purine metabolites such as deoxyinosine and hypoxanthine, low-water-soluble long-chain fatty alcohols/acids such as behenic acid, compounds rarely occurring in nature such as D-allose and D-arabinose, and microbe-related compounds such as 2,4-di-tert-butylphenol were depleted in the faeces of COVID-19 patients. Moreover, these metabolites significantly correlated with altered serum metabolites such as oxalate and gut microbesincluding Ruminococcaceae, Actinomyces, Sphingomonas and Aspergillus. Although levels of several faecal metabolites, such as sucrose, 1,5-anhydroglucitol and D-pinitol, of discharged patients were not different from those of healthy controls (HCs), those of oxalate and 2-palmitoyl-glycerol did differ. Therefore, alterations in the faecal metabolome of COVID-19 patients may reflect malnutrition and intestinal inflammation and warrant greater attention. The results of present study provide new insights into the pathogenesis and treatment of COVID-19.
Collapse
Affiliation(s)
- Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Hua Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Yingfeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China
| | - Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, Hangzhou, China.
| |
Collapse
|
28
|
Nóbrega S, Monteiro MP, Pereira-da-Silva L, Pereira SS, Hartmann B, Holst JJ, Barbosa Silva R, Cordeiro-Ferreira G. Congenital Glucagon-like Peptide-1 Deficiency in the Pathogenesis of Protracted Diarrhea in Mitchell-Riley Syndrome. J Clin Endocrinol Metab 2021; 106:1084-1090. [PMID: 33382423 DOI: 10.1210/clinem/dgaa916] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Mitchell-Riley syndrome due to RFX6 gene mutations is characterized by neonatal diabetes and protracted diarrhea. The RFX6 gene encodes a transcription factor involved in enteroendocrine cell differentiation required for beta-cell maturation. In contrast to the pathway by which RFX6 mutations leads to diabetes, the mechanisms underlying protracted diarrhea are unknown. OBJECTIVE To assess whether glucagon-like peptide-1 (GLP-1) was involved in the pathogenesis of Mitchell-Riley syndrome protracted diarrhea. METHODS Two case report descriptions. in a tertiary pediatric hospital. "Off-label" treatment with liraglutide. We describe 2 children diagnosed with Mitchell-Riley syndrome, presenting neonatal diabetes and protracted diarrhea. Both patients had nearly undetectable GLP-1 plasma levels and absence of GLP-1 immunostaining in distal intestine and rectum. The main outcome was to evaluate whether GLP-1 analogue therapy could improve Mitchell-Riley syndrome protracted diarrhea. RESULTS "Off-label" liraglutide treatment, licensed for type 2 diabetes treatment in children, was started as rescue therapy for protracted intractable diarrhea resulting in rapid improvement during the course of 12 months. CONCLUSION Congenital GLP-1 deficiency was identified in patients with Mitchell-Riley syndrome. The favorable response to liraglutide further supports GLP-1 involvement in the pathogenesis of protracted diarrhea and its potential therapeutic use.
Collapse
Affiliation(s)
- Sara Nóbrega
- Special Nutrition and Respiratory Care Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- Gastroenterology and Hepatology Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), Medicine of Woman, Childhood and Adolescence, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Luís Pereira-da-Silva
- Comprehensive Health Research Centre (CHRC), Medicine of Woman, Childhood and Adolescence, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- NICU, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Sofia S Pereira
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Raul Barbosa Silva
- Special Nutrition and Respiratory Care Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Gonçalo Cordeiro-Ferreira
- Special Nutrition and Respiratory Care Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- Gastroenterology and Hepatology Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC), Medicine of Woman, Childhood and Adolescence, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
- NICU, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| |
Collapse
|
29
|
Dong Y, Yin J, Zhang Y, Chen JDZ. Electronic Bypass for Diabetes: Optimization of Stimulation Parameters and Mechanisms of Glucagon-Like Peptide-1. Neuromodulation 2021; 25:1097-1105. [PMID: 33538043 DOI: 10.1111/ner.13367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/16/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Intestinal electrical stimulation (IES) has been proposed for treating diabetes; however, its parameters need to be further systematically optimized. This study aimed to optimize the parameters of IES and investigate its possible mechanisms involving glucagon-like peptide-1 (GLP-1) in diabetic rats. MATERIALS AND METHODS Thirty-six high-fat diet-induced diabetic rats were chronically implanted with a pair of bipolar electrodes at the duodenum for IES. The oral glucose tolerance test (OGTT) was performed in a number of sessions with IES using different parameters and biphasic charge-balanced waveforms to derive the best values for train on-time, pulse frequency, and pulse width. Incretin hormones such as GLP-1 were assessed and the GLP-1 antagonist Exendin 9-39 was used to assess the role of GLP-1 in the ameliorating effect of IES on hyperglycemia. RESULTS The most effective IES parameters in reducing blood glucose (BG) during the OGTT were derived: 1.2 sec on, 0.3 sec off, 80 Hz, 3 msec. IES with these parameters reduced BG level by at least 29% from 15 min to 180 min (p < 0.05 for all points, N = 10). IES with these stimulation parameters increased plasma GLP-1 level at 30 min, 60 min, 90 min and gastric inhibitory peptide (GIP) level at 30 min (N = 8). Exendin 9-39 blocked the inhibitory effect of IES on BG (p > 0.05, IES + Exendin 9-39 vs. sham-IES, N = 8). CONCLUSION IES with the most effective parameters derived in this study improves hyperglycemia in diabetic rats. The ameliorating effect of IES on hyperglycemia is attributed to the enhanced release of GLP-1. IES has great potential for treating diabetes.
