1
|
Affuso F, Micillo F, Fazio S. Insulin Resistance, a Risk Factor for Alzheimer's Disease: Pathological Mechanisms and a New Proposal for a Preventive Therapeutic Approach. Biomedicines 2024; 12:1888. [PMID: 39200352 PMCID: PMC11351221 DOI: 10.3390/biomedicines12081888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Peripheral insulin resistance (IR) is a well-documented, independent risk factor for the development of type 2 diabetes, cardiovascular disease, cancer and cellular senescence. Recently, the brain has also been identified as an insulin-responsive region, where insulin acts as regulator of the brain metabolism. Despite the clear link between IR and the brain, the exact mechanisms underlying this relationship remain unclear. Therapeutic intervention in patients showing symptoms of neurodegenerative diseases has produced little or no results. It has been demonstrated that insulin resistance plays a significant role in the pathogenesis of neurodegenerative diseases, particularly cognitive decline. Peripheral and brain IR may represent a modifiable state that could be used to prevent major brain disorders. In this review, we will analyse the scientific literature supporting IR as a risk factor for Alzheimer's disease and suggest some therapeutic strategies to provide a new proposal for the prevention of brain IR and its consequences.
Collapse
Affiliation(s)
- Flora Affuso
- Independent Researcher, Viale Raffaello, 74, 80129 Napoli, Italy
| | - Filomena Micillo
- UOC of Geriatric Medicine AORN S.G. Moscati, 83100 Avellino, Italy
| | - Serafino Fazio
- Department of Internal Medicine, School of Medicine, Federico II University of Naples, 80138 Naples, Italy;
| |
Collapse
|
2
|
Senthilkumaran M, Koch C, Herselman MF, Bobrovskaya L. Role of the Adrenal Medulla in Hypoglycaemia-Associated Autonomic Failure-A Diabetic Perspective. Metabolites 2024; 14:100. [PMID: 38392992 PMCID: PMC10890365 DOI: 10.3390/metabo14020100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Hypoglycaemia-associated autonomic failure (HAAF) is characterised by an impairment in adrenal medullary and neurogenic symptom responses following episodes of recurrent hypoglycaemia. Here, we review the status quo of research related to the regulatory mechanisms of the adrenal medulla in its response to single and recurrent hypoglycaemia in both diabetic and non-diabetic subjects with particular focus given to catecholamine synthesis, enzymatic activity, and the impact of adrenal medullary peptides. Short-term post-transcriptional modifications, particularly phosphorylation at specific residues of tyrosine hydroxylase (TH), play a key role in the regulation of catecholamine synthesis. While the effects of recurrent hypoglycaemia on catecholamine synthetic enzymes remain inconsistent, long-term changes in TH protein expression suggest species-specific responses. Adrenomedullary peptides such as neuropeptide Y (NPY), galanin, and proenkephalin exhibit altered gene and protein expression in response to hypoglycaemia, suggesting a potential role in the modulation of catecholamine secretion. Of note is NPY, since its antagonism has been shown to prevent reductions in TH protein expression. This review highlights the need for further investigation into the molecular mechanisms involved in the adrenal medullary response to hypoglycaemia. Despite advancements in our understanding of HAAF in non-diabetic rodents, a reliable diabetic rodent model of HAAF remains a challenge.
Collapse
Affiliation(s)
- Manjula Senthilkumaran
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Coen Koch
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Mauritz Frederick Herselman
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
3
|
Zhang Y, Qin C, Wang J, Yang L, Yan X, Zhi S, Nie G. Phosphofructokinase family genes in grass carp: Molecular identification and tissue-specific expression in response to glucose, insulin and glucagon. Comp Biochem Physiol B Biochem Mol Biol 2024; 269:110898. [PMID: 37673204 DOI: 10.1016/j.cbpb.2023.110898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/02/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
It is widely acknowledged that glucose serves as the primary energy source for organisms. However, fish exhibit persistent postprandial hyperglycemia and are thought to have low glucose tolerance. Glycolysis serves as the ubiquitous pathway for glucose catabolism, with phosphofructokinase (PFK) acting as a crucial rate-limiting enzyme in this process and playing an indispensable role. PFK is classified into three isoforms based on their major expression sites, i.e., PFKM (skeletal muscle type), PFKL (liver type) and PFKP (platelet type). In this study, grass carp (Ctenopharyngodon idella) was used as animal model and the open reading frame (ORF) sequences of six PFK genetic isoforms of grass carp were cloned. Real-time PCR was used to detect its tissue distribution, and expression changes in oral glucose tolerance test (OGTT), insulin and glucagon injection experiments. The results showed that the ORF of pfkla, pfklb, pfkma, pfkmb, pfkpa and pfkpb genes was 2343, 2340, 2355, 2331, 2364 and 2349 bp in length, respectively. The results of tissue distribution showed that pfkla and pfklb, homologous to mammalian pfkl, exhibited low expression levels in the liver of grass carp, but were expressed at the highest level in the brain. Muscle-type pfkma and pfkmb mRNA were found to be highly expressed in both red and white muscle, with pfkmb also exhibiting high expression levels in the heart, while platelet type pfkpa and pfkpb showed high mRNA abundances in the brain and heart. Oral glucose administration stimulated pfkma and pfkmb mRNA expression in the red muscle, and up-regulated pfklb mRNA levels in the liver at 3 h post treatment, but it suppressed liver-type and platelet-type PFK genes expression in the brain. The expression of pfkmb in white muscle and pfkmb and pfkpb in heart were promoted by insulin, whereas the expression of pfkla and pfkpb in the brain, pfkma and pfkmb in the red muscle, pfkma in the white muscle, and pfklb in the liver was suppressed by insulin. As for glucagon, it inhibited pfkma and pfkmb mRNA expression in the red muscle, as well as pfklb in the liver, but it up-regulated PFK genes expression in most tissues detected, such as brain (pfklb, pfkpa and pfkpb), white muscle (pfkma and pfkmb), liver (pfkla) and heart (pfkmb and pfkpb). Our results suggest that PFK family genes have different or even opposite expression patterns in response to glucose, insulin and glucagon stimulation in various tissues of grass carp, which may contribute to glucose intolerance in fish.
