151
|
Abstract
Leptin, as a major adipokine, positively correlates with the body's fat, while atopy is an important feature in the development of childhood asthma. We aimed to evaluate the relationship between leptin, parameters of obesity, and atopy in children with asthma. The study included 112 children (73 boys, 39 girls, mean age 11.1±2.4). 41 were overweight, 38 had asthma and a normal body mass index (BMI), and 33 were overweight asthmatics. Serum leptin levels, BMI, waist circumference (WC), and waist to hips ratio (WHR) were measured. Skin prick test (SPT)/CAP, total serum IgE, fractional exhaled nitric oxide (FeNO), and pulmonary function tests were performed. In asthmatic children, serum leptin median level was 9.2±16.2 ng/ml, in overweight children was 30.6±21.6 ng/ml, and in overweight asthmatics was 31.1±20.3 ng/ml with a significant difference between the groups (p=0.0374), yet with a significantly lower median level in the group of children with asthma compared to the overweight children: with asthma (p=0.00001) and without asthma (p=0.00001). In the three groups of patients, BMI and WC displayed a significant positive correlation with leptin (for BMI r=0.652 vs. r=0.530 vs. r=0.563, respectively and for WC r=0.508 vs. r=0.426 vs. r=0.527, respectively). No significant correlations of leptin within atopy parameters (Eo, IgE, SPT/CAP, FeNO) in all three analyzed groups (p>0.05) was detected. Conclusion: Atopy was not confirmed as an underlying mechanism of the association between asthma and being overweight. Leptin had a significant linear correlation as a parameter of central obesity with BMI and WC in all three groups, but not with WHR.
Collapse
|
152
|
O'Brien CJO, Haberman ER, Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol 2021; 37:549-573. [PMID: 34613819 PMCID: PMC7614880 DOI: 10.1146/annurev-cellbio-120319-114106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Emma R Haberman
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| |
Collapse
|
153
|
Spadini S, Ferro M, Lamanna J, Malgaroli A. Activity-based anorexia animal model: a review of the main neurobiological findings. J Eat Disord 2021; 9:123. [PMID: 34600568 PMCID: PMC8487535 DOI: 10.1186/s40337-021-00481-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The genesis of anorexia nervosa (AN), a severe eating disorder with a pervasive effect on many brain functions such as attention, emotions, reward processing, cognition and motor control, has not yet been understood. Since our current knowledge of the genetic aspects of AN is limited, we are left with a large and diversified number of biological, psychological and environmental risk factors, called into question as potential triggers of this chronic condition with a high relapse rate. One of the most valid and used animal models for AN is the activity-based anorexia (ABA), which recapitulates important features of the human condition. This model is generated from naïve rodents by a self-motivated caloric restriction, where a fixed schedule food delivery induces spontaneous increased physical activity. AIM In this review, we sought to provide a summary of the experimental research conducted using the ABA model in the pursuit of potential neurobiological mechanism(s) underlying AN. METHOD The experimental work presented here includes evidence for neuroanatomical and neurophysiological changes in several brain regions as well as for the dysregulation of specific neurochemical synaptic and neurohormonal pathways. RESULTS The most likely hypothesis for the mechanism behind the development of the ABA phenotype relates to an imbalance of the neural circuitry that mediates reward processing. Evidence collected here suggests that ABA animals show a large set of alterations, involving regions whose functions extend way beyond the control of reward mechanisms and eating habits. Hence, we cannot exclude a primary role of these alterations from a mechanistic theory of ABA induction. CONCLUSIONS These findings are not sufficient to solve such a major enigma in neuroscience, still they could be used to design ad hoc further experimental investigation. The prospect is that, since treatment of AN is still challenging, the ABA model could be more effectively used to shed light on the complex AN neurobiological framework, thus supporting the future development of therapeutic strategies but also the identification of biomarkers and diagnostic tools. Anorexia Nervosa (AN) is a severe eating disorder with a dramatic effect on many functions of our brain, such as attention, emotions, cognition and motion control. Since our current knowledge of the genetic aspects behind the development of AN is still limited, many biological, psychological and environmental factors must be taken into account as potential triggers of this condition. One of the most valid animal models for studying AN is the activity-based anorexia (ABA). In this model, rodents spontaneously limit food intake and start performing increased physical activity on a running wheel, a result of the imposition of a fixed time schedule for food delivery. In this review, we provide a detailed summary of the experimental research conducted using the ABA model, which includes extended evidence for changes in the anatomy and function of the brain of ABA rodents. The hope is that such integrated view will support the design of future experiments that will shed light on the complex brain mechanisms behind AN. Such advanced knowledge is crucial to find new, effective strategies for both the early diagnosis of AN and for its treatment.
Collapse
Affiliation(s)
- Sara Spadini
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Via Olgettina 58, 20132, Milan, Italy.
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
154
|
He Z, Lieu L, Dong Y, Afrin S, Chau D, Kabahizi A, Wallace B, Cao J, Hwang ES, Yao T, Huang Y, Okolo J, Cheng B, Gao Y, Hu L, Williams KW. PERK in POMC neurons connects celastrol with metabolism. JCI Insight 2021; 6:145306. [PMID: 34549728 PMCID: PMC8492333 DOI: 10.1172/jci.insight.145306] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 07/28/2021] [Indexed: 01/02/2023] Open
Abstract
ER stress and activation of the unfolded protein response in the periphery as well as the central nervous system have been linked to various metabolic abnormalities. Chemically lowering protein kinase R–like ER kinase (PERK) activity within the hypothalamus leads to decreased food intake and body weight. However, the cell populations required in this response remain undefined. In the current study, we investigated the effects of proopiomelanocortin-specific (POMC-specific) PERK deficiency on energy balance and glucose metabolism. Male mice deficient for PERK in POMC neurons exhibited improvements in energy balance on a high-fat diet, showing decreased food intake and body weight, independent of changes in glucose and insulin tolerances. The plant-based inhibitor of PERK, celastrol, increases leptin sensitivity, resulting in decreased food intake and body weight in a murine model of diet-induced obesity (DIO). Our data extend these observations by demonstrating that celastrol-induced improvements in leptin sensitivity and energy balance were attenuated in mice with PERK deficiency in POMC neurons. Altogether, these data suggest that POMC-specific PERK deficiency in male mice confers protection against DIO, possibly providing a new therapeutic target for the treatment of diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- Zhenyan He
- Department of Neurosurgery, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Linh Lieu
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Yanbin Dong
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA.,Institute of Gastroenterology and.,Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Sadia Afrin
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Dominic Chau
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Anita Kabahizi
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Briana Wallace
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Jianhong Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Eun-Sang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Ting Yao
- Division of Pediatric Endocrinology, Department of Pediatrics, UCLA Children's Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yiru Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jennifer Okolo
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Bo Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yong Gao
- Laboratory Department, Affiliated Hospital of Binzhou Medical College, Shandong, China
| | - Ling Hu
- Institute of Gastroenterology and.,Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
155
|
Sanborn V, Preis SR, Ang A, Devine S, Mez J, DeCarli C, Au R, Alosco ML, Gunstad J. Association Between Leptin, Cognition, and Structural Brain Measures Among "Early" Middle-Aged Adults: Results from the Framingham Heart Study Third Generation Cohort. J Alzheimers Dis 2021; 77:1279-1289. [PMID: 32831199 DOI: 10.3233/jad-191247] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND There is growing interest in the pathophysiological processes of preclinical Alzheimer's disease (AD), including the potential role of leptin. Human studies have shown that both low and high levels of leptin can be associated with worse neurocognitive outcomes, suggesting this relationship may be moderated by another risk factor. OBJECTIVE We examined the association between plasma leptin levels and both neuropsychological test performance and structural neuroimaging and assessed whether body mass index (BMI) is an effect modifier of these associations. METHODS Our study sample consisted of 2,223 adults from the Framingham Heart Study Third Generation Cohort (average age = 40 years, 53% women). RESULTS Among the entire sample, there was no association between leptin and any of the neuropsychological domain measures or any of the MRI brain volume measures, after adjustment for BMI, APOE4, and other clinical factors. However, we did observe that BMI category was an effect modifier for the association between leptin and verbal memory (p for interaction = 0.03), where higher levels of leptin were associated with better performance among normal weight participants (BMI 18.5-24.9) kg/m2 (beta = 0.12, p = 0.02). No association was observed between leptin level and verbal memory test performance among participants who were overweight or obese. CONCLUSION These findings suggest that the association between leptin and cognitive function is moderated by BMI category. Prospective examination of individuals transitioning from middle age to older adulthood will help to clarify the contribution of leptin to AD and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Victoria Sanborn
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| | - Sarah R Preis
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alvin Ang
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Sherral Devine
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA
| | - Charles DeCarli
- Department of Neurology, University of California at Davis Health System, Sacramento, CA, USA
| | - Rhoda Au
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Boston University Alzheimer's Disease Center and Boston University CTE Center, Boston University School of Medicine, Boston, MA, USA
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
156
|
Yu F, Wang Z, Zhang T, Chen X, Xu H, Wang F, Guo L, Chen M, Liu K, Wu B. Deficiency of intestinal Bmal1 prevents obesity induced by high-fat feeding. Nat Commun 2021; 12:5323. [PMID: 34493722 PMCID: PMC8423749 DOI: 10.1038/s41467-021-25674-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 08/19/2021] [Indexed: 12/18/2022] Open
Abstract
The role of intestine clock in energy homeostasis remains elusive. Here we show that mice with Bmal1 specifically deleted in the intestine (Bmal1iKO mice) have a normal phenotype on a chow diet. However, on a high-fat diet (HFD), Bmal1iKO mice are protected against development of obesity and related abnormalities such as hyperlipidemia and fatty livers. These metabolic phenotypes are attributed to impaired lipid resynthesis in the intestine and reduced fat secretion. Consistently, wild-type mice fed a HFD during nighttime (with a lower BMAL1 expression) show alleviated obesity compared to mice fed ad libitum. Mechanistic studies uncover that BMAL1 transactivates the Dgat2 gene (encoding the triacylglycerol synthesis enzyme DGAT2) via direct binding to an E-box in the promoter, thereby promoting dietary fat absorption. Supporting these findings, intestinal deficiency of Rev-erbα, a known BMAL1 repressor, enhances dietary fat absorption and exacerbates HFD-induced obesity and comorbidities. Moreover, small-molecule targeting of REV-ERBα/BMAL1 by SR9009 ameliorates HFD-induced obesity in mice. Altogether, intestine clock functions as an accelerator in dietary fat absorption and targeting intestinal BMAL1 may be a promising approach for management of metabolic diseases induced by excess fat intake.
Collapse
MESH Headings
- ARNTL Transcription Factors/deficiency
- ARNTL Transcription Factors/genetics
- Animals
- Circadian Rhythm/genetics
- Diacylglycerol O-Acyltransferase/genetics
- Diacylglycerol O-Acyltransferase/metabolism
- Diet, High-Fat/adverse effects
- Dietary Fats/administration & dosage
- Dietary Fats/metabolism
- Fatty Liver/etiology
- Fatty Liver/genetics
- Fatty Liver/metabolism
- Fatty Liver/prevention & control
- Gene Expression Regulation
- Homeostasis/drug effects
- Homeostasis/genetics
- Hyperlipidemias/etiology
- Hyperlipidemias/genetics
- Hyperlipidemias/metabolism
- Hyperlipidemias/prevention & control
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Lipid Metabolism/drug effects
- Lipid Metabolism/genetics
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Obesity/etiology
- Obesity/genetics
- Obesity/metabolism
- Obesity/prevention & control
- Promoter Regions, Genetic
- Protein Binding
- Pyrrolidines/pharmacology
- Signal Transduction
- Thiophenes/pharmacology
- Triglycerides/biosynthesis
Collapse
Affiliation(s)
- Fangjun Yu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhigang Wang
- Department of Intensive Care Unit, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tianpeng Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xun Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Haiman Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Fei Wang
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Lianxia Guo
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Min Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Kaisheng Liu
- Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China.