Collapse
Affiliation(s)
- Yan Dong
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jieyun Yin
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiling Zhang
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Guimarães M, Pereira SS, Monteiro MP. From Entero-Endocrine Cell Biology to Surgical Interventional Therapies for Type 2 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:273-297. [PMID: 32016913 DOI: 10.1007/5584_2020_480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The physiological roles of the enteroendocrine system in relation to energy and glucose homeostasis regulation have been extensively studied in the past few decades. Considerable advances were made that enabled to disclose the potential use of gastro-intestinal (GI) hormones to target obesity and type 2 diabetes (T2D). The recognition of the clinical relevance of these discoveries has led the pharmaceutical industry to design several hormone analogues to either to mitigate physiological defects or target pharmacologically T2D.Amongst several advances, a major breakthrough in the field was the unexpected observation that enteroendocrine system modulation to T2D target could be achieved by surgically induced anatomical rearrangement of the GI tract. These findings resulted from the widespread use of bariatric surgery procedures for obesity treatment, which despite initially devised to induce weight loss by limiting the systemic availably of nutrients, are now well recognized to influence GI hormone dynamics in a manner that is highly dependent on the type of anatomical rearrangement produced.This chapter will focus on enteroendocrine system related mechanisms leading to improved glycemic control in T2D after bariatric surgery interventions.
Collapse
Affiliation(s)
- Marta Guimarães
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Sofia S Pereira
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal. .,Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| |
Collapse
|
31
|
Smitka K, Prochazkova P, Roubalova R, Dvorak J, Papezova H, Hill M, Pokorny J, Kittnar O, Bilej M, Tlaskalova-Hogenova H. Current Aspects of the Role of Autoantibodies Directed Against Appetite-Regulating Hormones and the Gut Microbiome in Eating Disorders. Front Endocrinol (Lausanne) 2021; 12:613983. [PMID: 33953692 PMCID: PMC8092392 DOI: 10.3389/fendo.2021.613983] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
The equilibrium and reciprocal actions among appetite-stimulating (orexigenic) and appetite-suppressing (anorexigenic) signals synthesized in the gut, brain, microbiome and adipose tissue (AT), seems to play a pivotal role in the regulation of food intake and feeding behavior, anxiety, and depression. A dysregulation of mechanisms controlling the energy balance may result in eating disorders such as anorexia nervosa (AN) and bulimia nervosa (BN). AN is a psychiatric disease defined by chronic self-induced extreme dietary restriction leading to an extremely low body weight and adiposity. BN is defined as out-of-control binge eating, which is compensated by self-induced vomiting, fasting, or excessive exercise. Certain gut microbiota-related compounds, like bacterial chaperone protein Escherichia coli caseinolytic protease B (ClpB) and food-derived antigens were recently described to trigger the production of autoantibodies cross-reacting with appetite-regulating hormones and neurotransmitters. Gut microbiome may be a potential manipulator for AT and energy homeostasis. Thus, the regulation of appetite, emotion, mood, and nutritional status is also under the control of neuroimmunoendocrine mechanisms by secretion of autoantibodies directed against neuropeptides, neuroactive metabolites, and peptides. In AN and BN, altered cholinergic, dopaminergic, adrenergic, and serotonergic relays may lead to abnormal AT, gut, and brain hormone secretion. The present review summarizes updated knowledge regarding the gut dysbiosis, gut-barrier permeability, short-chain fatty acids (SCFA), fecal microbial transplantation (FMT), blood-brain barrier permeability, and autoantibodies within the ghrelin and melanocortin systems in eating disorders. We expect that the new knowledge may be used for the development of a novel preventive and therapeutic approach for treatment of AN and BN.
Collapse
Affiliation(s)
- Kvido Smitka
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
- *Correspondence: Kvido Smitka,
| | - Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiri Dvorak
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Hana Papezova
- Psychiatric Clinic, Eating Disorder Center, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Martin Hill
- Steroid Hormone and Proteofactors Department, Institute of Endocrinology, Prague, Czechia
| | - Jaroslav Pokorny
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
| | - Otomar Kittnar
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
| | - Martin Bilej
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
32
|
Zhang W, Waise TMZ, Toshinai K, Tsuchimochi W, Naznin F, Islam MN, Tanida R, Sakoda H, Nakazato M. Functional interaction between Ghrelin and GLP-1 regulates feeding through the vagal afferent system. Sci Rep 2020; 10:18415. [PMID: 33116243 PMCID: PMC7595212 DOI: 10.1038/s41598-020-75621-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract transmits feeding-regulatory signals to the brain via neuronal and hormonal pathways. Here we studied the interaction between the orexigenic gastric peptide, ghrelin, and the anorectic intestinal peptide, glucagon-like peptide 1 (GLP-1), in terms of feeding regulation via the vagal afferents. GLP-1 preadministration 30 min before ghrelin administration to rats and mice abolished ghrelin-induced food intake, while ghrelin preadministration abolished the anorectic effect of GLP-1. Ghrelin preadministration suppressed GLP-1-induced Fos expression in the nodose ganglia (NG). Electrophysiological assessment confirmed that the initially administered peptide abolished the vagal afferent electrical alteration induced by the subsequently administered peptide. Both the growth hormone secretagogue receptor (GHSR) and the GLP-1 receptor (GLP-1R) are co-localised in a major proportion of NG neurons that innervate the stomach. In these Ghsr+Glp1r+ neurons, ghrelin preadministration abolished the GLP-1-induced calcium response. Ghrelin generated a hyperpolarising current and GLP-1 generated a depolarising current in isolated NG neurons in a patch-clamp experiment. Ghrelin and GLP-1 potently influenced each other in terms of vagally mediated feeding regulation. This peptidergic interaction allows for fine control of the electrophysiological properties of NG neurons.