Collapse
Affiliation(s)
- Yingxin Zhang
- College of Life Science, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China.
| | - Chaobin Qin
- College of Fisheries, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China.
| | - Junli Wang
- College of Life Science, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China
| | - Liping Yang
- College of Fisheries, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China
| | - Xiao Yan
- College of Fisheries, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China
| | - Shaoyang Zhi
- College of Fisheries, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China
| | - Guoxing Nie
- College of Fisheries, Henan Normal University, No. 46 Jianshe Road, Xinxiang 453007, PR China.
| |
Collapse
|
4
|
Ye L, Ding W, Xiao D, Jia Y, Zhao Z, Ao X, Wang J. O-GlcNAcylation: cellular physiology and therapeutic target for human diseases. MedComm (Beijing) 2023; 4:e456. [PMID: 38116061 PMCID: PMC10728774 DOI: 10.1002/mco2.456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
O-linked-β-N-acetylglucosamine (O-GlcNAcylation) is a distinctive posttranslational protein modification involving the coordinated action of O-GlcNAc transferase and O-GlcNAcase, primarily targeting serine or threonine residues in various proteins. This modification impacts protein functionality, influencing stability, protein-protein interactions, and localization. Its interaction with other modifications such as phosphorylation and ubiquitination is becoming increasingly evident. Dysregulation of O-GlcNAcylation is associated with numerous human diseases, including diabetes, nervous system degeneration, and cancers. This review extensively explores the regulatory mechanisms of O-GlcNAcylation, its effects on cellular physiology, and its role in the pathogenesis of diseases. It examines the implications of aberrant O-GlcNAcylation in diabetes and tumorigenesis, highlighting novel insights into its potential role in cardiovascular diseases. The review also discusses the interplay of O-GlcNAcylation with other protein modifications and its impact on cell growth and metabolism. By synthesizing current research, this review elucidates the multifaceted roles of O-GlcNAcylation, providing a comprehensive reference for future studies. It underscores the potential of targeting the O-GlcNAcylation cycle in developing novel therapeutic strategies for various pathologies.
Collapse
Affiliation(s)
- Lin Ye
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Wei Ding
- The Affiliated Hospital of Qingdao UniversityQingdao Medical CollegeQingdao UniversityQingdaoChina
| | - Dandan Xiao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Yi Jia
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Zhonghao Zhao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Xiang Ao
- School of Basic MedicineQingdao UniversityQingdaoChina
| | - Jianxun Wang
- School of Basic MedicineQingdao UniversityQingdaoChina
| |
Collapse
|
5
|
Zhao D, Meng Y, Dian Y, Zhou Q, Sun Y, Le J, Zeng F, Chen X, He Y, Deng G. Molecular landmarks of tumor disulfidptosis across cancer types to promote disulfidptosis-target therapy. Redox Biol 2023; 68:102966. [PMID: 38035663 PMCID: PMC10698012 DOI: 10.1016/j.redox.2023.102966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023] Open
Abstract
The mystery about the mechanistic basis of disulfidptosis has recently been unraveled and shows promise as an effective treatment modality for triggering cancer cell death. However, the limited understanding of the role of disulfidptosis in tumor progression and drug sensitivity has hindered the development of disulfidptosis-targeted therapy and combinations with other therapeutic strategies. Here, we established a disulfidptosis signature model to estimate tumor disulfidptosis status in approximately 10,000 tumor samples across 33 cancer types and revealed its prognostic value. Then, we characterized disulfidptosis-associated molecular features and identified various types of molecular alterations that correlate with both drug-resistant and drug-sensitive responses to anti-tumor drugs. We further showed the vast heterogeneity in disulfidptosis status among 760 cancer cell lines across 25 cancer types. We experimentally validated that disulfidptosis score-high cell lines are more susceptible to glucose starvation-induced disulfidptosis compared to their counterparts with low scores. Finally, we investigated the impact of disulfidptosis status on drug response and revealed that disulfidptosis induction may enhance sensitivity to anti-cancer drugs, but in some cases, it could also lead to drug resistance in cultured cells. Overall, our multi-omics analysis firstly elucidates a comprehensive profile of disulfidptosis-related molecular alterations, prognosis, and potential therapeutic therapies at a pan-cancer level. These findings may uncover opportunities to utilize multiple drug sensitivities induced by disulfidptosis, thereby offering practical implications for clinical cancer therapy.