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
157
|
Del Vecchio G, Murashita K, Verri T, Gomes AS, Rønnestad I. Leptin receptor-deficient (knockout) zebrafish: Effects on nutrient acquisition. Gen Comp Endocrinol 2021; 310:113832. [PMID: 34089707 DOI: 10.1016/j.ygcen.2021.113832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/22/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022]
Abstract
In mammals, knockout of LEPR results in a hyperphagic, morbid obese, and diabetic phenotype, which supports that leptin plays an important role in the control of appetite and energy metabolism, and that its receptor, LEPR, mediates these effects. To date, little is known about the role(s) of lepr in teleost physiology. We investigated a zebrafish (Danio rerio) homozygous lepr knockout (lepr-/-) line generated by CRISPR/Cas9 in comparison to its wt counterpart with respect to nutrient acquisition, energy allocation, and metabolism. The metabolic characterization included oxygen consumption rate and morphometric parameters (yolk sac area, standard length, wet weight, and condition factor) as proxies for use and allocation of energy in developing (embryos, larvae, and juveniles) zebrafish and showed no particular differences between the two lines, in agreement with previous studies. One exception was found in oxygen consumption at 72 hpf, when zebrafish switch from embryonic to early larval stages and food-seeking behavior could be observed. In this case, the metabolic rate was significantly lower in lepr-/- than in wt. Both phenotypes showed similar responses, with respect to metabolic rate, to acute alterations (22 and 34 °C) in water temperature (measured in terms of Q10 and activation energy) compared to the standard (28 °C) rearing conditions. To assess lepr involvement in signaling the processing and handling of incoming nutrients when an exogenous meal is digested and absorbed, we conducted an in vivo analysis in lepr-/- and wt early (8 days post-fertilization) zebrafish larvae. The larvae were administered a bolus of protein hydrolysate (0%, 1%, 5%, and 15% lactalbumin) directly into the digestive tract lumen, and changes in the mRNA expression profile before and after (1 and 3 h) administration were quantified. The analysis showed transcriptional differences in the expressions of genes involved in the control of appetite and energy metabolism (cart, npy, agrp, and mc4r), sensing (casr, t1r1, t1r3, t1r2-1, t1r2-2, pept1a, and pept1b), and digestion (cck, pyy, try, ct, and amy), with more pronounced effects observed in the orexigenic than in the anorexigenic pathways, suggesting a role of lepr in their regulations. Differences in the mRNA levels of these genes in lepr-/-vs. wt larvae were also observed. Altogether, our analyses suggest an influence of lepr on physiological processes involved in nutrient acquisition, mainly control of food intake and digestion, during early development, whereas metabolism, energy allocation, and growth seem to be only slightly influenced.
Collapse
Affiliation(s)
- Gianmarco Del Vecchio
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy; Department of Biological Sciences, University of Bergen, PO Box 7803, NO-5020 Bergen, Norway
| | - Koji Murashita
- Department of Biological Sciences, University of Bergen, PO Box 7803, NO-5020 Bergen, Norway; Aquaculture Research Department, Fisheries Technology Institute, Fisheries Research and Education Agency, 224-1 Hiruda, Tamaki, Watarai, Mie 519-0423, Japan
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy
| | - Ana S Gomes
- Department of Biological Sciences, University of Bergen, PO Box 7803, NO-5020 Bergen, Norway
| | - Ivar Rønnestad
- Department of Biological Sciences, University of Bergen, PO Box 7803, NO-5020 Bergen, Norway.
| |
Collapse
|
158
|
Guo Z, Zhang Y, Liu C, Youn JY, Cai H. Toll-Like Receptor 2 (TLR2) Knockout Abrogates Diabetic and Obese Phenotypes While Restoring Endothelial Function via Inhibition of NOX1. Diabetes 2021; 70:2107-2119. [PMID: 34127487 PMCID: PMC8576422 DOI: 10.2337/db20-0591] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/08/2021] [Indexed: 11/13/2022]
Abstract
We have previously demonstrated a novel role of bone morphogenic protein 4 (BMP4) in inducing NOX1-dependent endothelial nitric oxide synthase (eNOS) uncoupling, endothelial dysfunction, and inflammatory activation in type 2 diabetes mellitus (T2DM). However, how BMP4 activates NOX1 and whether targeting the new mechanistic pathway revealed is effective in preserving endothelial function in T2DM remains unclear. In this study, we observed that BMP4 induced a marked, time-dependent increase in physiological binding between TLR2 and NOX1 in aortic endothelial cells as well as increased binding of TLR2 to NOXO1. In TLR2 knockout (Tlr2 -/-) mice fed high-fat diet, body weight gain was significantly less compared with wild-type (WT) mice both in males and females. The high-fat diet-induced increases in fasting blood glucose levels, as well as in circulating insulin and leptin levels, were absent in Tlr2 -/- mice. High-fat feeding induced increases in overall fat mass, and in fat mass of different pockets were abrogated in Tlr2 -/- mice. Whereas energy intake was similar in high-fat-fed WT and Tlr2 -/- mice, TLR2 deficiency resulted in higher energy expenditure attributable to improved physical activity, which was accompanied by restored skeletal muscle mitochondrial function. In addition, TLR2 deficiency recoupled eNOS, reduced total superoxide production, improved H4B and NO bioavailabilities in aortas, and restored endothelium-dependent vasorelaxation. Collectively, our data strongly indicate that TLR2 plays important roles in the development of metabolic features of T2DM and its related endothelial/vascular dysfunction. Therefore, targeting TLR2 may represent a novel therapeutic strategy for T2DM, obesity, and cardiovascular complications via specific inhibition of NOX1.
Collapse
Affiliation(s)
- Zhen Guo
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Chang Liu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
159
|
Berger C, Heyne HO, Heiland T, Dommel S, Höfling C, Guiu-Jurado E, Roßner S, Dannemann M, Kelso J, Kovacs P, Blüher M, Klöting N. A novel compound heterozygous leptin receptor mutation causes more severe obesity than in Lepr db/db mice. J Lipid Res 2021; 62:100105. [PMID: 34390703 PMCID: PMC8450258 DOI: 10.1016/j.jlr.2021.100105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/04/2023] Open
Abstract
The leptin receptor (Lepr) pathway is important for food intake regulation, energy expenditure, and body weight. Mutations in leptin and the Lepr have been shown to cause early-onset severe obesity in mice and humans. In studies with C57BL/6NCrl mice, we found a mouse with extreme obesity. To identify a putative spontaneous new form of monogenic obesity, we performed backcross studies with this mouse followed by a quantitative trait locus (QTL) analysis and sequencing of the selected chromosomal QTL region. We thereby identified a novel Lepr mutation (C57BL/6N-LeprL536Hfs*6-1NKB), which is located at chromosome 4, exon 11 within the CRH2-leptin-binding site. Compared with C57BL/6N mice, LeprL536Hfs*6 develop early onset obesity and their body weight exceeds that of Leprdb/db mice at an age of 30 weeks. Similar to Leprdb/db mice, the LeprL536Hfs*6 model is characterized by hyperphagia, obesity, lower energy expenditure and activity, hyperglycemia, and hyperinsulinemia compared with C57BL/6N mice. Crossing Leprdb/wt with LeprL536Hfs*6/wt mice results in compound heterozygous LeprL536Hfs*6/db mice, which develop even higher body weight and fat mass than both homozygous Leprdb/db and LeprL536Hfs*6 mice. Compound heterozygous Lepr deficiency affecting functionally different regions of the Lepr causes more severe obesity than the parental homozygous mutations.
Collapse
Affiliation(s)
- Claudia Berger
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany
| | - Henrike O Heyne
- Medical Department, Institute for Human Genetics, University of Leipzig Medical Center, Leipzig, Germany; Institute for Molecular Medicine Finland: FIMM, Helsinki, Finland; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tina Heiland
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Sebastian Dommel
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany
| | - Corinna Höfling
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Esther Guiu-Jurado
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Leipzig, Germany
| | - Steffen Roßner
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Michael Dannemann
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany; Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Janet Kelso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Medical Department III, Endocrinology, Nephrology, Rheumatology, CRC1052, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Leipzig, Germany.
| |
Collapse
|
160
|
Peñaloza-Sancho V, Pérez-Valenzuela C, Gonzalez C, Jujihara G, Bustos P, Dagnino-Subiabre A. Cannabinoid receptor type 1 modulates the effects of polyunsaturated fatty acids on memory of stressed rats. Nutr Neurosci 2021; 24:583-600. [PMID: 31637966 DOI: 10.1080/1028415x.2019.1659561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Memory and GABAergic activity in the hippocampus of stressed rats improve after n-3 polyunsaturated fatty acid (PUFA) supplementation. On the other hand, cannabinoid receptor type 1 (CB1) strongly regulates inhibitory neurotransmission in the hippocampus. Speculation about a possible relation between stress, endocannabinoids, and PUFAs. Here, we examined whether the effects of PUFAs on memory of chronically stressed rats depends on pharmacological manipulation of CB1 receptors. Male Sprague-Dawley rats were orally supplemented with n-3 (fish oil) or n-6 (primrose oil) PUFAs during chronic restraint stress (CRS) protocol (6 h/day; 21 days). First, we studied if the expression of CB1 receptors in the hippocampus may be affected by CRS and PUFAs supplementation by real-time PCR and immunofluorescence. CRS up-regulated the CB1 expression compared with the non-stressed rats, while only n-3 PUFAs countered this effect. Memory was evaluated in the Morris water maze. Stressed rats were co-treated with PUFAs and/or modulators of CB1 receptor (AM251, antagonist, 0.3 mg/kg/day; WIN55,212-2, agonist, 0.5 mg/kg/day) by intraperitoneal injections. Memory improved in the stressed rats that were treated with AM251 and/or n-3 PUFAs. Supplementation with n-6 PUFAs did not affect memory of stressed rats, but co-treatment with AM251 improved it, while co-treatment with WIN55,212-2 did not affect memory. Our results demonstrate that activity of the CB1 receptors may modulate the effects of PUFAs on memory of stressed rats. This study suggests that endocannabinoids and PUFAs can both become a singular system by being self-regulated in limbic areas, so they control the effects of stress on the brain.
Collapse
Affiliation(s)
- Valentín Peñaloza-Sancho
- Laboratory of Stress Neurobiology, Institute of Physiology, Center for Neurobiology and Integrative Pathophysiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - Catherine Pérez-Valenzuela
- Laboratory of Stress Neurobiology, Institute of Physiology, Center for Neurobiology and Integrative Pathophysiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - Celindo Gonzalez
- Laboratory of Stress Neurobiology, Institute of Physiology, Center for Neurobiology and Integrative Pathophysiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - German Jujihara
- Laboratory of Stress Neurobiology, Institute of Physiology, Center for Neurobiology and Integrative Pathophysiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - Paulina Bustos
- PhD Program in Aquaculture, School of Marine Sciences, Pontifica Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Institute of Physiology, Center for Neurobiology and Integrative Pathophysiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
161
|
Bake T, Peris-Sampedro F, Wáczek Z, Ohlsson C, Pálsdóttir V, Jansson JO, Dickson SL. The gravitostat protects diet-induced obese rats against fat accumulation and weight gain. J Neuroendocrinol 2021; 33:e12997. [PMID: 34240761 DOI: 10.1111/jne.12997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/23/2021] [Accepted: 05/31/2021] [Indexed: 11/29/2022]
Abstract
The gravitostat is a novel homeostatic body weight-regulating mechanism, mostly studied in mice, and recently confirmed in obese humans. In the present study, we explored the effect of weight loading on metabolic outcomes, meal patterns and parameters linked to energy expenditure in both obese and lean rats. Diet-induced obese (DIO) and lean rats were implanted with capsules weighing either 15% of biological body weight (load) or empty capsules (1.3% of body weight; controls). Loading protected against fat accumulation more markedly in the DIO group. In line with this, the obesity-related impairment in insulin sensitivity was notably ameliorated in DIO rats upon loading, as revealed by the reduction in serum insulin levels and homeostatic model assessment for insulin resistance index scores. Although 24-hour caloric intake was reduced in both groups, this effect was greater in loaded DIO rats than in loaded lean peers. During days 10-16, after recovery from surgery, loading: (i) decreased meal size in both groups (only during the light phase in DIO rats) but this was compensated in lean rats by an increase in meal frequency; (ii) reduced dark phase locomotor activity only in lean rats; and (iii) reduced mean caloric efficiency in DIO rats. Muscle weight was unaffected by loading in either group. Dietary-obese rats are therefore more responsive than lean rats to loading.