Collapse
Affiliation(s)
- Weidong Zhang
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - T M Zaved Waise
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.,Toronto General Hospital Research Institute, UHN, Toronto, ON, M5G 1L7, Canada
| | - Koji Toshinai
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.,Department of Sports and Fitness, Faculty of Wellness, Shigakkan University, 55 Nakoyama, Yokone, Obu, 474-8651, Japan
| | - Wakaba Tsuchimochi
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Farhana Naznin
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.,Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada
| | - Md Nurul Islam
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Ryota Tanida
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.,Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology, and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan. .,AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan.
| |
Collapse
|
33
|
Crisóstomo L, Pereira SC, Monteiro MP, Raposo JF, Oliveira PF, Alves MG. Lifestyle, metabolic disorders and male hypogonadism - A one-way ticket? Mol Cell Endocrinol 2020; 516:110945. [PMID: 32707080 DOI: 10.1016/j.mce.2020.110945] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/27/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Hypogonadism is more frequent among men with common metabolic diseases, notably obesity and type 2 diabetes. Indeed, endocrine disruption caused by metabolic diseases can trigger the onset of hypogonadism, although the underlying molecular mechanisms are not entirely understood. Metabolic diseases are closely related to unhealthy lifestyle choices, such as dietary habits and sedentarism. Therefore, hypogonadism is part of a pathological triad gathering unhealthy lifestyle, metabolic disease and genetic background. Additionally, hypogonadism harbors the potential to aggravate underlying metabolic disorders, further sustaining the mechanisms leading to disease. To what extent does lifestyle intervention in men suffering from these metabolic disorders can prevent, improve or reverse hypogonadism, is still controversial. Moreover, recent evidence suggests that the metabolic status of the father is related to the risk of inter and transgenerational inheritance of hypogonadism. In this review, we will address the proposed mechanisms of disease, as well as currently available interventions for hypogonadism.
Collapse
Affiliation(s)
- Luís Crisóstomo
- Department of Microscopy, Laboratory of Cell Biology, And Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal
| | - Sara C Pereira
- Department of Microscopy, Laboratory of Cell Biology, And Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal
| | - Mariana P Monteiro
- Department of Anatomy, And Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal
| | - João F Raposo
- NOVA Medical School - New University Lisbon, Lisbon, Portugal; APDP - Diabetes Portugal, Lisbon, Portugal
| | - Pedro F Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, And Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal.
| |
Collapse
|
34
|
Koliaki C, Liatis S, Dalamaga M, Kokkinos A. The Implication of Gut Hormones in the Regulation of Energy Homeostasis and Their Role in the Pathophysiology of Obesity. Curr Obes Rep 2020; 9:255-271. [PMID: 32647952 DOI: 10.1007/s13679-020-00396-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the role of gut hormones and their interactions in the regulation of energy homeostasis, describes gut hormone adaptations in obesity and in response to weight loss, and summarizes the current evidence on the role of gut hormone-based therapies for obesity treatment. RECENT FINDINGS Gut hormones play a key role in regulating eating behaviour, energy and glucose homeostasis. Dysregulated gut hormone responses have been proposed to be pathogenetically involved in the development and perpetuation of obesity. Summarizing the major gut hormone changes in obesity, obese individuals are characterized by blunted postprandial ghrelin suppression, loss of premeal ghrelin peaks, impaired diurnal ghrelin variability and reduced fasting and postprandial levels of anorexigenic peptides. Adaptive alterations of gut hormone levels are implicated in weight regain, thus complicating hypocaloric dietary interventions, and can further explain the profound weight loss and metabolic improvement following bariatric surgery. A plethora of compounds mimicking gut hormone changes after bariatric surgery are currently under investigation, introducing a new era in the pharmacotherapy of obesity. The current trend is to combine different gut hormone receptor agonists and target multiple systems simultaneously, in order to replicate as closely as possible the gut hormone milieu after bariatric surgery and circumvent the counter-regulatory adaptive changes associated with dietary energy restriction. An increasing number of preclinical and early-phase clinical trials reveal the additive benefits obtained with dual or triple gut peptide receptor agonists in reducing body weight and improving glycaemia. Gut hormones act as potent regulators of energy and glucose homeostasis. Therapeutic strategies targeting their levels or receptors emerge as a promising approach to treat patients with obesity and hyperglycaemia.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece.