Collapse
Affiliation(s)
- Deze Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan 410008, China; Furong Laboratory, Changsha, Hunan 410008, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
6
|
Lee HG, Jung IH, Park BS, Yang HR, Kim KK, Tu TH, Yeh JY, Lee S, Yang S, Lee BJ, Kim JG, Nam-Goong IS. Altered Metabolic Phenotypes and Hypothalamic Neuronal Activity Triggered by Sodium-Glucose Cotransporter 2 Inhibition. Diabetes Metab J 2023; 47:784-795. [PMID: 37915185 PMCID: PMC10695712 DOI: 10.4093/dmj.2022.0261] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/17/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGRUOUND Sodium-glucose cotransporter 2 (SGLT-2) inhibitors are currently used to treat patients with diabetes. Previous studies have demonstrated that treatment with SGLT-2 inhibitors is accompanied by altered metabolic phenotypes. However, it has not been investigated whether the hypothalamic circuit participates in the development of the compensatory metabolic phenotypes triggered by the treatment with SGLT-2 inhibitors. METHODS Mice were fed a standard diet or high-fat diet and treated with dapagliflozin, an SGLT-2 inhibitor. Food intake and energy expenditure were observed using indirect calorimetry system. The activity of hypothalamic neurons in response to dapagliflozin treatment was evaluated by immunohistochemistry with c-Fos antibody. Quantitative real-time polymerase chain reaction was performed to determine gene expression patterns in the hypothalamus of dapagliflozin-treated mice. RESULTS Dapagliflozin-treated mice displayed enhanced food intake and reduced energy expenditure. Altered neuronal activities were observed in multiple hypothalamic nuclei in association with appetite regulation. Additionally, we found elevated immunosignals of agouti-related peptide neurons in the paraventricular nucleus of the hypothalamus. CONCLUSION This study suggests the functional involvement of the hypothalamus in the development of the compensatory metabolic phenotypes induced by SGLT-2 inhibitor treatment.
Collapse
Affiliation(s)
- Ho Gyun Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Il Hyeon Jung
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Byong Seo Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Hye Rim Yang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Kwang Kon Kim
- Department of Biological Science, University of Ulsan, Ulsan, Korea
| | - Thai Hien Tu
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Jung-Yong Yeh
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
| | - Sewon Lee
- Division of Sport Science, College of Arts & Physical Education, Incheon National University, Incheon, Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
| | - Sunggu Yang
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
- Department of Nano-Bioengineering, College of Life Science and Technology, Incheon National University, Incheon, Korea
| | - Byung Ju Lee
- Department of Biological Science, University of Ulsan, Ulsan, Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea
- Research Center of Brain-Machine Interface, Incheon National University, Incheon, Korea
| | - Il Seong Nam-Goong
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| |
Collapse
|
7
|
Carrasco RA, Breen KM. Allostasis in Neuroendocrine Systems Controlling Reproduction. Endocrinology 2023; 164:bqad125. [PMID: 37586095 PMCID: PMC10461221 DOI: 10.1210/endocr/bqad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
Allostasis provides a supporting role to the homeostatic control of biological variables in mammalian species. While the concept of homeostasis is related to the control of variables within a set point or range that are essential to life, allostasis refers to systems that facilitate adaptation to challenges that the organism faces and the new requirements for survival. Essential for such adaptation is the role played by the brain in eliciting neural and neuroendocrine responses. Reproductive function is fundamental for the survival of species but is costly in energetic terms and requires a synchrony with an ever-changing environment. Thus, in many species reproductive function is blocked or delayed over immediate challenges. This review will cover the physiological systems and neuroendocrine pathways that supply allostatic control over reproductive neuroendocrine systems. Light, hypoxia, temperature, nutrition, psychosocial, and immune mediators influence the neuroendocrine control of reproductive functions through pathways that are confluent at the paraventricular nucleus; however, understanding of the integrative responses to these stimuli has not been clarified. Likely, the ultimate consequence of these allostatic mechanisms is the modification of kisspeptin and gonadotropin-releasing hormone neuronal activity, thus compromising reproduction function in the short term, while preserving species survivability.
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| | - Kellie M Breen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093-0674, USA
| |
Collapse
|
8
|
Raut S, Bhalerao A, Powers M, Gonzalez M, Mancuso S, Cucullo L. Hypometabolism, Alzheimer's Disease, and Possible Therapeutic Targets: An Overview. Cells 2023; 12:2019. [PMID: 37626828 PMCID: PMC10453773 DOI: 10.3390/cells12162019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
The brain is a highly dynamic organ that requires a constant energy source to function normally. This energy is mostly supplied by glucose, a simple sugar that serves as the brain's principal fuel source. Glucose transport across the blood-brain barrier (BBB) is primarily controlled via sodium-independent facilitated glucose transport, such as by glucose transporter 1 (GLUT1) and 3 (GLUT3). However, other glucose transporters, including GLUT4 and the sodium-dependent transporters SGLT1 and SGLT6, have been reported in vitro and in vivo. When the BBB endothelial layer is crossed, neurons and astrocytes can absorb the glucose using their GLUT1 and GLUT3 transporters. Glucose then enters the glycolytic pathway and is metabolized into adenosine triphosphate (ATP), which supplies the energy to support cellular functions. The transport and metabolism of glucose in the brain are impacted by several medical conditions, which can cause neurological and neuropsychiatric symptoms. Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, traumatic brain injury (TBI), schizophrenia, etc., are a few of the most prevalent disorders, characterized by a decline in brain metabolism or hypometabolism early in the course of the disease. Indeed, AD is considered a metabolic disorder related to decreased brain glucose metabolism, involving brain insulin resistance and age-dependent mitochondrial dysfunction. Although the conventional view is that reduced cerebral metabolism is an effect of neuronal loss and consequent brain atrophy, a growing body of evidence points to the opposite, where hypometabolism is prodromal or at least precedes the onset of brain atrophy and the manifestation of clinical symptoms. The underlying processes responsible for these glucose transport and metabolic abnormalities are complicated and remain poorly understood. This review article provides a comprehensive overview of the current understanding of hypometabolism in AD and potential therapeutic targets.