Collapse
Affiliation(s)
- Tina Bake
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Fiona Peris-Sampedro
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Zita Wáczek
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre of Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Vilborg Pálsdóttir
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - John-Olov Jansson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Department of Physiology/Endocrine, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
162
|
Rostami Rayeni N, Abdollahzad H, Alibakhshi P, Morvaridzadeh M, Heydari H, Dehnad A, Khorshidi M, Izadi A, Shidfar F, Dulce Estêvão M, Omidi A, Heshmati J. Effects of body weight regain on leptin levels: A systematic review and meta-analysis. Cytokine 2021; 148:155647. [PMID: 34344588 DOI: 10.1016/j.cyto.2021.155647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND There are different changes observed before and after diet therapy, and also after weight regain. However, there is not sufficient information regarding weight regain and hormonal changes. PURPOSE The purpose of this study was to review the connection between weight regain and leptin concentration levels. METHODS MEDLINE, SCOPUS, Web of Science, and the Cochrane Library were searched for interventional articles published from January 1, 1980, to June 30, 2020. Randomized clinical trials with parallel or cross over design assessing leptin concentrations at the baseline and at the end of study were reviewed. Two independent reviewers extracted data related to study design, year of publication, country, age, gender, body mass index (BMI), duration of the following up period and mean ± SD of other intended variables. RESULTS Four articles were included, published between 2004 and 2016. Three of them were conducted in the US and one of them in Netherland. Sample size of the studies ranged between 25 and 148 participants. The range of following up period was from13 to 48 weeks. The age range of participants was from 34 to 44 years. Our analysis shows that weight regain could reduce leptin levels, but this change is not statistically significant. CONCLUSION This review suggests that weight regain may induce a non-significant reduction in leptin level. However, the limited number and great heterogeneity between the included studies may affect the presented results and there are still need to well-designed, large population studies to determine the relationship between weight regain and leptin levels.
Collapse
Affiliation(s)
- Najme Rostami Rayeni
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Hadi Abdollahzad
- Department of Nutritional Science, School of Nutritional Science and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pooya Alibakhshi
- Department of Internal Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojgan Morvaridzadeh
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hafez Heydari
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Afsaneh Dehnad
- Department of English Language, School of Health Management and Information Sciences, Center for Educational Research in Medical Sciences (CERMS), Iran University of Medical Sciences, Iran.
| | - Masoud Khorshidi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azimeh Izadi
- Iran Football Medical Assessment and Rehabilitation Center (IFMARC), Tehran, Iran; Department of Biochemistry and Diet Therapy, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - M Dulce Estêvão
- Universidade do Algarve, Escola Superior de Saúde, Campus de Gambelas, Faro, Portugal.
| | - Amirhosein Omidi
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javad Heshmati
- Songhor Healthcare Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
163
|
Abstract
Gut microbiota has emerged as a major metabolically active organ with critical functions in both health and disease. The trillions of microorganisms hosted by the gastrointestinal tract are involved in numerous physiological and metabolic processes including modulation of appetite and regulation of energy in the host spanning from periphery to the brain. Indeed, bacteria and their metabolic byproducts are working in concert with the host chemosensory signaling pathways to affect both short- and long-term ingestive behavior. Sensing of nutrients and taste by specialized G protein-coupled receptor cells is important in transmitting food-related signals, optimizing nutrition as well as in prevention and treatment of several diseases, notably obesity, diabetes and associated metabolic disorders. Further, bacteria metabolites interact with specialized receptors cells expressed by gut epithelium leading to taste and appetite response changes to nutrients. This review describes recent advances on the role of gut bacteria in taste perception and functions. It further discusses how intestinal dysbiosis characteristic of several pathological conditions may alter and modulate taste preference and food consumption via changes in taste receptor expression.
Collapse
|
164
|
Lkhagvasuren B, Mee-Inta O, Zhao ZW, Hiramoto T, Boldbaatar D, Kuo YM. Pancreas-Brain Crosstalk. Front Neuroanat 2021; 15:691777. [PMID: 34354571 PMCID: PMC8329585 DOI: 10.3389/fnana.2021.691777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
The neural regulation of glucose homeostasis in normal and challenged conditions involves the modulation of pancreatic islet-cell function. Compromising the pancreas innervation causes islet autoimmunity in type 1 diabetes and islet cell dysfunction in type 2 diabetes. However, despite the richly innervated nature of the pancreas, islet innervation remains ill-defined. Here, we review the neuroanatomical and humoral basis of the cross-talk between the endocrine pancreas and autonomic and sensory neurons. Identifying the neurocircuitry and neurochemistry of the neuro-insular network would provide clues to neuromodulation-based approaches for the prevention and treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Battuvshin Lkhagvasuren
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Onanong Mee-Inta
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Zi-Wei Zhao
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Tetsuya Hiramoto
- Department of Psychosomatic Medicine, Fukuoka Hospital, National Hospital Organization, Fukuoka, Japan
| | - Damdindorj Boldbaatar
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan.,Department of Cell Biology and Anatomy, National Cheng Kung University College of Medicine, Tainan, Taiwan
| |
Collapse
|
165
|
Mahesh M, Pandey H, Raja Gopal Reddy M, Prabhakaran Sobhana P, Korrapati D, Uday Kumar P, Vajreswari A, Jeyakumar SM. Carrot Juice Consumption Reduces High Fructose-Induced Adiposity in Rats and Body Weight and BMI in Type 2 Diabetic Subjects. Nutr Metab Insights 2021; 14:11786388211014917. [PMID: 34349520 PMCID: PMC8287410 DOI: 10.1177/11786388211014917] [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: 11/18/2020] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
Nutritional intervention is a key strategy in the control and management of non-communicable diseases. Here, initially, we evaluated the effects of carrot juice (CJ) on some of the physical and biochemical parameters in rats fed with high-fructose diet, then in type 2 diabetic subjects. For the animal study, weanling male Wistar rats were given control (n = 6) or high fructose (HFr; n = 24) diet for 8 weeks. Then, the HFr group rats were subdivided into 4 groups (n = 6 in each) and continued either on HFr diet or shifted to control diet, with or without CJ (0.3 mg β-carotene) ingestion orally for 8 weeks. At the end, the ingestion of CJ reversed the HFr-induced adiposity (23 ± 1.6 vs 18 ± 1.1, P = .038), hypertriglyceridemia (182 ± 18.2 vs 90 ± 10.5 mg/dL, P<0.001), and hyperinsulinemia (81 ± 14.7 vs 40 ± 7.5 µU/mL, P = .014), while increased the retinol levels in liver (240 ± 38.4 vs 492 ± 61.2 µg/g, P = .002) and adipose tissue (1.8 ± 0.09 vs 2.5 ± 0.18 µg/g, P = .026). On the other hand, in the diabetic subjects (7 males and females each, n = 14) compared to their baseline, the daily consumption of 50 mL CJ (~2400 µg β-carotene) for 6 weeks significantly reduced the body weight (69.4 ± 4.13 vs 69.0 ± 4.09 kg, P = .014), BMI (27.4 ± 1.07 vs 27.2 ± 1.06 kg/m2, P = .007), and fat% (33.4 ± 1.87 vs 31.9 ± 2.13, P = .029) with an increase in plasma β-carotene levels (0.21 ± 0.045 vs 0.45 ± 0.089 µmol/L, P = .044). Although CJ increased the glucose (145 ± 10.4 vs 165 ± 11.4 mg/dL, P = .039), insulin, and glycated hemoglobin levels remained unaltered. In conclusion, the consumption of carrot juice reversed the HFr-induced metabolic abnormalities in a rat model and decreased body weight and BMI of diabetic subjects.
Collapse
Affiliation(s)
- Malleswarapu Mahesh
- Division of Lipid Biochemistry, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Himanshi Pandey
- Division of Dietetics, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Mooli Raja Gopal Reddy
- Division of Lipid Biochemistry, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | | | - Damayanti Korrapati
- Division of Dietetics, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | - Putcha Uday Kumar
- Division of Pathology, ICMR-National Institute of Nutrition, Hyderabad, Telangana, India
| | | | | |
Collapse
|
166
|
Kim LJ, Shin MK, Pho H, Otvos L, Tufik S, Andersen ML, Pham LV, Polotsky VY. Leptin Receptor Blockade Attenuates Hypertension, but Does Not Affect Ventilatory Response to Hypoxia in a Model of Polygenic Obesity. Front Physiol 2021; 12:688375. [PMID: 34276408 PMCID: PMC8283021 DOI: 10.3389/fphys.2021.688375] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022] Open
Abstract
Background Obesity can cause hypertension and exacerbates sleep-disordered breathing (SDB). Leptin is an adipocyte-produced hormone, which increases metabolic rate, suppresses appetite, modulates control of breathing, and increases blood pressure. Obese individuals with high circulating levels of leptin are resistant to metabolic and respiratory effects of leptin, but they appear to be sensitive to hypertensive effects of this hormone. Obesity-induced hypertension has been associated with hyperleptinemia. New Zealand obese (NZO) mice, a model of polygenic obesity, have high levels of circulating leptin and hypertension, and are prone to develop SDB, similarly to human obesity. We hypothesize that systemic leptin receptor blocker Allo-aca will treat hypertension in NZO mice without any effect on body weight, food intake, or breathing. Methods Male NZO mice, 12–13 weeks of age, were treated with Allo-aca (n = 6) or a control peptide Gly11 (n = 12) for 8 consecutive days. Doses of 0.2 mg/kg were administered subcutaneously 2×/day, at 10 AM and 6 PM. Blood pressure was measured by telemetry for 48 h before and during peptide infusion. Ventilation was assessed by whole-body barometric plethysmography, control of breathing was examined by assessing the hypoxic ventilatory response (HVR), and polysomnography was performed during light-phase at baseline and during treatment. Heart rate variability analyses were performed to estimate the cardiac autonomic balance. Results Systemic leptin receptor blockade with Allo-aca did not affect body weight, body temperature, and food intake in NZO mice. Plasma levels of leptin did not change after the treatment with either Allo-aca or the control peptide Gy11. NZO mice were hypertensive at baseline and leptin receptor blocker Allo-aca significantly reduced the mean arterial pressure from 134.9 ± 3.1 to 124.9 ± 5.7 mmHg during the light phase (P < 0.05), whereas the control peptide had no effect. Leptin receptor blockade did not change the heart rate or cardiac autonomic balance. Allo-aca did not affect minute ventilation under normoxic or hypoxic conditions and HVR. Ventilation, apnea index, and oxygen desaturation during NREM and REM sleep did not change with leptin receptor blockade. Conclusion Systemic leptin receptor blockade attenuates hypertension in NZO mice, but does not exacerbate obesity and SDB. Thus, leptin receptor blockade represents a potential pharmacotherapy for obesity-associated hypertension.