| | - Stavros Liatis
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| |
Collapse
|
35
|
Purnell JQ. Management of chronic diseases in preventive cardiology: Revisiting “the Problem of Obesity.”. Am J Prev Cardiol 2020; 1:100005. [PMID: 34327449 PMCID: PMC8315319 DOI: 10.1016/j.ajpc.2020.100005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 11/29/2022] Open
|
36
|
Ghoshal UC, Goel A, Quigley EMM. Gut microbiota abnormalities, small intestinal bacterial overgrowth, and non-alcoholic fatty liver disease: An emerging paradigm. Indian J Gastroenterol 2020; 39:9-21. [PMID: 32291578 DOI: 10.1007/s12664-020-01027-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Evidence accumulates to implicate a role for the gut microbiota in non-alcoholic fatty liver disease (NAFLD)-a disorder that has reached almost epidemic proportions around the globe. For some time a disturbance in the gut microbiome, small intestinal bacterial overgrowth (SIBO), has been described among patients with liver disease, in general, and in the development and progression of NAFLD to nonalcoholic steatohepatitis (NASH), decompensated liver disease and hepatocellular cancer (HCC), in particular. More recently and permitted by the advent of high-throughput sequencing and allied molecular techniques, a much more detailed analysis of gut microbiota in NAFLD and NASH has become possible. In animal models, several mechanisms have been delineated which reveal how gut bacteria and their products could promote steatosis, hepatic inflammation, fibrosis, cirrhosis, and carcinogenesis. For understandable reasons evidence from human studies is less complete, but here again a plausible case is beginning to emerge to incriminate microbiota in NAFLD and NASH pathogenesis. Therapeutic interventions based on the modulation of the microbiome have been explored to some extent, but their application to everyday medical practice is still in the future.
Collapse
Affiliation(s)
- Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India
| | - Amit Goel
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India
| | - Eamonn M M Quigley
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226 014, India. .,Gastroenterology and Hepatology, Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, 6550 Fannin St, SM 1201, Houston, TX, 77030, USA.
| |
Collapse
|
37
|
Ohki J, Sakashita A, Aihara E, Inaba A, Uchiyama H, Matsumoto M, Ninomiya Y, Yamane T, Oishi Y, Iwatsuki K. Comparative analysis of enteroendocrine cells and their hormones between mouse intestinal organoids and native tissues. Biosci Biotechnol Biochem 2020; 84:936-942. [PMID: 31916916 DOI: 10.1080/09168451.2020.1713043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endocrine cells in the gastrointestinal tract secrete multiple hormones to maintain homeostasis in the body. In the present study, we generated intestinal organoids from the duodenum, jejunum, and ileum of Neurogenin 3 (Ngn3)-EGFP mice and examined how enteroendocrine cells (EECs) within organoid cultures resemble native epithelial cells in the gut. Transcriptome analysis of EGFP-positive cells from Ngn3-EGFP organoids showed gene expression pattern comparable to EECs in vivo. We also compared mRNAs of five major hormones, namely, ghrelin (Ghrl), cholecystokinin (Cck), Gip, secretin (Sct), and glucagon (Gcg) in organoids and small intestine along the longitudinal axis and found that expression patterns of these hormones in organoids were similar to those in native tissues. These findings suggest that an intestinal organoid culture system can be utilized as a suitable model to study enteroendocrine cell functions in vitro.
Collapse
Affiliation(s)
- Junko Ohki
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan.,Department of Health and Nutrition, Tsukuba International University, Ibaraki, Japan
| | - Akihiko Sakashita
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eitaro Aihara
- Department of Pharmacology and System Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Akihiko Inaba
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Hironobu Uchiyama
- Genome Research Center, Tokyo University of Agriculture, Tokyo, Japan
| | | | - Yuzo Ninomiya
- Division of Sensory Physiology, Research & Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan.,Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Takumi Yamane
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Yuichi Oishi
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| | - Ken Iwatsuki
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
38
|
Dai F, Yin J, Chen JDZ. Effects and Mechanisms of Vagal Nerve Stimulation on Body Weight in Diet-Induced Obese Rats. Obes Surg 2020; 30:948-956. [DOI: 10.1007/s11695-019-04365-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Lobato CB, Pereira SS, Guimarães M, Hartmann B, Wewer Albrechtsen NJ, Hilsted L, Holst JJ, Nora M, Monteiro MP. A Potential Role for Endogenous Glucagon in Preventing Post-Bariatric Hypoglycemia. Front Endocrinol (Lausanne) 2020; 11:608248. [PMID: 33424773 PMCID: PMC7793799 DOI: 10.3389/fendo.2020.608248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/22/2020] [Indexed: 01/03/2023] Open
Abstract