Collapse
Affiliation(s)
- Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Michael Powers
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA;
| | - Minelly Gonzalez
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Salvatore Mancuso
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| |
Collapse
|
9
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
10
|
Huang Y, Wang JB, Parker JJ, Shivacharan R, Lal RA, Halpern CH. Spectro-spatial features in distributed human intracranial activity proactively encode peripheral metabolic activity. Nat Commun 2023; 14:2729. [PMID: 37169738 PMCID: PMC10174612 DOI: 10.1038/s41467-023-38253-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Mounting evidence demonstrates that the central nervous system (CNS) orchestrates glucose homeostasis by sensing glucose and modulating peripheral metabolism. Glucose responsive neuronal populations have been identified in the hypothalamus and several corticolimbic regions. However, how these CNS gluco-regulatory regions modulate peripheral glucose levels is not well understood. To better understand this process, we simultaneously measured interstitial glucose concentrations and local field potentials in 3 human subjects from cortical and subcortical regions, including the hypothalamus in one subject. Correlations between high frequency activity (HFA, 70-170 Hz) and peripheral glucose levels are found across multiple brain regions, notably in the hypothalamus, with correlation magnitude modulated by sleep-wake cycles, circadian coupling, and hypothalamic connectivity. Correlations are further present between non-circadian (ultradian) HFA and glucose levels which are higher during awake periods. Spectro-spatial features of neural activity enable decoding of peripheral glucose levels both in the present and up to hours in the future. Our findings demonstrate proactive encoding of homeostatic glucose dynamics by the CNS.
Collapse
Affiliation(s)
- Yuhao Huang
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Jeffrey B Wang
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, 94305, USA
- Medical Scientist Training Program, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Jonathon J Parker
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Rajat Shivacharan
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Rayhan A Lal
- Department of Medicine (Endocrinology), Stanford University Medical Center, Stanford, CA, 94305, USA.
- Department of Pediatrics (Endocrinology), Stanford University Medical Center, Stanford, CA, 94305, USA.
| | - Casey H Halpern
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA, 94305, USA.
| |
Collapse
|
11
|
Alonge KM, Porte D, Schwartz MW. Distinct Roles for Brain and Pancreas in Basal and Postprandial Glucose Homeostasis. Diabetes 2023; 72:547-556. [PMID: 37146276 PMCID: PMC10130484 DOI: 10.2337/db22-0969] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/04/2023] [Indexed: 05/07/2023]
Abstract
The glucose homeostasis system ensures that the circulating glucose level is maintained within narrow physiological limits both in the fasting (or basal) state and following a nutrient challenge. Although glucose homeostasis is traditionally conceptualized as a single overarching system, evidence reviewed here suggests that basal glycemia and glucose tolerance are governed by distinct control systems. Specifically, whereas glucose tolerance appears to be determined largely by interactions between insulin secretion and insulin sensitivity, basal-state glucose homeostasis is predominated by insulin-independent mechanisms governed largely by the brain. In addition to a new perspective on how glucose homeostasis is achieved, this "dual control system" hypothesis offers a feasible and testable explanation for observations that are otherwise difficult to reconcile and sheds new light on the integration of central and peripheral metabolic control mechanisms. The implications of this model for the pathogenesis and treatment of impaired fasting glucose, impaired glucose tolerance, and type 2 diabetes are also discussed.
Collapse
Affiliation(s)
- Kimberly M. Alonge
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| | - Daniel Porte
- Division of Endocrinology, School of Medicine, University of California San Diego, San Diego, CA
| | - Michael W. Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| |
Collapse
|
12
|
Schwartz MW, Krinsley JS, Faber CL, Hirsch IB, Brownlee M. Brain Glucose Sensing and the Problem of Relative Hypoglycemia. Diabetes Care 2023; 46:237-244. [PMID: 36701597 PMCID: PMC9887623 DOI: 10.2337/dc22-1445] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/22/2022] [Indexed: 01/27/2023]
Abstract
"Relative hypoglycemia" is an often-overlooked complication of diabetes characterized by an increase in the glycemic threshold for detecting and responding to hypoglycemia. The clinical relevance of this problem is linked to growing evidence that among patients with critical illness, higher blood glucose in the intensive care unit is associated with higher mortality among patients without diabetes but lower mortality in patients with preexisting diabetes and an elevated prehospitalization HbA1c. Although additional studies are needed, the cardiovascular stress associated with hypoglycemia perception, which can occur at normal or even elevated glucose levels in patients with diabetes, offers a plausible explanation for this difference in outcomes. Little is known, however, regarding how hypoglycemia is normally detected by the brain, much less how relative hypoglycemia develops in patients with diabetes. In this article, we explore the role in hypoglycemia detection played by glucose-responsive sensory neurons supplying peripheral vascular beds and/or circumventricular organs. These observations support a model wherein relative hypoglycemia results from diabetes-associated impairment of this neuronal glucose-sensing process. By raising the glycemic threshold for hypoglycemia perception, this impairment may contribute to the increased mortality risk associated with standard glycemic management of critically ill patients with diabetes.