Collapse
Affiliation(s)
- Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laszlo Otvos
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary.,Arrevus, Inc., Raleigh, NC, United States.,OLPE, LLC, Audubon, PA, United States
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
167
|
van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev 2021; 22:e13210. [PMID: 33559362 PMCID: PMC8243944 DOI: 10.1111/obr.13210] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
The role of serotonin in food intake has been studied for decades. Food intake is mainly regulated by two brain circuitries: (i) the homeostatic circuitry, which matches energy intake to energy expenditure, and (ii) the hedonic circuitry, which is involved in rewarding and motivational aspects of energy consumption. In the homeostatic circuitry, serotonergic signaling contributes to the integration of metabolic signals that convey the body's energy status and facilitates the ability to suppress food intake when homeostatic needs have been met. In the hedonic circuitry, serotonergic signaling may reduce reward-related, motivational food consumption. In contrast, peripherally acting serotonin promotes energy absorption and storage. Disturbed serotonergic signaling is associated with obesity, emphasizing the importance to understand the role of serotonergic signaling in food intake. However, unraveling the serotonin-mediated regulation of food intake is complex, as the effects of serotonergic signaling in different brain regions depend on the regional expression of serotonin receptor subtypes and downstream effects via connections to other brain regions. We therefore provide an overview of the effects of serotonergic signaling in brain regions of the homeostatic and hedonic regulatory systems on food intake. Furthermore, we discuss the disturbances in serotonergic signaling in obesity and its potential therapeutic implications.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
168
|
Shin MK, Mitrut R, Gu C, Kim LJ, Yeung BH, Lee R, Pham L, Tang WY, Sham JSK, Cui H, Polotsky VY. Pharmacological and Genetic Blockade of Trpm7 in the Carotid Body Treats Obesity-Induced Hypertension. Hypertension 2021; 78:104-114. [PMID: 33993722 PMCID: PMC8192446 DOI: 10.1161/hypertensionaha.120.16527] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roxana Mitrut
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bonnie H.Y. Yeung
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rachel Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wan-Yee Tang
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - James S. K. Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
169
|
Correa‐da‐Silva F, Fliers E, Swaab DF, Yi C. Hypothalamic neuropeptides and neurocircuitries in Prader Willi syndrome. J Neuroendocrinol 2021; 33:e12994. [PMID: 34156126 PMCID: PMC8365683 DOI: 10.1111/jne.12994] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Prader-Willi Syndrome (PWS) is a rare and incurable congenital neurodevelopmental disorder, resulting from the absence of expression of a group of genes on the paternally acquired chromosome 15q11-q13. Phenotypical characteristics of PWS include infantile hypotonia, short stature, incomplete pubertal development, hyperphagia and morbid obesity. Hypothalamic dysfunction in controlling body weight and food intake is a hallmark of PWS. Neuroimaging studies have demonstrated that PWS subjects have abnormal neurocircuitry engaged in the hedonic and physiological control of feeding behavior. This is translated into diminished production of hypothalamic effector peptides which are responsible for the coordination of energy homeostasis and satiety. So far, studies with animal models for PWS and with human post-mortem hypothalamic specimens demonstrated changes particularly in the infundibular and the paraventricular nuclei of the hypothalamus, both in orexigenic and anorexigenic neural populations. Moreover, many PWS patients have a severe endocrine dysfunction, e.g. central hypogonadism and/or growth hormone deficiency, which may contribute to the development of increased fat mass, especially if left untreated. Additionally, the role of non-neuronal cells, such as astrocytes and microglia in the hypothalamic dysregulation in PWS is yet to be determined. Notably, microglial activation is persistently present in non-genetic obesity. To what extent microglia, and other glial cells, are affected in PWS is poorly understood. The elucidation of the hypothalamic dysfunction in PWS could prove to be a key feature of rational therapeutic management in this syndrome. This review aims to examine the evidence for hypothalamic dysfunction, both at the neuropeptidergic and circuitry levels, and its correlation with the pathophysiology of PWS.
Collapse
Affiliation(s)
- Felipe Correa‐da‐Silva
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Eric Fliers
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
| | - Dick F. Swaab
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Chun‐Xia Yi
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| |
Collapse
|
170
|
García-Estévez L, Calvo I, Pérez S, Gallegos I, Díaz E, Sampayo-Cordero M, Oltra SS, Moreno-Bueno G. Predictive Role of Leptin Receptor (Ob-R) Overexpression in Patients with Early Breast Cancer Receiving Neoadjuvant Systemic Treatment. Cancers (Basel) 2021; 13:cancers13133269. [PMID: 34210055 PMCID: PMC8268260 DOI: 10.3390/cancers13133269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
The primary aim of this retrospective study was to investigate the correlation between the immunohistochemical expression of Ob-R (leptin receptor) with pCR (pathological complete response) in early breast cancer patients receiving neoadjuvant systemic treatment (NST). A total of 100 women with breast cancer receiving NST (2017-2020) followed by surgical resection were retrospectively obtained. Demographic parameters and clinicopathological factors (e.g., treatment modalities, immunohistochemistry (IHC), and cancer subtype) were obtained from the patient's clinical records. In the analyzed breast cancer cohort, high expression of Ob-R was found in 52% of tumors and there was a significantly higher incidence in the HER2+ and TNBC subgroups. Overall, a significantly greater percentage of patients with Ob-R positive tumors achieved pCR compared with Ob-R negative patients (57.7% vs. 27.1%; p = 0.002). This result was observed in most breast cancer subtypes. In patients with HER2+ breast cancer, there was no difference in Ob-R expression in relation to the HR status. Ob-R cell positivity was significantly higher in younger breast cancer patients (p = 0.008), those who were premenopausal (p = 0.011), and in those with a BMI > 25 kg/m2 (p = 0.019). A significantly greater percentage of early breast cancer patients with Ob-R positive tumors achieved pCR compared with Ob-R negative patients. Furthermore, breast cancer patients with positive Ob-R expression were significantly younger than those with negative Ob-R expression. This association was not explained by differences in BMI between young and old patients.
Collapse
Affiliation(s)
- Laura García-Estévez
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| | - Isabel Calvo
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
| | - Silvia Pérez
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
| | - Isabel Gallegos
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
| | - Eva Díaz
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
| | - Miguel Sampayo-Cordero
- Medica Scientia Innovation Research (MedSIR), 08018 Barcelona, Spain;
- Optimapharm, Biostatistics Department, Parc Bit Edifici Disset A2, 07121 Palma de Mallorca, Spain
| | - Sara S Oltra
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Biochemistry Department, Instituto de Investigaciones Biomédoicas ‘Alberto Sols’ (CSIC-UAM), Universidad Autónoma de Madrid (UAM), IdiPaz, 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Foundation MD Anderson International, C/Gómez Hemans 2, 28033 Madrid, Spain; (I.C.); (S.P.); (I.G.); (E.D.); (S.S.O.); (G.M.-B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Biochemistry Department, Instituto de Investigaciones Biomédoicas ‘Alberto Sols’ (CSIC-UAM), Universidad Autónoma de Madrid (UAM), IdiPaz, 28029 Madrid, Spain
| |
Collapse
|
171
|
Blaszczak AM, Jalilvand A, Hsueh WA. Adipocytes, Innate Immunity and Obesity: A Mini-Review. Front Immunol 2021; 12:650768. [PMID: 34248937 PMCID: PMC8264354 DOI: 10.3389/fimmu.2021.650768] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
The role of adipose tissue (AT) inflammation in obesity and its multiple related-complications is a rapidly expanding area of scientific interest. Within the last 30 years, the role of the adipocyte as an endocrine and immunologic cell has been progressively established. Like the macrophage, the adipocyte is capable of linking the innate and adaptive immune system through the secretion of adipokines and cytokines; exosome release of lipids, hormones, and microRNAs; and contact interaction with other immune cells. Key innate immune cells in AT include adipocytes, macrophages, neutrophils, and innate lymphoid cells type 2 (ILC2s). The role of the innate immune system in promoting adipose tissue inflammation in obesity will be highlighted in this review. T cells and B cells also play important roles in contributing to AT inflammation and are discussed in this series in the chapter on adaptive immunity.
Collapse
Affiliation(s)
- Alecia M Blaszczak
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| | - Anahita Jalilvand
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| | - Willa A Hsueh
- Hsueh Laboratory, The Ohio State University Wexner Medical Center, Diabetes and Metabolism Research Center, Columbus, OH, United States
| |
Collapse
|
172
|
Amorim MR, Dergacheva O, Fleury-Curado T, Pho H, Freire C, Mendelowitz D, Branco LGS, Polotsky VY. The Effect of DREADD Activation of Leptin Receptor Positive Neurons in the Nucleus of the Solitary Tract on Sleep Disordered Breathing. Int J Mol Sci 2021; 22:6742. [PMID: 34201760 PMCID: PMC8269100 DOI: 10.3390/ijms22136742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/19/2021] [Indexed: 12/14/2022] Open
Abstract
Obstructive sleep apnea (OSA) is recurrent obstruction of the upper airway due to the loss of upper airway muscle tone during sleep. OSA is highly prevalent, especially in obesity. There is no pharmacotherapy for OSA. Previous studies have demonstrated the role of leptin, an adipose-tissue-produced hormone, as a potent respiratory stimulant. Leptin signaling via a long functional isoform of leptin receptor, LEPRb, in the nucleus of the solitary tract (NTS), has been implicated in control of breathing. We hypothesized that leptin acts on LEPRb positive neurons in the NTS to increase ventilation and maintain upper airway patency during sleep in obese mice. We expressed designer receptors exclusively activated by designer drugs (DREADD) selectively in the LEPRb positive neurons of the NTS of Leprb-Cre-GFP mice with diet-induced obesity (DIO) and examined the effect of DREADD ligand, J60, on tongue muscle activity and breathing during sleep. J60 was a potent activator of LEPRb positive NTS neurons, but did not stimulate breathing or upper airway muscles during NREM and REM sleep. We conclude that, in DIO mice, the stimulating effects of leptin on breathing during sleep are independent of LEPRb signaling in the NTS.
Collapse
Affiliation(s)
- Mateus R. Amorim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; (T.F.-C.); (H.P.); (C.F.)
- Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-904, Brazil;
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA; (O.D.); (D.M.)
| | - Thomaz Fleury-Curado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; (T.F.-C.); (H.P.); (C.F.)
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; (T.F.-C.); (H.P.); (C.F.)
| | - Carla Freire
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; (T.F.-C.); (H.P.); (C.F.)
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA; (O.D.); (D.M.)
| | - Luiz G. S. Branco
- Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-904, Brazil;
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA; (T.F.-C.); (H.P.); (C.F.)
| |
Collapse
|
173
|
Impaired Leptin Signalling in Obesity: Is Leptin a New Thermolipokine? Int J Mol Sci 2021; 22:ijms22126445. [PMID: 34208585 PMCID: PMC8235268 DOI: 10.3390/ijms22126445] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
Leptin is a principal adipose-derived hormone mostly implicated in the regulation of energy balance through the activation of anorexigenic neuronal pathways. Comprehensive studies have established that the maintenance of certain concentrations of circulating leptin is essential to avoid an imbalance in nutrient intake. Indeed, genetic modifications of the leptin/leptin receptor axis and the obesogenic environment may induce changes in leptin levels or action in a manner that accelerates metabolic dysfunctions, resulting in a hyperphagic status and adipose tissue expansion. As a result, a vicious cycle begins wherein hyperleptinaemia and leptin resistance occur, in turn leading to increased food intake and fat enlargement, which is followed by leptin overproduction. In addition, in the context of obesity, a defective thermoregulatory response is associated with impaired leptin signalling overall within the ventromedial nucleus of the hypothalamus. These recent findings highlight the role of leptin in the regulation of adaptive thermogenesis, thus suggesting leptin to be potentially considered as a new thermolipokine. This review provides new insight into the link between obesity, hyperleptinaemia, leptin resistance and leptin deficiency, focusing on the ability to restore leptin sensitiveness by way of enhanced thermogenic responses and highlighting novel anti-obesity therapeutic strategies.