Obesity and obesity-related diseases are major public health concerns that have been exponentially growing in the last decades. Bariatric surgery is an effective long-term treatment to achieve weight loss and obesity comorbidity remission. Post-bariatric hypoglycemia (PBH) is a late complication of bariatric surgery most commonly reported after Roux-en-Y gastric bypass (RYGB). PBH is the end result of postprandial hyperinsulinemia but additional endocrine mechanisms involved are still under debate. Our aim was to characterize entero-pancreatic hormone dynamics associated with postprandial hypoglycemia after RYGB. Individuals previously submitted to RYGB (N=23) in a single tertiary hospital presenting PBH symptoms (Sym, n=14) and asymptomatic weight-matched controls (Asy, n=9) were enrolled. Participants underwent a mixed-meal tolerance test (MMTT) to assess glucose, total amino acids (total AA), insulin, C-peptide, glucagon, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and neurotensin (NT). We found that hypoglycemia during the MMTT was equally frequent in Sym and Asy groups (p=1.000). Re-grouped according to glucose nadir during the MMTT (Hypo n=11 vs NoHypo n=12; nadir <3.05 mmol/l vs ≥3.05 mmol/l), subjects presented no differences in anthropometric (BMI: p=0.527) or metabolic features (HbA1c: p=0.358), yet distinct meal-elicited hormone dynamics were identified. Postprandial glucose excursion and peak glucose levels were similar (p>0.05), despite distinct late glycemic outcomes (t=60 min and t=90 min: p<0.01), with overall greater glycemic variability in Hypo group (minimum-to-maximum glucose ratio: p<0.001). Hypo group meal-triggered hormone profile was characterized by lower early glucagon (t=15 min: p<0.01) and higher insulin (t=30 min: p<0.05, t=45 min: p<0.001), C-peptide (t=30 min: p<0.01, t=45 min: p<0.001, t=60 min: p<0.05), and GLP-1 (t=45 min: p<0.05) levels. Hyperinsulinemia was an independent risk factor for hypoglycemia (p<0.05). After adjusting for hyperinsulinemia, early glucagon correlated with glycemic nadir (p<0.01), and prevented postprandial hypoglycemia (p<0.05). A higher insulin to glucagon balance in Hypo was observed (p<0.05). No differences were observed in total AA, GIP or NT excursions (p>0.05). In sum, after RYGB, postprandial hyperinsulinemia is key in triggering PBH, but a parallel and earlier rise in endogenous glucagon might sustain the inter-individual variability in glycemic outcome beyond the effect of hyperinsulinism, advocating a potential pivotal role for glucagon in preventing hyperinsulinemic hypoglycemia.
Collapse
Affiliation(s)
- Carolina B. Lobato
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Sofia S. Pereira
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Marta Guimarães
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J. Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Linda Hilsted
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J. Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mário Nora
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Mariana P. Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- *Correspondence: Mariana P. Monteiro,
| |
Collapse
|
40
|
Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. S100 proteins in obesity: liaisons dangereuses. Cell Mol Life Sci 2020; 77:129-147. [PMID: 31363816 PMCID: PMC11104817 DOI: 10.1007/s00018-019-03257-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| |
Collapse
|
41
|
Dietary adzuki bean paste dose-dependently reduces visceral fat accumulation in rats fed a normal diet. Food Res Int 2019; 130:108890. [PMID: 32156348 DOI: 10.1016/j.foodres.2019.108890] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/29/2019] [Accepted: 12/12/2019] [Indexed: 01/15/2023]
Abstract
The aim of this study was to evaluate the dose-dependent effect of adzuki bean (Vigna angularis) paste (ABP) on visceral fat accumulation in rats. ABP is a rich source of indigestible carbohydrates (18.5%) with fiber and resistant starch (RS) contents of 14.5% and 4.0%, respectively. Animals were fed one of the following diets, control (CON), 30% ABP or 58.9% ABP for 28 days. The daily dietary energy intake was lowered (p < 0.05) and reduced visceral fat accumulation and lower serum lipid levels were observed in ABP fed groups. ABP consumption dose-dependently increased (p < 0.05) the daily fecal lipid and fecal acidic sterol excretions. On the other hand, cecal content and fecal moisture content in the 58.9% ABP group were greater (p < 0.05) than the CON group, while there was no significant difference between the two ABP fed groups. Both 30% and 58.9% ABP diets had significantly (p < 0.05) higher contents of cecal acetic, propionic and n-butyric acids, and lowered cecal pH, independently of the ABP dose. Microbial community data of rats fed ABP diets exhibited higher alpha-diversities than the rats fed CON diet, based on the Shannon Index and the number of observed species index, where the two ABP groups exhibited a similar alpha diversity. The weighted UniFrac-based principal coordinate analysis plot of cecal microbial community data showed that the ABP had a substantial effect on the cecal microbial composition. Furthermore, cecal bacterial 16S rRNA gene sequencing revealed that the ABP supplemented diets decreased the ratio of Firmicutes to Bacteroidetes. These findings suggested that the cecal fermentation of fiber and RS in ABP, might have decreased the energy intake, altered the gut microbiota composition, increased fecal lipid output, and thereby reduced fat accumulation in rats.