Collapse
Affiliation(s)
- Michael W. Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| | - James S. Krinsley
- Stamford Hospital, Stamford, CT
- Columbia Vagelos College of Physicians and Surgeons, New York, NY
| | - Chelsea L. Faber
- Ivy Brain Tumor Center, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ
| | - Irl B. Hirsch
- Department of Medicine, University of Washington Medicine Diabetes Institute, Seattle, WA
| | - Michael Brownlee
- Einstein Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
13
|
Drosophila suzukii energetic pathways are differently modulated by nutritional geometry in males and females. Sci Rep 2022; 12:21194. [PMID: 36476948 PMCID: PMC9729594 DOI: 10.1038/s41598-022-25509-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
As a polyphagous pest, Drosophila suzukii has a variety of host fruits available for feeding and oviposition, but how the nutritional geometry of different hosts influences its metabolism is still poorly understood. This work aimed to evaluate how D. suzukii metabolic and transcriptional pathways are influenced by feeding on different host fruits, and how sex influences these responses. Adult flies were allowed to feed on five different fruit-based media. Lipids, glucose, glycogen, and energy pathways-associated gene expression, were quantified. Females showed an energetic metabolism easily adaptable to the food's nutritional characteristics; in contrast, males' energetic metabolism was particularly influenced by food, predominantly those fed on raspberry media who showed changes in glucose levels and in the expression of genes associated with metabolic pathways, suggesting activation of gluconeogenesis and trehaloneogenesis as a result of nutritional deficiency. Here we present novel insight into how D. suzukii's energetic pathways are modulated depending on fruits' nutritional geometry and sex. While the females showed high adaptability in their energetic metabolism to the diet, males were more feeding-sensitive. These findings might be used not only to control this pest population but to better advise producers to invest in less suitable fruits based on the hosts' nutritional geometry.
Collapse
|
14
|
Lv X, Gao F, Cao X. Skeletal interoception in bone homeostasis and pain. Cell Metab 2022; 34:1914-1931. [PMID: 36257317 PMCID: PMC9742337 DOI: 10.1016/j.cmet.2022.09.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Accumulating evidence indicates that interoception maintains proper physiological status and orchestrates metabolic homeostasis by regulating feeding behaviors, glucose balance, and lipid metabolism. Continuous skeletal remodeling consumes a tremendous amount of energy to provide skeletal scaffolding, support muscle movement, store vital minerals, and maintain a niche for hematopoiesis, which are processes that also contribute to overall metabolic balance. Although skeletal innervation has been described for centuries, recent work has shown that skeletal metabolism is tightly regulated by the nervous system and that skeletal interoception regulates bone homeostasis. Here, we provide a general discussion of interoception and its effects on the skeleton and whole-body metabolism. We also discuss skeletal interoception-mediated regulation in the context of pathological conditions and skeletal pain as well as future challenges to our understanding of these process and how they can be leveraged for more effective therapy.
Collapse
Affiliation(s)
- Xiao Lv
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Feng Gao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xu Cao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Gasser E, Sancar G, Downes M, Evans RM. Metabolic Messengers: fibroblast growth factor 1. Nat Metab 2022; 4:663-671. [PMID: 35681108 PMCID: PMC9624216 DOI: 10.1038/s42255-022-00580-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022]
Abstract
While fibroblast growth factor (FGF) 1 is expressed in multiple tissues, only adipose-derived and brain FGF1 have been implicated in the regulation of metabolism. Adipose FGF1 production is upregulated in response to dietary stress and is essential for adipose tissue plasticity in these conditions. Similarly, in the brain, FGF1 secretion into the ventricular space and the adjacent parenchyma is increased after a hypercaloric challenge induced by either feeding or glucose infusion. Potent anorexigenic properties have been ascribed to both peripheral and centrally injected FGF1. The ability of recombinant FGF1 and variants with reduced mitogenicity to lower glucose, suppress adipose lipolysis and promote insulin sensitization elevates their potential as candidates in the treatment of type 2 diabetes mellitus and associated comorbidities. Here, we provide an overview of the known metabolic functions of endogenous FGF1 and discuss its therapeutic potential, distinguishing between peripherally or centrally administered FGF1.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Gencer Sancar
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
16
|
Tu L, Fukuda M, Tong Q, Xu Y. The ventromedial hypothalamic nucleus: watchdog of whole-body glucose homeostasis. Cell Biosci 2022; 12:71. [PMID: 35619170 PMCID: PMC9134642 DOI: 10.1186/s13578-022-00799-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
The brain, particularly the ventromedial hypothalamic nucleus (VMH), has been long known for its involvement in glucose sensing and whole-body glucose homeostasis. However, it is still not fully understood how the brain detects and responds to the changes in the circulating glucose levels, as well as brain-body coordinated control of glucose homeostasis. In this review, we address the growing evidence implicating the brain in glucose homeostasis, especially in the contexts of hypoglycemia and diabetes. In addition to neurons, we emphasize the potential roles played by non-neuronal cells, as well as extracellular matrix in the hypothalamus in whole-body glucose homeostasis. Further, we review the ionic mechanisms by which glucose-sensing neurons sense fluctuations of ambient glucose levels. We also introduce the significant implications of heterogeneous neurons in the VMH upon glucose sensing and whole-body glucose homeostasis, in which sex difference is also addressed. Meanwhile, research gaps have also been identified, which necessities further mechanistic studies in future.
Collapse
Affiliation(s)
- Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Makoto Fukuda
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Caretta A, Mucignat-Caretta C. Not Only COVID-19: Involvement of Multiple Chemosensory Systems in Human Diseases. Front Neural Circuits 2022; 16:862005. [PMID: 35547642 PMCID: PMC9081982 DOI: 10.3389/fncir.2022.862005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Chemosensory systems are deemed marginal in human pathology. In appraising their role, we aim at suggesting a paradigm shift based on the available clinical and experimental data that will be discussed. Taste and olfaction are polymodal sensory systems, providing inputs to many brain structures that regulate crucial visceral functions, including metabolism but also endocrine, cardiovascular, respiratory, and immune systems. Moreover, other visceral chemosensory systems monitor different essential chemical parameters of “milieu intérieur,” transmitting their data to the brain areas receiving taste and olfactory inputs; hence, they participate in regulating the same vital functions. These chemosensory cells share many molecular features with olfactory or taste receptor cells, thus they may be affected by the same pathological events. In most COVID-19 patients, taste and olfaction are disturbed. This may represent only a small portion of a broadly diffuse chemosensory incapacitation. Indeed, many COVID-19 peculiar symptoms may be explained by the impairment of visceral chemosensory systems, for example, silent hypoxia, diarrhea, and the “cytokine storm”. Dysregulation of chemosensory systems may underlie the much higher mortality rate of COVID-19 Acute Respiratory Distress Syndrome (ARDS) compared to ARDSs of different origins. In chronic non-infectious diseases like hypertension, diabetes, or cancer, the impairment of taste and/or olfaction has been consistently reported. This may signal diffuse chemosensory failure, possibly worsening the prognosis of these patients. Incapacitation of one or few chemosensory systems has negligible effects on survival under ordinary life conditions but, under stress, like metabolic imbalance or COVID-19 pneumonia, the impairment of multiple chemosensory systems may lead to dire consequences during the course of the disease.