Collapse
|
174
|
Kuiper-Makris C, Selle J, Nüsken E, Dötsch J, Alejandre Alcazar MA. Perinatal Nutritional and Metabolic Pathways: Early Origins of Chronic Lung Diseases. Front Med (Lausanne) 2021; 8:667315. [PMID: 34211985 PMCID: PMC8239134 DOI: 10.3389/fmed.2021.667315] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Lung development is not completed at birth, but expands beyond infancy, rendering the lung highly susceptible to injury. Exposure to various influences during a critical window of organ growth can interfere with the finely-tuned process of development and induce pathological processes with aberrant alveolarization and long-term structural and functional sequelae. This concept of developmental origins of chronic disease has been coined as perinatal programming. Some adverse perinatal factors, including prematurity along with respiratory support, are well-recognized to induce bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease that is characterized by arrest of alveolar and microvascular formation as well as lung matrix remodeling. While the pathogenesis of various experimental models focus on oxygen toxicity, mechanical ventilation and inflammation, the role of nutrition before and after birth remain poorly investigated. There is accumulating clinical and experimental evidence that intrauterine growth restriction (IUGR) as a consequence of limited nutritive supply due to placental insufficiency or maternal malnutrition is a major risk factor for BPD and impaired lung function later in life. In contrast, a surplus of nutrition with perinatal maternal obesity, accelerated postnatal weight gain and early childhood obesity is associated with wheezing and adverse clinical course of chronic lung diseases, such as asthma. While the link between perinatal nutrition and lung health has been described, the underlying mechanisms remain poorly understood. There are initial data showing that inflammatory and nutrient sensing processes are involved in programming of alveolarization, pulmonary angiogenesis, and composition of extracellular matrix. Here, we provide a comprehensive overview of the current knowledge regarding the impact of perinatal metabolism and nutrition on the lung and beyond the cardiopulmonary system as well as possible mechanisms determining the individual susceptibility to CLD early in life. We aim to emphasize the importance of unraveling the mechanisms of perinatal metabolic programming to develop novel preventive and therapeutic avenues.
Collapse
Affiliation(s)
- Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Member of the German Centre for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Gießen, Germany
| |
Collapse
|
175
|
Pho H, Berger S, Freire C, Kim LJ, Shin MK, Streeter SR, Hosamane N, Cabassa ME, Anokye-Danso F, Dergacheva O, Amorim MR, Fleury-Curado T, Jun JC, Schwartz AR, Ahima RS, Mendelowitz D, Polotsky VY. Leptin receptor expression in the dorsomedial hypothalamus stimulates breathing during NREM sleep in db/db mice. Sleep 2021; 44:6149135. [PMID: 33624805 PMCID: PMC8193564 DOI: 10.1093/sleep/zsab046] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY OBJECTIVES Obesity leads to obstructive sleep apnea (OSA), which is recurrent upper airway obstruction during sleep, and obesity hypoventilation syndrome (OHS), hypoventilation during sleep resulting in daytime hypercapnia. Impaired leptin signaling in the brain was implicated in both conditions, but mechanisms are unknown. We have previously shown that leptin stimulates breathing and treats OSA and OHS in leptin-deficient ob/ob mice and leptin-resistant diet-induced obese mice and that leptin's respiratory effects may occur in the dorsomedial hypothalamus (DMH). We hypothesized that leptin receptor LepRb-deficient db/db mice have obesity hypoventilation and that restoration of leptin signaling in the DMH will increase ventilation during sleep in these animals. METHODS We measured arterial blood gas in unanesthetized awake db/db mice. We subsequently infected these animals with Ad-LepRb or control Ad-mCherry virus into the DMH and measured ventilation during sleep as well as CO2 production after intracerebroventricular (ICV) infusions of phosphate-buffered saline or leptin. RESULTS Awake db/db mice had elevated CO2 levels in the arterial blood. Ad-LepRb infection resulted in LepRb expression in the DMH neurons in a similar fashion to wildtype mice. In LepRb-DMH db/db mice, ICV leptin shortened REM sleep and increased inspiratory flow, tidal volume, and minute ventilation during NREM sleep without any effect on the quality of NREM sleep or CO2 production. Leptin had no effect on upper airway obstruction in these animals. CONCLUSION Leptin stimulates breathing and treats obesity hypoventilation acting on LepRb-positive neurons in the DMH.
Collapse
Affiliation(s)
- Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Slava Berger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla Freire
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stone R Streeter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nishitha Hosamane
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meaghan E Cabassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Mateus R Amorim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomaz Fleury-Curado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan C Jun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Corresponding author. Vsevolod (Seva) Y. Polotsky, Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Johns Hopkins Asthma and Allergy Center, Rm 4B65, Baltimore, MD 21224.
| |
Collapse
|
176
|
Fatty acids role on obesity induced hypothalamus inflammation: From problem to solution – A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
177
|
Shuai L, Li BH, Jiang HW, Yang L, Li J, Li JY. DOT1L Regulates Thermogenic Adipocyte Differentiation and Function via Modulating H3K79 Methylation. Diabetes 2021; 70:1317-1333. [PMID: 33795413 DOI: 10.2337/db20-1110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022]
Abstract
Brown and beige adipocytes are characterized as thermogenic adipocytes and have great potential for treating obesity and associated metabolic diseases. In this article, we identify a conserved mammalian lysine 79 of histone H3 (H3K79) methyltransferase, disruptor of telomeric silencing-1 like (DOT1L), as a new epigenetic regulator that controls thermogenic adipocyte differentiation and function. We show that deletion of DOT1L in thermogenic adipocytes potently protects mice from diet-induced obesity, improves glucose homeostasis, alleviates hepatic steatosis, and facilitates adaptive thermogenesis in vivo. Loss of DOT1L in primary preadipocytes significantly promotes brown and beige adipogenesis and thermogenesis in vitro. Mechanistically, DOT1L epigenetically regulates the brown adipose tissue-selective gene program by modulating H3K79 methylation, in particular H3K79me2 modification. Thus, our study demonstrates that DOT1L exerts an important role in energy homeostasis by regulating thermogenic adipocyte differentiation and function.
Collapse
Affiliation(s)
- Lin Shuai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Bo-Han Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Wen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lin Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Jing-Ya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
178
|
Hayden MR, Banks WA. Deficient Leptin Cellular Signaling Plays a Key Role in Brain Ultrastructural Remodeling in Obesity and Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:5427. [PMID: 34063911 PMCID: PMC8196569 DOI: 10.3390/ijms22115427] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
The triad of obesity, metabolic syndrome (MetS), Type 2 diabetes mellitus (T2DM) and advancing age are currently global societal problems that are expected to grow over the coming decades. This triad is associated with multiple end-organ complications of diabetic vasculopathy (maco-microvessel disease), neuropathy, retinopathy, nephropathy, cardiomyopathy, cognopathy encephalopathy and/or late-onset Alzheimer's disease. Further, obesity, MetS, T2DM and their complications are associated with economical and individual family burdens. This review with original data focuses on the white adipose tissue-derived adipokine/hormone leptin and how its deficient signaling is associated with brain remodeling in hyperphagic, obese, or hyperglycemic female mice. Specifically, the ultrastructural remodeling of the capillary neurovascular unit, brain endothelial cells (BECs) and their endothelial glycocalyx (ecGCx), the blood-brain barrier (BBB), the ventricular ependymal cells, choroid plexus, blood-cerebrospinal fluid barrier (BCSFB), and tanycytes are examined in female mice with impaired leptin signaling from either dysfunction of the leptin receptor (DIO and db/db models) or the novel leptin deficiency (BTBR ob/ob model).
Collapse
Affiliation(s)
- Melvin R. Hayden
- Departments of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri-Columbia School of Medicine, One Hospital Drive, Columbia, MO 65212, USA;
| | - William A. Banks
- Geriatrics Research, Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, 1660 S. Columbian Way, 810C/Bldg 1, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98108, USA
| |
Collapse
|
179
|
Yang M, Qiu S, He Y, Li L, Wu T, Ding N, Li F, Zhao AZ, Yang G. Genetic ablation of C-reactive protein gene confers resistance to obesity and insulin resistance in rats. Diabetologia 2021; 64:1169-1183. [PMID: 33544171 DOI: 10.1007/s00125-021-05384-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 11/06/2020] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Besides serving as a traditional inflammatory marker, C-reactive protein (CRP) is closely associated with the development of obesity, diabetes and cardiovascular diseases as a metabolic and inflammatory marker. We hypothesise that CRP protein directly participates in the regulation of energy and glucose metabolism rather than just being a surrogate marker, and that genetic deficiency of CRP will lead to resistance to obesity and insulin resistance. METHODS Crp gene deletion was achieved by transcription activator-like effector nuclease (TALEN) technology in rats. The Crp knockout animals were placed on either a standard chow diet or a high-fat diet. Phenotypic changes in body weight, glucose metabolism, insulin sensitivity, energy expenditure and inflammation condition were examined. The central impact of CRP deficiency on leptin and insulin hypothalamic signalling, as well as glucose homeostasis, were examined via intracerebral ventricular delivery of leptin and CRP plus glucose clamp studies in the wild-type and Crp knockout rats. RESULTS CRP deficiency led to a significant reduction in weight gain and food intake, elevated energy expenditure and improved insulin sensitivity after exposure to high-fat diet. Glucose clamp studies revealed enhanced hepatic insulin signalling and actions. Deficiency of CRP enhanced and prolonged the weight-reducing effect of central injected leptin and promoted the central and peripheral roles of leptin. By contrast, reinstatement of CRP into the hypothalamus of the knockout rats attenuated the effects of central leptin signalling on insulin sensitivity and peripheral glucose metabolism. CONCLUSIONS/INTERPRETATION This study represents the first line of genetic evidence that CRP is not merely a surrogate blood marker for inflammation and metabolic syndromes but directly regulates energy balance, body weight, insulin sensitivity and glucose homeostasis through direct regulation of leptin's central effect and hypothalamic signalling.
Collapse
Affiliation(s)
- Mengliu Yang
- Department of Endocrinology, the 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing and Department of Endocrinology, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Sheng Qiu
- Department of Endocrinology, the 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yirui He
- Department of Endocrinology, the 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling Li
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Tong Wu
- Department of Endocrinology, the 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ning Ding
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Fanghong Li
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Allan Z Zhao
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Gangyi Yang
- Department of Endocrinology, the 2nd Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
180
|
Lima RS, Mattos RT, Medeiros NI, Kattah FM, Nascimento JRS, Menezes CA, Rios-Santos F, Dutra WO, Gomes JAS, Moreira PR. CXCL8 expression and methylation are correlated with anthropometric and metabolic parameters in childhood obesity. Cytokine 2021; 143:155538. [PMID: 33926776 DOI: 10.1016/j.cyto.2021.155538] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Childhood obesity is a global and increasing health issue. Inflammation and dysregulated adipose tissue secretion are common findings in obesity and have been related to poor metabolic function. Given that DNA methylation impacts gene expression and is responsive to environmental changes, we aimed, in addition to characterize the patients in anthropometric and biochemical terms, to determine the expression of cytokines and adipokines, assess the methylation on regulatory regions of the genes that code for these molecules, and investigate the association of the expression and gene methylation with anthropometric and biochemical parameters in childhood obesity. Obese children present dyslipidemia, dysregulated serum levels of adipokines and their ratios, altered leukocytic expression of cytokines, and higher methylation at the CXCL8 promoter as compared to the control group. However, no significant results were observed in the fasting plasma glucose levels or the methylation of TGFB1, LEP, and the enhancer region of ADIPOQ. We also found negative correlations of CXCL8 expression with anthropometric and biochemical parameters, and positive correlation of CXCL8 promoter methylation and the serum levels of hepatic enzymes. Our results indicate that changes in metabolic parameters observed in childhood obesity are associated with the expression of adipokines and cytokines, and the methylation status at the CXCL8 promoter. CXCL8 may be a key factor for these alterations, as it correlates with many of the parameters assessed in the present study.