Collapse
|
42
|
Makaronidis JM, Batterham RL. The role of gut hormones in the pathogenesis and management of obesity. CURRENT OPINION IN PHYSIOLOGY 2019. [DOI: 10.1016/j.cophys.2019.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
43
|
Racz R, Nagy A, Rakonczay Z, Dunavari EK, Gerber G, Varga G. Defense Mechanisms Against Acid Exposure by Dental Enamel Formation, Saliva and Pancreatic Juice Production. Curr Pharm Des 2019; 24:2012-2022. [PMID: 29769002 PMCID: PMC6225347 DOI: 10.2174/1381612824666180515125654] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/30/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022]
Abstract
The pancreas, the salivary glands and the dental enamel producing ameloblasts have marked developmental, structural and functional similarities. One of the most striking similarities is their bicarbonate-rich secretory product, serving acid neutralization. An important difference between them is that while pancreatic juice and saliva are delivered into a lumen where they can be collected and analyzed, ameloblasts produce locally precipitating hydroxyapatite which cannot be easily studied. Interestingly, the ion and protein secretion by the pancreas, the salivary glands, and maturation ameloblasts are all two-step processes, of course with significant differences too. As they all have to defend against acid exposure by producing extremely large quantities of bicarbonate, the failure of this function leads to deteriorating consequences. The aim of the present review is to describe and characterize the defense mechanisms of the pancreas, the salivary glands and enamel-producing ameloblasts against acid exposure and to compare their functional capabilities to do this by producing bicarbonate.
Collapse
Affiliation(s)
- Robert Racz
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| | - Akos Nagy
- Department of Dentistry, Oral and Maxillofacial Surgery, University of Pecs, Pecs, Hungary
| | - Zoltan Rakonczay
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - Erika Katalin Dunavari
- Department of Dentistry, Oral and Maxillofacial Surgery, University of Pecs, Pecs, Hungary
| | - Gabor Gerber
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gabor Varga
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
44
|
Bonaz B, Sinniger V, Pellissier S. Vagus Nerve Stimulation at the Interface of Brain-Gut Interactions. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034199. [PMID: 30201788 DOI: 10.1101/cshperspect.a034199] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vagus nerve, a key component of the cross-communication between the gut and the brain, is a major element of homeostasis sensing the "milieu intérieur" and boosting the nervous and endocrine responses to maintain the gastrointestinal health status. This nerve has anti-inflammatory properties regulating the gut through the activation of the hypothalamic-pituitary-adrenal axis and the release of cortisol and through a vagovagal reflex, which has an anti-tumor necrosis factor (TNF) effect called the cholinergic anti-inflammatory pathway. Stimulating this nerve is an interesting tool as a nondrug therapy for the treatment of gastrointestinal diseases in which brain-gut communication is dysfunctional, such as inflammatory bowel disorders and others. This review presents the rationale of vagal gastrointestinal physiology and diseases and the most recent advances in vagus nerve stimulation. It also highlights the main issues to be addressed in the future to improve this bioelectronic therapy for gastrointestinal disorders.
Collapse
Affiliation(s)
- Bruno Bonaz
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Valérie Sinniger
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Sonia Pellissier
- University Grenoble Alpes, University Savoie Mont Blanc, 38000 Grenoble, France
| |
Collapse
|
45
|
Gelberg L, Rico MW, Herman DR, Belin TR, Chandler M, Ramirez E, Love S, McCarthy WJ. Comparative effectiveness trial comparing MyPlate to calorie counting for mostly low-income Latino primary care patients of a federally qualified community health center: study design, baseline characteristics. BMC Public Health 2019; 19:990. [PMID: 31340800 PMCID: PMC6651946 DOI: 10.1186/s12889-019-7294-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Primary care-based behavior change obesity treatment has long featured the Calorie restriction (CC), portion control approach. By contrast, the MyPlate-based obesity treatment approach encourages eating more high-satiety/high-satiation foods and requires no calorie-counting. This report describes study methods of a comparative effectiveness trial of CC versus MyPlate. It also describes baseline findings involving demographic characteristics and their associations with primary outcome measures and covariates, including satiety/satiation, dietary quality and acculturation. METHODS A comparative effectiveness trial was designed to compare the CC approach (n = 130) versus a MyPlate-based approach (n = 131) to treating patient overweight. Intervenors were trained community health workers. The 11 intervention sessions included two in-home health education sessions, two group education sessions, and seven telephone coaching sessions. Questionnaire and anthropometric assessments occurred at baseline, 6- and 12 months; food frequency questionnaires were administered at baseline and 12 months. Participants were overweight adult primary care patients of a federally qualified health center in Long Beach, California. Two measures of satiety/satiation and one measure of post-meal hunger comprised the primary outcome measures. Secondary outcomes included weight, waist circumference, blood pressure, dietary quality, sugary beverage intake, water intake, fruit and vegetable fiber intake, mental health and health-related quality of life. Covariates included age, gender, nativity status (U.S.-born, not U.S.-born), race/ethnicity, education, and acculturation. ANALYSIS Baseline characteristics were compared using chi square tests. Associations between covariates and outcome measures were evaluated using multiple regression and logistic regression. RESULTS Two thousand eighty-six adult patients were screened, yielding 261 enrollees who were 86% Latino, 8% African American, 4% White and 2% Other. Women predominated (95%). Mean age was 42 years. Most (82%) were foreign-born; 74% chose the Spanish language option. Mean BMI was 33.3 kg/m2; mean weight was 82 kg; mean waist circumference was 102 cm. Mean blood pressure was 122/77 mm. Study arms on key baseline measures did not differ except on dietary quality and sugary beverage intake. Nativity status was significantly associated with dietary quality. CONCLUSIONS The two treatment arms were well-balanced demographically at baseline. Nativity status is inversely related to dietary quality. TRIAL REGISTRATION NCT02514889 , posted on 8/4/2015.