Collapse
Affiliation(s)
- Antonio Caretta
- National Institute for Biostructures and Biosystems (NIBB), Rome, Italy
- Department of Food and Drug Science, University of Parma, Parma, Italy
| | - Carla Mucignat-Caretta
- National Institute for Biostructures and Biosystems (NIBB), Rome, Italy
- Department of Molecular Medicine, University of Padova, Padua, Italy
- *Correspondence: Carla Mucignat-Caretta,
| |
Collapse
|
18
|
Nutrient Sensing via Gut in Drosophila melanogaster. Int J Mol Sci 2022; 23:ijms23052694. [PMID: 35269834 PMCID: PMC8910450 DOI: 10.3390/ijms23052694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Nutrient-sensing mechanisms in animals' sense available nutrients to generate a physiological regulatory response involving absorption, digestion, and regulation of food intake and to maintain glucose and energy homeostasis. During nutrient sensing via the gastrointestinal tract, nutrients interact with receptors on the enteroendocrine cells in the gut, which in return respond by secreting various hormones. Sensing of nutrients by the gut plays a critical role in transmitting food-related signals to the brain and other tissues informing the composition of ingested food to digestive processes. These signals modulate feeding behaviors, food intake, metabolism, insulin secretion, and energy balance. The increasing significance of fly genetics with the availability of a vast toolbox for studying physiological function, expression of chemosensory receptors, and monitoring the gene expression in specific cells of the intestine makes the fly gut the most useful tissue for studying the nutrient-sensing mechanisms. In this review, we emphasize on the role of Drosophila gut in nutrient-sensing to maintain metabolic homeostasis and gut-brain cross talk using endocrine and neuronal signaling pathways stimulated by internal state or the consumption of various dietary nutrients. Overall, this review will be useful in understanding the post-ingestive nutrient-sensing mechanisms having a physiological and pathological impact on health and diseases.
Collapse
|
19
|
Boland BB, Laker RC, O'Brien S, Sitaula S, Sermadiras I, Nielsen JC, Barkholt P, Roostalu U, Hecksher-Sørensen J, Sejthen SR, Thorbek DD, Suckow A, Burmeister N, Oldham S, Will S, Howard VG, Gill BM, Newton P, Naylor J, Hornigold DC, Austin J, Lantier L, McGuinness OP, Trevaskis JL, Grimsby JS, Rhodes CJ. Peptide-YY 3-36/glucagon-like peptide-1 combination treatment of obese diabetic mice improves insulin sensitivity associated with recovered pancreatic β-cell function and synergistic activation of discrete hypothalamic and brainstem neuronal circuitries. Mol Metab 2021; 55:101392. [PMID: 34781035 PMCID: PMC8717237 DOI: 10.1016/j.molmet.2021.101392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/22/2021] [Accepted: 11/04/2021] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Obesity-linked type 2 diabetes (T2D) is a worldwide health concern and many novel approaches are being considered for its treatment and subsequent prevention of serious comorbidities. Co-administration of glucagon like peptide 1 (Fc-GLP-1) and peptide YY3-36 (Fc-PYY3-36) renders a synergistic decrease in energy intake in obese men. However, mechanistic details of the synergy between these peptide agonists and their effects on metabolic homeostasis remain relatively scarce. METHODS In this study, we utilized long-acting analogues of GLP-1 and PYY3-36 (via Fc-peptide conjugation) to better characterize the synergistic pharmacological benefits of their co-administration on body weight and glycaemic regulation in obese and diabetic mouse models. Hyperinsulinemic-euglycemic clamps were used to measure weight-independent effects of Fc-PYY3-36 + Fc-GLP-1 on insulin action. Fluorescent light sheet microscopy analysis of whole brain was performed to assess activation of brain regions. RESULTS Co-administration of long-acting Fc-IgG/peptide conjugates of Fc-GLP-1 and Fc-PYY3-36 (specific for PYY receptor-2 (Y2R)) resulted in profound weight loss, restored glucose homeostasis, and recovered endogenous β-cell function in two mouse models of obese T2D. Hyperinsulinemic-euglycemic clamps in C57BLKS/J db/db and diet-induced obese Y2R-deficient (Y2RKO) mice indicated Y2R is required for a weight-independent improvement in peripheral insulin sensitivity and enhanced hepatic glycogenesis. Brain cFos staining demonstrated distinct temporal activation of regions of the hypothalamus and hindbrain following Fc-PYY3-36 + Fc-GLP-1R agonist administration. CONCLUSIONS These results reveal a therapeutic approach for obesity/T2D that improved insulin sensitivity and restored endogenous β-cell function. These data also highlight the potential association between the gut-brain axis in control of metabolic homeostasis.