Collapse
Affiliation(s)
- Rafael S Lima
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Rafael T Mattos
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Nayara I Medeiros
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Fabiana M Kattah
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Julya R S Nascimento
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Carlos A Menezes
- Department of Genetics, State University of Santa Cruz, Bahia, Brazil; Service of Preventive Medicine - Unimed, Sergipe, Brazil
| | - Fabricio Rios-Santos
- Department of Basic and Health Sciences, Federal University of Mato Grosso, Mato Grosso, Brazil
| | - Walderez O Dutra
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil; National Institute of Science and Technology in Tropical Diseases - INCT-DT, Brazil
| | - Juliana A S Gomes
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Paula R Moreira
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil.
| |
Collapse
|
181
|
Yeung BHY, Griffiths K, Berger L, Paudel O, Shin MK, Rui L, Sham JSK, Polotsky VY, Tang WY. Leptin Induces Epigenetic Regulation of Transient Receptor Potential Melastatin 7 in Rat Adrenal Pheochromocytoma Cells. Am J Respir Cell Mol Biol 2021; 65:214-221. [PMID: 33891828 DOI: 10.1165/rcmb.2020-0374oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Obesity elevates the plasma level of leptin, which has been associated with hypertension. Our recent studies in mice demonstrated that leptin increases blood pressure by activating the carotid sinus nerve, which transmits the chemosensory input from carotid bodies (CBs) to the medullary centers, and that the effect of leptin is mediated via Trpm7 (TRP [transient receptor potential] melastatin 7) channels in CB glomus cells. We also found that Trpm7 overexpression and Trpm7 promoter demethylation in CBs correlate positively with the hyperleptinemia and leptin receptor overexpression in CBs. Hence, we postulated that leptin epigenetically regulates Trpm7 expression in CBs. We addressed our hypothesis by using rat adrenal pheochromocytoma (PC12) cells as a model of CB glomus cells. PC12 cells expressing LEPRb (long, active form of leptin receptor) showed dramatic induction of the promoter activity and expression of Trpm7 upon leptin treatment. The increased Trpm7 expression coincided with the reduction of CpG site-specific methylation and trimethylation of H3K27 (H3 [histone 3] K27 [lysine 27]) and the increase of acetylation of H3K27 and trimethylation of H3K4 (H3 lysine 4) at the Trpm7 promoter. The inhibitor of STAT3 (signal transducer and activator of transcription 3) signaling, SD1008, reversed the leptin-induced Trpm7 promoter activity via modulations of the binding of pSTAT3 (phosphorylated STAT3) and DNMT3B (DNA methyltransferase 3B) and modifications of H3K27 and H3K4 at the Trpm7 promoter. Our results suggest that leptin-activated pSTAT3 epigenetically regulates the transcription of Trpm7 through DNA methylation and histone modifications. Because epigenetic changes are reversible, targeting epigenetic modifications of Trpm7 may serve as a new therapeutic approach for the treatment of hypertension in obesity.
Collapse
Affiliation(s)
- Bonnie Ho-Yee Yeung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kelly Griffiths
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and
| | - Liron Berger
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Omkar Paudel
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Mi-Kyung Shin
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan
| | - James S K Sham
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Vsevolod Y Polotsky
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Wan-Yee Tang
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, and.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
182
|
Irving A, Harvey J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog Lipid Res 2021; 82:101098. [PMID: 33895229 DOI: 10.1016/j.plipres.2021.101098] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Significant advances have been made in our understanding of the hormone, leptin and its CNS actions in recent years. It is now evident that leptin has a multitude of brain functions, that extend beyond its established role in the hypothalamic control of energy balance. Additional brain regions including the hippocampus are important targets for leptin, with a high density of leptin receptors (LepRs) expressed in specific hippocampal regions and localised to CA1 synapses. Extensive evidence indicates that leptin has pro-cognitive actions, as it rapidly modifies synaptic efficacy at excitatory Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 synapses and enhances performance in hippocampal-dependent memory tasks. There is a functional decline in hippocampal responsiveness to leptin with age, with significant reductions in the modulatory effects of leptin at SC-CA1 and TA-CA1 synapses in aged, compared to adult hippocampus. As leptin has pro-cognitive effects, this decline in leptin sensitivity is likely to have negative consequences for cognitive function during the aging process. Here we review how evaluation of the hippocampal actions of leptin has improved our knowledge of the regulatory brain functions of leptin in health and provided significant insight into the impact of leptin in age-related neurodegenerative disorders linked to cognitive decline.
Collapse
Affiliation(s)
- Andrew Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
183
|
Wołodko K, Castillo-Fernandez J, Kelsey G, Galvão A. Revisiting the Impact of Local Leptin Signaling in Folliculogenesis and Oocyte Maturation in Obese Mothers. Int J Mol Sci 2021; 22:4270. [PMID: 33924072 PMCID: PMC8074257 DOI: 10.3390/ijms22084270] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022] Open
Abstract
The complex nature of folliculogenesis regulation accounts for its susceptibility to maternal physiological fitness. In obese mothers, progressive expansion of adipose tissue culminates with severe hyperestrogenism and hyperleptinemia with detrimental effects for ovarian performance. Indeed, maternal obesity is associated with the establishment of ovarian leptin resistance. This review summarizes current knowledge on potential effects of impaired leptin signaling throughout folliculogenesis and oocyte developmental competence in mice and women.
Collapse
Affiliation(s)
- Karolina Wołodko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, Tuwima 10, 10-748 Olsztyn, Poland;
| | | | - Gavin Kelsey
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; (J.C.-F.); (G.K.)
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - António Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of PAS, Tuwima 10, 10-748 Olsztyn, Poland;
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; (J.C.-F.); (G.K.)
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
184
|
Evaluation of Appetite-Regulating Hormones ın Young Children with Autism Spectrum Disorder. J Autism Dev Disord 2021; 51:632-643. [PMID: 32583136 DOI: 10.1007/s10803-020-04579-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study aimed to investigate the role of leptin, ghrelin, neuropeptide Y, and nesfatin-1 in young children with autism spectrum disorder (ASD). A total of 44 children with ASD and 44 healthy controls aged 18-60 months were included. Plasma levels of hormones were measured using commercial enzyme-linked immunosorbent assay kits. Plasma leptin and ghrelin levels were significantly higher in the ASD group than in the control group. However, no significant difference for plasma neuropeptide Y and nesfatin-1 levels was detected between the groups. No relation was found between the severity of ASD symptoms, severity of eating problems, and plasma levels of hormones. Leptin and ghrelin may play a potential role in the pathogenesis of ASD.
Collapse
|
185
|
de Candia P, Prattichizzo F, Garavelli S, Alviggi C, La Cava A, Matarese G. The pleiotropic roles of leptin in metabolism, immunity, and cancer. J Exp Med 2021; 218:211994. [PMID: 33857282 PMCID: PMC8056770 DOI: 10.1084/jem.20191593] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The discovery of the archetypal adipocytokine leptin and how it regulates energy homeostasis have represented breakthroughs in our understanding of the endocrine function of the adipose tissue and the biological determinants of human obesity. Investigations on leptin have also been instrumental in identifying physio-pathological connections between metabolic regulation and multiple immunological functions. For example, the description of the promoting activities of leptin on inflammation and cell proliferation have recognized the detrimental effects of leptin in connecting dysmetabolic conditions with cancer and with onset and/or progression of autoimmune disease. Here we review the multiple biological functions and complex framework of operations of leptin, discussing why and how the pleiotropic activities of this adipocytokine still pose major hurdles in the development of effective leptin-based therapeutic opportunities for different clinical conditions.
Collapse
Affiliation(s)
- Paola de Candia
- Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Milan, Italy
| | | | - Silvia Garavelli
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, Università di Napoli "Federico II," Naples, Italy
| | - Antonio La Cava
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Giuseppe Matarese
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Naples, Italy.,T reg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II," Naples, Italy
| |
Collapse
|
186
|
Anderson MR, Kim JS, Allison M, Giles JT, Hoffman EA, Ding J, Barr RG, Podolanczuk A. Adiposity and Interstitial Lung Abnormalities in Community-Dwelling Adults: The MESA Cohort Study. Chest 2021; 160:582-594. [PMID: 33844978 DOI: 10.1016/j.chest.2021.03.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Obesity is associated with restrictive ventilatory defects and a faster rate of decline in FVC. This association is not exclusively mediated by mechanical factors and may reflect direct pulmonary injury by adipose-derived mediators. RESEARCH QUESTION Is adipose tissue involved in the pathogenesis of interstitial lung disease (ILD)? STUDY DESIGN AND METHODS We evaluated the association of CT measures of pericardial, abdominal visceral, and abdominal subcutaneous adipose tissue with high-attenuation areas (HAAs) and interstitial lung abnormalities (ILAs) in a large multicenter cohort study of community-dwelling adults, using multivariable-adjusted models. We secondarily evaluated the association of adipose depot size with FVC and biomarkers of obesity and inflammation. RESULTS In fully adjusted models, every doubling in pericardial adipose tissue volume was associated with a 63.4-unit increase in HAA (95% CI, 55.5-71.3), 20% increased odds of ILA (95% CI, -2% to 50%), and a 5.5% decrease in percent predicted FVC (95% CI, -6.8% to -4.3%). IL-6 levels accounted for 8% of the association between pericardial adipose tissue and HAA. Every doubling in visceral adipose tissue area was associated with a 41.5-unit increase in HAA (95% CI, 28.3-54.7), 30% increased odds of ILA (95% CI, -10% to 80%), and a 5.4% decrease in percent predicted FVC (95% CI, -6.6% to -4.3%). IL-6 and leptin accounted for 17% and 18%, respectively, of the association between visceral adipose tissue and HAA. INTERPRETATION Greater amounts of pericardial and abdominal visceral adipose tissue were associated with CT measures of early lung injury and lower FVC in a cohort of community-dwelling adults. Adipose tissue may represent a modifiable risk factor for ILD.