Collapse
Affiliation(s)
- Lillian Gelberg
- Department of Family Medicine, David Geffen School of Medicine at UCLA, University of California, 10880 Wilshire Blvd, Ste 1800, Los Angeles, CA 90024 USA
| | - Melvin W. Rico
- Charles R. Drew/UCLA Medical Education Program, Los Angeles, USA
| | - Dena R. Herman
- Family and Consumer Sciences, College of Health and Human Development, SQ 200M 18111 Nordhoff Street, Northridge, CA 91330 USA
| | - Thomas R. Belin
- UCLA Fielding School of Public Health, 51-267 CHS, mc 177220, 650 Charles Young Drive, Los Angeles, CA 90095 USA
| | - Maria Chandler
- TCC, 701 East 28th Street, Suite 200, Long Beach, CA 90806 USA
| | - Evangelina Ramirez
- The Children’s Clinic of Long Beach Patient Representative, 1715 E. Anaheim St., Apt. 2, Long Beach, CA 90813 USA
| | | | - William J. McCarthy
- UCLA Fielding School of Public Health, A2-125 CHS, mc 690015, 650 Charles Young Drive, Los Angeles, CA 90095 USA
| |
Collapse
|
46
|
Roubalová R, Procházková P, Papežová H, Smitka K, Bilej M, Tlaskalová-Hogenová H. Anorexia nervosa: Gut microbiota-immune-brain interactions. Clin Nutr 2019; 39:676-684. [PMID: 30952533 DOI: 10.1016/j.clnu.2019.03.023] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/14/2022]
Abstract
Anorexia nervosa is a psychiatric disorder defined by an extremely low body weight, a devastating fear of weight gain, and body image disturbance, however the etiopathogenesis remains unclear. The objective of the article is to provide a comprehensive review on the potential role of gut microbiota in pathogenesis of anorexia nervosa. Recent advances in sequencing techniques used for microbial detection revealed that this disease is associated with disruption of the composition of normal gut microbiota (dysbiosis), manifested by low microbial diversity and taxonomic differences as compared to healthy individuals. Microorganisms present in the gut represent a part of the so called "microbiota-gut-brain" axis that affect the central nervous system and thus human behavior via the production of various neuroactive compounds. In addition, cells of the immune system are equipped with receptors for these neuroactive substances. Microbiota of the intestinal system also represent a very important antigenic source. These antigens can mimic some host neuropeptides and neurohormones and thus trigger the production of autoantibodies which cross-react with these compounds. The levels and affinities of these antibodies are thought to be associated with neuropsychiatric conditions including anxiety, depression, and eating and sleep disorders. The study of microbiota function in diseases could bring new insights to the pathogenetic mechanisms.
Collapse
Affiliation(s)
- Radka Roubalová
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Prague 4, Czech Republic.
| | - Petra Procházková
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Prague 4, Czech Republic
| | - Hana Papežová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, Prague 2, Czech Republic
| | - Kvido Smitka
- The Institute of Physiology, First Faculty of Medicine, Charles University, Albertov 5, Prague 2, Czech Republic
| | - Martin Bilej
- Institute of Microbiology, Czech Academy of Sciences, Videnska 1083, Prague 4, Czech Republic
| | | |
Collapse
|
47
|
Miron I, Dumitrascu DL. GASTROINTESTINAL MOTILITY DISORDERS IN OBESITY. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:497-504. [PMID: 32377248 DOI: 10.4183/aeb.2019.497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) motility, which is important for the digestion and absorption, may be altered in obesity. The aim of this review is to present the GI motility changes occurring in obesity, as well as their underlying mechanisms. We have conducted a systematic review of the published literature concerning GI motility and obesity and have described recent published data on the changes throughout the entire GI tract. Most recent discoveries include evidence supporting the increase of gastroesophageal reflux disease in obesity and inhibition of gastric motility. Intestinal transit of the distal small bowel generally slows down, ensuring enough time for digestion and absorption. Constipation is more frequent in obese patients than in those with a normal weight. The gut-brain axis plays an important role in the pathophysiology of GI motility disorders in obesity. This bidirectional communication is achieved by way of neurons, hormones, metabolites derived from intestinal microbiota and cytokines. The molecular mechanisms of GI motility changes in obesity are complex. Current data offer a starting point for further research needed to clarify the association of obesity with GI motility disorders.