Collapse
Affiliation(s)
- Brandon B Boland
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Gubra ApS, Horsholm, Denmark; PRECISIONscientia, Yardley, PA, USA
| | - Rhianna C Laker
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Siobhan O'Brien
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sadichha Sitaula
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Isabelle Sermadiras
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | | | | | | | | | - Arthur Suckow
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; DTX Pharma, San Diego, CA, USA
| | - Nicole Burmeister
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Roche, Penzberg, Germany
| | - Stephanie Oldham
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sarah Will
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Victor G Howard
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benji M Gill
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Philip Newton
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jacqueline Naylor
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David C Hornigold
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jotham Austin
- University of Chicago Advanced Electron Microscopy Core Facility, Chicago, IL, USA
| | - Louise Lantier
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - James L Trevaskis
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Gilead Sciences, Foster City, CA, USA
| | - Joseph S Grimsby
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
20
|
Kessler S, Labouèbe G, Croizier S, Gaspari S, Tarussio D, Thorens B. Glucokinase neurons of the paraventricular nucleus of the thalamus sense glucose and decrease food consumption. iScience 2021; 24:103122. [PMID: 34622169 PMCID: PMC8481977 DOI: 10.1016/j.isci.2021.103122] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/08/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
The paraventricular nucleus of the thalamus (PVT) controls goal-oriented behavior through its connections to the nucleus accumbens (NAc). We previously characterized Glut2aPVT neurons that are activated by hypoglycemia, and which increase sucrose seeking behavior through their glutamatergic projections to the NAc. Here, we identified glucokinase (Gck)-expressing neurons of the PVT (GckaPVT) and generated a mouse line expressing the Cre recombinase from the glucokinase locus (Gck Cre/+ mice). Ex vivo calcium imaging and whole-cell patch clamp recordings revealed that GckaPVT neurons that project to the NAc were mostly activated by hyperglycemia. Their chemogenetic inhibition or optogenetic stimulation, respectively, enhanced food intake or decreased sucrose-seeking behavior. Collectively, our results describe a neuronal population of Gck-expressing neurons in the PVT, which has opposite glucose sensing properties and control over feeding behavior than the previously characterized Glut2aPVT neurons. This study allows a better understanding of the complex regulation of feeding behavior by the PVT.
Collapse
Affiliation(s)
- Sébastien Kessler
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| | - Sophie Croizier
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| | - Sevasti Gaspari
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne, Switzerland
| |
Collapse
|
21
|
Cavalcanti-de-Albuquerque JP, Donato J. Rolling out physical exercise and energy homeostasis: Focus on hypothalamic circuitries. Front Neuroendocrinol 2021; 63:100944. [PMID: 34425188 DOI: 10.1016/j.yfrne.2021.100944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023]
Abstract
Energy balance is the fine regulation of energy expenditure and energy intake. Negative energy balance causes body weight loss, while positive energy balance promotes weight gain. Modern societies offer a maladapted way of life, where easy access to palatable foods and the lack of opportunities to perform physical activity are considered the roots of the obesity pandemic. Physical exercise increases energy expenditure and, consequently, is supposed to promote weight loss. Paradoxically, physical exercise acutely drives anorexigenic-like effects, but the mechanisms are still poorly understood. Using an evolutionary background, this review aims to highlight the potential involvement of the melanocortin system and other hypothalamic neural circuitries regulating energy balance during and after physical exercise. The physiological significance of these changes will be explored, and possible signalling agents will be addressed. The knowledge discussed here might be important for clarifying obesity aetiology as well as new therapeutic approaches for body weight loss.
Collapse
Affiliation(s)
| | - José Donato
- Department of Physiology and Biophysics, University of São Paulo, São Paulo 05508-900, Brazil.
| |
Collapse
|
22
|
Abstract
The ventromedial nucleus of the hypothalamus (VMH) is a complex brain structure that is integral to many neuroendocrine functions, including glucose regulation, thermogenesis, and appetitive, social, and sexual behaviors. As such, it is of little surprise that the nucleus is under intensive investigation to decipher the mechanisms which underlie these diverse roles. Developments in genetic and investigative tools, for example the targeting of steroidogenic factor-1-expressing neurons, have allowed us to take a closer look at the VMH, its connections, and how it affects competing behaviors. In the current review, we aim to integrate recent findings into the literature and contemplate the conclusions that can be drawn.
Collapse
Affiliation(s)
- Tansi Khodai
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK
| | - Simon M Luckman
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK
- Correspondence: Simon M. Luckman, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, UK.
| |
Collapse
|
23
|
Yan H, Fan W, Chen X, Liu L, Wang H, Jiang X. Terahertz signatures and quantitative analysis of glucose anhydrate and monohydrate mixture. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 258:119825. [PMID: 33901947 DOI: 10.1016/j.saa.2021.119825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Glucose, as the main energy carrier and significant source of nutrition, generally comes in two available forms of anhydrate and monohydrate in commercial production. Considering their respective application occasions, proper identification of glucose in single composition or binary-mixture and quantification of the mixture are crucial in industry monitoring to guarantee merchandise quality. Simultaneously, public confusions of glucose are rather ubiquitous partly due to anhydrate and monohydrate with identical white crystalline appearance. In this paper, utilizing the molecular fingerprints of terahertz (THz) technology that are corresponding to structural characteristics of anhydrous and hydrated form, THz signatures of glucose anhydrate, monohydrate and their mixture, as well as THz spectral transformation from monohydrate to anhydrate with the dehydrating process are systematically studied. Some visible peaks of monohydrate were noted at 1.82 and 1.99 THz signifying the presence of hydrated structure. However, with the dehydrating process, the peaks related to the hydrated structure are not very apparent when the peaks at 1.44 and 2.08 THz appear due to changes in the molecular structure of anhydrate, which provide clear indication for hydrogen-bond network reconstruction at the micro level. Furthermore, characteristic peaks at 1.44 and 1.82 THz can be specified as the main quantitative indicators for quantitative detection. The linear relationships between the amplitudes of characteristic peaks and the percentage compositions of anhydrate and monohydrate are revealed. Three commercially available brands of edible glucose powder A, B, C were effectively identified by THz signatures. While powder C was recognized as binary-mixture and the proportion of anhydrate and monohydrate was further quantified. THz spectroscopy technology has advantages of direct recognition, simple quantitative model based on THz absorption peaks, and no need for complicated chemical treatment. It may be potentially shed light on industrial monitoring of glucose production and other related mixture in the future.