Collapse
Affiliation(s)
| | - John S Kim
- Department of Medicine, University of Virginia, Charlottesville, VA
| | - Matthew Allison
- Department of Preventive Medicine, University of California San Diego, San Diego, CA
| | - Jon T Giles
- Department of Medicine, Columbia University Medical Center, New York, NY
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Des Moines, IA
| | - Jingzhong Ding
- Department of Epidemiology and Prevention, Wake Forest University, Winston-Salem, NC; Department of Gerontology and Geriatric Science, Wake Forest University, Winston-Salem, NC
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, NY; Department of Epidemiology, Columbia University Medical Center, New York, NY
| | - Anna Podolanczuk
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
187
|
Blanco AM, Soengas JL. Leptin signalling in teleost fish with emphasis in food intake regulation. Mol Cell Endocrinol 2021; 526:111209. [PMID: 33588023 DOI: 10.1016/j.mce.2021.111209] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/14/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Leptin, the product of the obese (ob or Lep) gene, was first cloned in teleost fish in 2005, more than a decade after its identification in mammals. This was because bony fish and mammalian leptins share a very low amino acid sequence identity, which suggests different functionality of the leptin system in fish compared to that of mammals. Indeed, major differences are evident between the mammalian and fish leptin system. Thus, for instance, mammalian leptin is synthesized and released by the adipose tissue in response to the amount of fat depots, while several tissues (mainly the liver) are the main sources of leptin in fish, whose determining factors of production are still unclear. In mammals, the main physiological role for leptin is its involvement in the maintenance of energy balance by decreasing food intake and increasing energy expenditure, although a wide variety of actions have been attributed to this hormone (e.g., regulation of lipid and carbohydrate metabolism, reproduction and immune functions). In fish, available literature also points towards a multifunctional nature for leptin, although knowledge on its functions is limited. In this review, we offer an overview of teleostean leptin structure and mechanism of action, and discuss the available knowledge on the role of this hormone in food intake regulation in teleost fish, aiming to provide a comparative overview between the functioning of the teleostean and mammalian leptin systems.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Pontevedra, Spain
| | - José Luis Soengas
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Pontevedra, Spain.
| |
Collapse
|
188
|
Garrido-Amaro C, Cardona P, Gassó D, Arias L, Velarde R, Tvarijonativiciute A, Serrano E, Cardona PJ. Protective Effect of Intestinal Helminthiasis Against Tuberculosis Progression Is Abrogated by Intermittent Food Deprivation. Front Immunol 2021; 12:627638. [PMID: 33936040 PMCID: PMC8079633 DOI: 10.3389/fimmu.2021.627638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background Tuberculosis (TB) is still a major challenge for humankind. Because regions with the highest incidence also have a high prevalence of helminthiasis and nutritional scarcity, we wanted to understand the impact of these on TB progression. Methods We have developed an experimental murine model for active TB in C3HeB/FeJ, coinfected with Trichuris muris and Heligmosomoides polygyrus nematodes, and exposed to an environmental mycobacterium (M. manresensis) and intermittent fasting. Cause-effect relationships among these factors were explored with Partial Least Squares Path modelling (PLSPM). Results Previous parasitization had a major anti-inflammatory effect and reduced systemic levels of ADA, haptoglobin, local pulmonary levels of IL-1β, IL-6, TNF-α, CXCL-1, CXCL-5 and IL-10. Oral administration of heat-killed M. manresensis resulted in a similar outcome. Both interventions diminished pulmonary pathology and bacillary load, but intermittent food deprivation reduced this protective effect increasing stress and inflammation. The PLSPM revealed nematodes might have protective effects against TB progression. Conclusions Significantly higher cortisol levels in food-deprivation groups showed it is a stressful condition, which might explain its deleterious effect. This highlights the impact of food security on TB eradication policies and the need to prioritize food supply over deworming activities.
Collapse
Affiliation(s)
- Cristina Garrido-Amaro
- Wildlife Ecology & Health Group (WE&H) and Servei d’Ecopatologia de Fauna Salvatge (SEFaS), Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Paula Cardona
- Unitat de Tuberculosi Experimental, Institut Germans Trias i Pujol, UAB, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Diana Gassó
- Wildlife Ecology & Health Group (WE&H) and Servei d’Ecopatologia de Fauna Salvatge (SEFaS), Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
- Departament of Animal Science, Agrifood, Forestry and Veterinary Campus, University of Lleida, Lleida, Spain
| | - Lilibeth Arias
- Unitat de Tuberculosi Experimental, Institut Germans Trias i Pujol, UAB, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Roser Velarde
- Wildlife Ecology & Health Group (WE&H) and Servei d’Ecopatologia de Fauna Salvatge (SEFaS), Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Asta Tvarijonativiciute
- Interdisciplinary Laboratory of Clinical Analysis Interlab-UMU, Regional Campus of International Excellence Campus Mare Nostrum, Universidad de Murcia, Murcia, Spain
| | - Emmanuel Serrano
- Wildlife Ecology & Health Group (WE&H) and Servei d’Ecopatologia de Fauna Salvatge (SEFaS), Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Pere-Joan Cardona
- Unitat de Tuberculosi Experimental, Institut Germans Trias i Pujol, UAB, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
189
|
Bishop EL, Gudgeon N, Dimeloe S. Control of T Cell Metabolism by Cytokines and Hormones. Front Immunol 2021; 12:653605. [PMID: 33927722 PMCID: PMC8076900 DOI: 10.3389/fimmu.2021.653605] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Dynamic, coordinated changes in metabolic pathway activity underpin the protective and inflammatory activity of T cells, through provision of energy and biosynthetic precursors for effector functions, as well as direct effects of metabolic enzymes, intermediates and end-products on signaling pathways and transcriptional mechanisms. Consequently, it has become increasingly clear that the metabolic status of the tissue microenvironment directly influences T cell activity, with changes in nutrient and/or metabolite abundance leading to dysfunctional T cell metabolism and interlinked immune function. Emerging evidence now indicates that additional signals are integrated by T cells to determine their overall metabolic phenotype, including those arising from interaction with cytokines and hormones in their environment. The impact of these on T cell metabolism, the mechanisms involved and the pathological implications are discussed in this review article.
Collapse
Affiliation(s)
| | | | - Sarah Dimeloe
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
190
|
Abstract
Obesity is epidemiologically linked to 13 forms of cancer. The local and systemic obese environment is complex and likely affect tumors through multiple avenues. This includes modulation of cancer cell phenotypes and the composition of the tumor microenvironment. A molecular understanding of how obesity links to cancer holds promise for identifying candidate genes for targeted therapy for obese cancer patient. Herein, we review both the cell-autonomous and non-cell-autonomous mechanisms linking obesity and cancer as well as provide an overview of the mouse model systems applied to study this.
Collapse
Affiliation(s)
- Xiao-Zheng Liu
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Line Pedersen
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, N-5020 Bergen, Norway
| |
Collapse
|
191
|
O'Connor S, Murphy EA, Szwed SK, Kanke M, Marchildon F, Sethupathy P, Darnell RB, Cohen P. AGO HITS-CLIP reveals distinct miRNA regulation of white and brown adipose tissue identity. Genes Dev 2021; 35:771-781. [PMID: 33832988 PMCID: PMC8091975 DOI: 10.1101/gad.345447.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are short, noncoding RNAs that associate with Argonaute (AGO) to influence mRNA stability and translation, thereby regulating cellular determination and phenotype. While several individual miRNAs have been shown to control adipocyte function, including energy storage in white fat and energy dissipation in brown fat, a comprehensive analysis of miRNA activity in these tissues has not been performed. We used high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP) to comprehensively characterize the network of high-confidence, in vivo mRNA:miRNA interactions across white and brown fat, revealing >20,000 unique AGO binding sites. When coupled with miRNA and mRNA sequencing, we found an inverse correlation between depot-enriched miRNAs and their targets. To illustrate the functionality of our HITS-CLIP data set in identifying specific miRNA:mRNA interactions, we show that miR-29 is a novel regulator of leptin, an adipocyte-derived hormone that coordinates food intake and energy homeostasis. Two independent miR-29 binding sites in the leptin 3' UTR were validated using luciferase assays, and miR-29 gain and loss of function modulated leptin mRNA and protein secretion in primary adipocytes. This work represents the only experimentally generated miRNA targetome in adipose tissue and identifies multiple regulatory pathways that may specify the unique identities of white and brown fat.
Collapse
Affiliation(s)
- Sean O'Connor
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York 10065, USA
| | - Elisabeth A Murphy
- Laboratory of Molecular Neuro-Oncology, The Rockefeller University, New York, New York 10065, USA
| | - Sarah K Szwed
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York 10065, USA.,Weill-Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York 10065, USA
| | - Matt Kanke
- Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York 10065, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Robert B Darnell
- Laboratory of Molecular Neuro-Oncology, The Rockefeller University, New York, New York 10065, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
192
|
Fujikawa T. Central regulation of glucose metabolism in an insulin-dependent and -independent manner. J Neuroendocrinol 2021; 33:e12941. [PMID: 33599044 DOI: 10.1111/jne.12941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS) contributes significantly to glucose homeostasis. The available evidence indicates that insulin directly acts on the CNS, in particular the hypothalamus, to regulate hepatic glucose production, thereby controlling whole-body glucose metabolism. Additionally, insulin also acts on the brain to regulate food intake and fat metabolism, which may indirectly regulate glucose metabolism. Studies conducted over the last decade have found that the CNS can regulate glucose metabolism in an insulin-independent manner. Enhancement of central leptin signalling reverses hyperglycaemia in insulin-deficient rodents. Here, I review the mechanisms by which central insulin and leptin actions regulate glucose metabolism. Although clinical studies have shown that insulin treatment is currently indispensable for managing diabetes, unravelling the neuronal mechanisms underlying the central regulation of glucose metabolism will pave the way for the design of novel therapeutic drugs for diabetes.
Collapse
Affiliation(s)
- Teppei Fujikawa
- Center for Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
193
|
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Overweight and obesity are strongly associated with comorbidities such as hypertension and insulin resistance, which collectively contribute to the development of cardiovascular diseases and resultant morbidity and mortality. Forty-two percent of adults in the United States are obese, and a total of 1.9 billion adults worldwide are overweight or obese. These alarming numbers, which continue to climb, represent a major health and economic burden. Adipose tissue is a highly dynamic organ that can be classified based on the cellular composition of different depots and their distinct anatomical localization. Massive expansion and remodeling of adipose tissue during obesity differentially affects specific adipose tissue depots and significantly contributes to vascular dysfunction and cardiovascular diseases. Visceral adipose tissue accumulation results in increased immune cell infiltration and secretion of vasoconstrictor mediators, whereas expansion of subcutaneous adipose tissue is less harmful. Therefore, fat distribution more than overall body weight is a key determinant of the risk for cardiovascular diseases. Thermogenic brown and beige adipose tissue, in contrast to white adipose tissue, is associated with beneficial effects on the vasculature. The relationship between the type of adipose tissue and its influence on vascular function becomes particularly evident in the context of the heterogenous phenotype of perivascular adipose tissue that is strongly location dependent. In this review, we address the abnormal remodeling of specific adipose tissue depots during obesity and how this critically contributes to the development of hypertension, endothelial dysfunction, and vascular stiffness. We also discuss the local and systemic roles of adipose tissue derived secreted factors and increased systemic inflammation during obesity and highlight their detrimental impact on cardiovascular health.
Collapse
Affiliation(s)
- Mascha Koenen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York (M.K., P.C.)
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.)
- Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York (M.K., P.C.)
| | - James R Sowers
- Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.)
- Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia
- Diabetes and Cardiovascular Center (J.R.S.), University of Missouri School of Medicine, Columbia
- Department of Medicine (J.R.S.), University of Missouri School of Medicine, Columbia
| |
Collapse
|
194
|
Bhardwaj P, Brown KA. Obese Adipose Tissue as a Driver of Breast Cancer Growth and Development: Update and Emerging Evidence. Front Oncol 2021; 11:638918. [PMID: 33859943 PMCID: PMC8042134 DOI: 10.3389/fonc.2021.638918] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an established risk factor for breast cancer growth and progression. A number of advances have been made in recent years revealing new insights into this link. Early events in breast cancer development involve the neoplastic transformation of breast epithelial cells to cancer cells. In obesity, breast adipose tissue undergoes significant hormonal and inflammatory changes that create a mitogenic microenvironment. Many factors that are produced in obesity have also been shown to promote tumorigenesis. Given that breast epithelial cells are surrounded by adipose tissue, the crosstalk between the adipose compartment and breast epithelial cells is hypothesized to be a significant player in the initiation and progression of breast cancer in individuals with excess adiposity. The present review examines this crosstalk with a focus on obese breast adipose-derived estrogen, inflammatory mediators and adipokines, and how they are mechanistically linked to breast cancer risk and growth through stimulation of oxidative stress, DNA damage, and pro-oncogenic transcriptional programs. Pharmacological and lifestyle strategies targeting these factors and their downstream effects are evaluated for feasibility and efficacy in decreasing the risk of obesity-induced breast epithelial cell transformation and consequently, breast cancer development.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
195
|
Njunge JM, Gonzales GB, Ngari MM, Thitiri J, Bandsma RH, Berkley JA. Systemic inflammation is negatively associated with early post discharge growth following acute illness among severely malnourished children - a pilot study. Wellcome Open Res 2021; 5:248. [PMID: 33969227 PMCID: PMC8080977 DOI: 10.12688/wellcomeopenres.16330.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Rapid growth should occur among children with severe malnutrition (SM) with medical and nutritional management. Systemic inflammation (SI) is associated with death among children with SM and is negatively associated with linear growth. However, the relationship between SI and weight gain during therapeutic feeding following acute illness is unknown. We hypothesised that growth post-hospital discharge is associated with SI among children with SM. Methods: We conducted secondary analysis of data from HIV-uninfected children with SM (n=98) who survived and were not readmitted to hospital during one year of follow-up. We examined the relationship between changes in absolute deficits in weight and mid-upper-arm circumference (MUAC) from enrolment at stabilisation to 60 days and one year later, and untargeted plasma proteome, targeted cytokines/chemokines, leptin, and soluble CD14 using multivariate regularized linear regression. Results: The mean change in absolute deficit in weight and MUAC was -0.50kg (standard deviation; SD±0.69) and -1.20cm (SD±0.89), respectively, from enrolment to 60 days later. During the same period, mean weight and MUAC gain was 3.3g/kg/day (SD±2.4) and 0.22mm/day (SD±0.2), respectively. Enrolment interleukins; IL17-alpha and IL-2, and serum amyloid P were negatively associated with weight and MUAC gain during 60 days. Lipopolysaccharide binding protein and complement component 2 were negatively associated with weight gain only. Leptin was positively associated with weight gain. Soluble CD14, beta-2 microglobulin, and macrophage inflammatory protein 1 beta were negatively associated with MUAC gain only. Glutathione peroxidase 3 was positively associated with weight and MUAC gain during one year. Conclusions: Early post-hospital discharge weight and MUAC gain were rapid and comparable to children with uncomplicated SM treated in the community. Higher concentrations of SI markers were associated with less weight and MUAC gain, suggesting inflammation negatively impacts recovery from wasting. This finding warrants further research on reducing inflammation on growth among children with SM.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Gerard Bryan Gonzales
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Moses M. Ngari
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Robert H.J. Bandsma
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James A. Berkley
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
196
|
Chong L, Li H, Zhu L, Yu G. Regulatory effect of mitoQ on the mtROS-NLRP3 inflammasome pathway in leptin-pretreated BEAS-2 cells. Exp Ther Med 2021; 21:466. [PMID: 33763153 PMCID: PMC7983181 DOI: 10.3892/etm.2021.9897] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 11/26/2020] [Indexed: 01/10/2023] Open
Abstract
Obese asthma is a phenotype of asthma whose occurrence is gradually increasing in both adults and children. The majority of studies have demonstrated that obesity is a major risk factor for asthma and the effect of obesity on the lungs is considerable. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome has been previously demonstrated to serve a role in obese asthma mediated by mitochondrial reactive oxygen species (mtROS). The aim of the present in vitro study was to investigate the effect of leptin on airway epithelial cells and the protective effect of the mitochondrial-targeted antioxidant mitoquinone (mitoQ). Human normal bronchial epithelial cell lines BEAS-2 cells were used and divided into 6 groups: Control group (negative control), DMSO group (solvent control), lipopolysaccharide (LPS) group (positive control), LPS + mitoQ group, Leptin group and Leptin + mitoQ group. CCK8 assay was used to establish the optimal concentration and incubation time of the drugs. mitoTracker probe and mitoSOX reagent were used to detect the integrity of mitochondrial membranes and the content of mtROS. mRNA expression levels were detected by reverse transcription-quantitative PCR analysis. It was revealed that the mitochondrial membrane was disrupted in the Leptin group, which recovered after treatment with mitoQ. As a result, the production of mitochondrial reactive oxygen species (mtROS) in the Leptin group was significantly increased (P<0.01), but following treatment with mitoQ, this overproduction of mtROS was significantly decreased to normal levels (P<0.01). Furthermore, the expression levels of NOD-, LRR- and pyrin domain-containing protein 3 NLRP3 and caspase-1 mRNA in the leptin-pretreated BEAS-2 cells were significantly increased compared with those in the control group (P<0.01), while they were decreased following mitoQ treatment (P<0.01). Taken together, these data suggested that leptin may promote airway inflammation partially through upregulating the mtROS-NLRP3 inflammasome signaling pathway in airway epithelial cells and mitoQ may be a potential treatment for obese asthma.
Collapse
Affiliation(s)
- Lei Chong
- Institute of Pediatrics, National Key Clinical Specialty of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Haiyan Li
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lili Zhu
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Gang Yu
- Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
197
|
Patsalos O, Keeler J, Schmidt U, Penninx BWJH, Young AH, Himmerich H. Diet, Obesity, and Depression: A Systematic Review. J Pers Med 2021; 11:176. [PMID: 33802480 PMCID: PMC7999659 DOI: 10.3390/jpm11030176] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity and depression co-occur in a significant proportion of the population. Mechanisms linking the two disorders include the immune and the endocrine system, psychological and social mechanisms. The aim of this systematic review was to ascertain whether weight loss through dietary interventions has the additional effect of ameliorating depressive symptoms in obese patients. METHODS We systematically searched three databases (Pubmed, Medline, Embase) for longitudinal clinical trials testing a dietary intervention in people with obesity and depression or symptoms of depression. RESULTS Twenty-four longitudinal clinical studies met the eligibility criteria with a total of 3244 included patients. Seventeen studies examined the effects of calorie-restricted diets and eight studies examined dietary supplements (two studies examined both). Only three studies examined people with a diagnosis of both obesity and depression. The majority of studies showed that interventions using a calorie-restricted diet resulted in decreases in depression scores, with effect sizes between ≈0.2 and ≈0.6. The results were less clear for dietary supplements. CONCLUSIONS People with obesity and depression appear to be a specific subgroup of depressed patients in which calorie-restricted diets might constitute a promising personalized treatment approach. The reduction of depressive symptoms may be related to immunoendocrine and psychosocial mechanisms.
Collapse
Affiliation(s)
- Olivia Patsalos
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (O.P.); (J.K.); (U.S.); (A.H.Y.)
| | - Johanna Keeler
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (O.P.); (J.K.); (U.S.); (A.H.Y.)
| | - Ulrike Schmidt
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (O.P.); (J.K.); (U.S.); (A.H.Y.)
- South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Brenda W. J. H. Penninx
- Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, 1081 BT Amsterdam, The Netherlands;
| | - Allan H. Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (O.P.); (J.K.); (U.S.); (A.H.Y.)
- South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| | - Hubertus Himmerich
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK; (O.P.); (J.K.); (U.S.); (A.H.Y.)
- South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
| |
Collapse
|
198
|
Heyde I, Begemann K, Oster H. Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism. Endocrinology 2021; 162:6102571. [PMID: 33453099 PMCID: PMC7864004 DOI: 10.1210/endocr/bqab009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/17/2022]
Abstract
The term energy metabolism comprises the entirety of chemical processes associated with uptake, conversion, storage, and breakdown of nutrients. All these must be tightly regulated in time and space to ensure metabolic homeostasis in an environment characterized by cycles such as the succession of day and night. Most organisms evolved endogenous circadian clocks to achieve this goal. In mammals, a ubiquitous network of cellular clocks is coordinated by a pacemaker residing in the hypothalamic suprachiasmatic nucleus. Adipocytes harbor their own circadian clocks, and large aspects of adipose physiology are regulated in a circadian manner through transcriptional regulation of clock-controlled genes. White adipose tissue (WAT) stores energy in the form of triglycerides at times of high energy levels that then serve as fuel in times of need. It also functions as an endocrine organ, releasing factors in a circadian manner to regulate food intake and energy turnover in other tissues. Brown adipose tissue (BAT) produces heat through nonshivering thermogenesis, a process also controlled by the circadian clock. We here review how WAT and BAT contribute to the circadian regulation of energy metabolism. We describe how adipose rhythms are regulated by the interplay of systemic signals and local clocks and summarize how adipose-originating circadian factors feed-back on metabolic homeostasis. The role of adipose tissue in the circadian control of metabolism becomes increasingly clear as circadian disruption leads to alterations in adipose tissue regulation, promoting obesity and its sequelae. Stabilizing adipose tissue rhythms, in turn, may help to combat disrupted energy homeostasis and obesity.
Collapse
Affiliation(s)
- Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Correspondence: Henrik Oster, PhD, Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
199
|
Hillers-Ziemer LE, Williams AE, Janquart A, Grogan C, Thompson V, Sanchez A, Arendt LM. Obesity-Activated Lung Stromal Cells Promote Myeloid Lineage Cell Accumulation and Breast Cancer Metastasis. Cancers (Basel) 2021; 13:1005. [PMID: 33670906 PMCID: PMC7957630 DOI: 10.3390/cancers13051005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is correlated with increased incidence of breast cancer metastasis; however, the mechanisms underlying how obesity promotes metastasis are unclear. In a diet-induced obese mouse model, obesity enhanced lung metastasis in both the presence and absence of primary mammary tumors and increased recruitment of myeloid lineage cells into the lungs. In the absence of tumors, obese mice demonstrated increased numbers of myeloid lineage cells and elevated collagen fibers within the lung stroma, reminiscent of premetastatic niches formed by primary tumors. Lung stromal cells isolated from obese tumor-naïve mice showed increased proliferation, contractility, and expression of extracellular matrix, inflammatory markers and transforming growth factor beta-1 (TGFβ1). Conditioned media from lung stromal cells from obese mice promoted myeloid lineage cell migration in vitro in response to colony-stimulating factor 2 (CSF2) expression and enhanced invasion of tumor cells. Together, these results suggest that prior to tumor formation, obesity alters the lung microenvironment, creating niches conducive to metastatic growth.
Collapse
Affiliation(s)
- Lauren E. Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Abbey E. Williams
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Amanda Janquart
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Caitlin Grogan
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Victoria Thompson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Adriana Sanchez
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| | - Lisa M. Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Program in Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (A.J.); (C.G.); (V.T.); (A.S.)
| |
Collapse
|
200
|
Saadi S, Ghazali HM, Saari N, Abdulkarim SM. The structural reconformation of peptides in enhancing functional and therapeutic properties: Insights into their solid state crystallizations. Biophys Chem 2021; 273:106565. [PMID: 33780688 DOI: 10.1016/j.bpc.2021.106565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 01/05/2023]
Abstract
Therapeutic peptides derived proteins with alpha-reconformation states like antibody shape have shown potential effects in combating terrible diseases linked with earlier signs of angiogensis, mutagenesis and transgenesis. Alpha reconformation in material design refers to the folding of the peptide chains and their transitions under reversible chemical bonds of disulfide chemical bridges and further non-covalence lesions. Thus, the rational design of signal peptides into alpha-helix is intended in increasing the defending effects of peptides into cores like adjuvant antibiotic and/or vaccines. Thereby, the signal peptides are able in displaying multiple eradicating regions by changing crystal-depositions and deviation angles. These types of molecular structures could have multiple advantages in tracing disease syndromes and impurities by increasing the host defense against the fates of pathogens and viruses, eventually leading to the loss in signaling by increasing peptide susceptibility levels to folding and unfolding and therefore, formation of transgenic peptide models. Alpha reconformation peptides is aimed in triggering as well as other regulatory functions such as remodulating metabolic chain disorders of lipolysis and glucolysis by increasing the insulin and leptin resistance for best lipid storages and lipoprotein density distributions.
Collapse
Affiliation(s)
- Sami Saadi
- Institut de la Nutrition, de l'Alimentation et des Technologies Agro-alimentaires INATAA 25017, Université Frères Mentouri, Constantine 1, Algeria; Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Hasanah Mohd Ghazali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Nazamid Saari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Sabo Mohammed Abdulkarim
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|