Collapse
Affiliation(s)
- I Miron
- "Iuliu Hatieganu" University of Medicine and Pharmacy, 3 Medical Clinic, Cluj-Napoca, Romania
| | - D L Dumitrascu
- "Iuliu Hatieganu" Dept of Internal Medicine, Cluj-Napoca, Romania
| |
Collapse
|
48
|
Mantelmacher FD, Zvibel I, Cohen K, Epshtein A, Pasmanik-Chor M, Vogl T, Kuperman Y, Weiss S, Drucker DJ, Varol C, Fishman S. GIP regulates inflammation and body weight by restraining myeloid-cell-derived S100A8/A9. Nat Metab 2019; 1:58-69. [PMID: 32694806 DOI: 10.1038/s42255-018-0001-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/01/2018] [Indexed: 12/25/2022]
Abstract
Enteroendocrine cells relay energy-derived signals to immune cells to signal states of nutrient abundance and control immunometabolism. Emerging data suggest that the gut-derived nutrient-induced incretin glucose-dependent insulinotropic polypeptide (GIP) operates at the interface of metabolism and inflammation. Here we show that high-fat diet (HFD)-fed mice with immune cell-targeted GIP receptor (GIPR) deficiency exhibit greater weight gain, insulin resistance, hepatic steatosis and significant myelopoiesis concomitantly with impaired energy expenditure and inguinal white adipose tissue (WAT) beiging. Expression of the S100 calcium-binding protein S100A8 was increased in the WAT of mice with immune cell-targeted GIPR deficiency and co-deletion of GIPR and the heterodimer S100A8/A9 in immune cells ameliorated the aggravated metabolic and inflammatory phenotype following a HFD. Specific GIPR deletion in myeloid cells identified this lineage as the target of GIP effects. Furthermore, GIP directly downregulated S100A8 expression in adipose tissue macrophages. Collectively, our results identify a myeloid-GIPR-S100A8/A9 signalling axis coupling nutrient signals to the control of inflammation and adaptive thermogenesis.
Collapse
Affiliation(s)
- Fernanda Dana Mantelmacher
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Isabel Zvibel
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Cohen
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alona Epshtein
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Shai Weiss
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Daniel J Drucker
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sigal Fishman
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
49
|
Neuroendocrinology of Adipose Tissue and Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2018; 19:49-70. [PMID: 28933061 DOI: 10.1007/978-3-319-63260-5_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Food intake and energy expenditure are closely regulated by several mechanisms which involve peripheral organs and nervous system, in order to maintain energy homeostasis.Short-term and long-term signals express the size and composition of ingested nutrients and the amount of body fat, respectively. Ingested nutrients trigger mechanical forces and gastrointestinal peptide secretion which provide signals to the brain through neuronal and endocrine pathways. Pancreatic hormones also play a role in energy balance exerting a short-acting control regulating the start, end, and composition of a meal. In addition, insulin and leptin derived from adipose tissue are involved in long-acting adiposity signals and regulate body weigh as well as the amount of energy stored as fat over time.This chapter focuses on the gastrointestinal-, pancreatic-, and adipose tissue-derived signals which are integrated in selective orexigenic and anorexigenic brain areas that, in turn, regulate food intake, energy expenditure, and peripheral metabolism.
Collapse
|
50
|
Idrizaj E, Garella R, Castellini G, Mohr H, Pellegata NS, Francini F, Ricca V, Squecco R, Baccari MC. Adiponectin affects the mechanical responses in strips from the mouse gastric fundus. World J Gastroenterol 2018; 24:4028-4035. [PMID: 30254407 PMCID: PMC6148421 DOI: 10.3748/wjg.v24.i35.4028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/12/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether the adipocytes derived hormone adiponectin (ADPN) affects the mechanical responses in strips from the mouse gastric fundus.
METHODS For functional experiments, gastric strips from the fundal region were cut in the direction of the longitudinal muscle layer and placed in organ baths containing Krebs-Henseleit solution. Mechanical responses were recorded via force-displacement transducers, which were coupled to a polygraph for continuous recording of isometric tension. Electrical field stimulation (EFS) was applied via two platinum wire rings through which the preparation was threaded. The effects of ADPN were investigated on the neurally-induced contractile and relaxant responses elicited by EFS. The expression of ADPN receptors, Adipo-R1 and Adipo-R2, was also evaluated by touchdown-PCR analysis.
RESULTS In the functional experiments, EFS (4-16 Hz) elicited tetrodotoxin (TTX)-sensitive contractile responses. Addition of ADPN to the bath medium caused a reduction in amplitude of the neurally-induced contractile responses (P < 0.05). The effects of ADPN were no longer observed in the presence of the nitric oxide (NO) synthesis inhibitor L-NG-nitro arginine (L-NNA) (P > 0.05). The direct smooth muscle response to methacholine was not influenced by ADPN (P > 0.05). In carbachol precontracted strips and in the presence of guanethidine, EFS induced relaxant responses. Addition of ADPN to the bath medium, other than causing a slight and progressive decay of the basal tension, increased the amplitude of the neurally-induced relaxant responses (P < 0.05). Touchdown-PCR analysis revealed the expression of both Adipo-R1 and Adipo-R2 in the gastric fundus.
CONCLUSION The results indicate for the first time that ADPN is able to influence the mechanical responses in strips from the mouse gastric fundus.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence 50134, Italy
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence 50134, Italy
| | - Giovanni Castellini
- Psychiatry Unit, Department of Health Sciences, University of Florence, Florence 50134, Italy
| | - Hermine Mohr
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Natalia S Pellegata
- Institute for Diabetes and Cancer, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Fabio Francini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence 50134, Italy
| | - Valdo Ricca
- Psychiatry Unit, Department of Health Sciences, University of Florence, Florence 50134, Italy
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence 50134, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence 50134, Italy
| |
Collapse
|