Collapse
Affiliation(s)
- Hui Yan
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China; College of Science, Zhongyuan University of Technology, Zhengzhou 450007, China; Zhengzhou Key Laboratory of Low-dimensional Quantum Materials and Devices; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenhui Fan
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China; University of Chinese Academy of Sciences, Beijing 100049, China; Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan 030006, China.
| | - Xu Chen
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China
| | - Lutao Liu
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hanqi Wang
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoqiang Jiang
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an 710119, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
24
|
De Angelis LC, Brigati G, Polleri G, Malova M, Parodi A, Minghetti D, Rossi A, Massirio P, Traggiai C, Maghnie M, Ramenghi LA. Neonatal Hypoglycemia and Brain Vulnerability. Front Endocrinol (Lausanne) 2021; 12:634305. [PMID: 33796072 PMCID: PMC8008815 DOI: 10.3389/fendo.2021.634305] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Neonatal hypoglycemia is a common condition. A transient reduction in blood glucose values is part of a transitional metabolic adaptation following birth, which resolves within the first 48 to 72 h of life. In addition, several factors may interfere with glucose homeostasis, especially in case of limited metabolic stores or increased energy expenditure. Although the effect of mild transient asymptomatic hypoglycemia on brain development remains unclear, a correlation between severe and prolonged hypoglycemia and cerebral damage has been proven. A selective vulnerability of some brain regions to hypoglycemia including the second and the third superficial layers of the cerebral cortex, the dentate gyrus, the subiculum, the CA1 regions in the hippocampus, and the caudate-putamen nuclei has been observed. Several mechanisms contribute to neuronal damage during hypoglycemia. Neuronal depolarization induced by hypoglycemia leads to an elevated release of glutamate and aspartate, thus promoting excitotoxicity, and to an increased release of zinc to the extracellular space, causing the extensive activation of poly ADP-ribose polymerase-1 which promotes neuronal death. In this review we discuss the cerebral glucose homeostasis, the mechanisms of brain injury following neonatal hypoglycemia and the possible treatment strategies to reduce its occurrence.
Collapse
Affiliation(s)
- Laura Costanza De Angelis
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Giorgia Brigati
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giulia Polleri
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mariya Malova
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Alessandro Parodi
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Diego Minghetti
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Andrea Rossi
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Neuroradiology Unit, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Massirio
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Cristina Traggiai
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mohamad Maghnie
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Luca Antonio Ramenghi
- Neonatal Intensive Care Unit, Department Mother and Child, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| |
Collapse
|
25
|
Duca FA, Waise TMZ, Peppler WT, Lam TKT. The metabolic impact of small intestinal nutrient sensing. Nat Commun 2021; 12:903. [PMID: 33568676 PMCID: PMC7876101 DOI: 10.1038/s41467-021-21235-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract maintains energy and glucose homeostasis, in part through nutrient-sensing and subsequent signaling to the brain and other tissues. In this review, we highlight the role of small intestinal nutrient-sensing in metabolic homeostasis, and link high-fat feeding, obesity, and diabetes with perturbations in these gut-brain signaling pathways. We identify how lipids, carbohydrates, and proteins, initiate gut peptide release from the enteroendocrine cells through small intestinal sensing pathways, and how these peptides regulate food intake, glucose tolerance, and hepatic glucose production. Lastly, we highlight how the gut microbiota impact small intestinal nutrient-sensing in normal physiology, and in disease, pharmacological and surgical settings. Emerging evidence indicates that the molecular mechanisms of small intestinal nutrient sensing in metabolic homeostasis have physiological and pathological impact as well as therapeutic potential in obesity and diabetes. The gastrointestinal tract participates in maintaining metabolic homeostasis in part through nutrient-sensing and subsequent gut-brain signalling. Here the authors review the role of small intestinal nutrient-sensing in regulation of energy intake and systemic glucose metabolism, and link high-fat diet, obesity and diabetes with perturbations in these pathways.
Collapse
Affiliation(s)
- Frank A Duca
- BIO5 Institute, University of Arizona, Tucson, AZ, USA. .,School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA.
| | - T M Zaved Waise
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Willem T Peppler
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Tony K T Lam
- Toronto General Hospital Research Institute, UHN, Toronto, Canada. .,Department of Physiology, University of Toronto, Toronto, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
26
|
Simon JJ, Stopyra MA, Mönning E, Sailer S, Lavandier N, Kihm LP, Bendszus M, Preissl H, Herzog W, Friederich HC. Neuroimaging of hypothalamic mechanisms related to glucose metabolism in anorexia nervosa and obesity. J Clin Invest 2021; 130:4094-4103. [PMID: 32315289 DOI: 10.1172/jci136782] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUNDGiven the heightened tolerance to self-starvation in anorexia nervosa (AN), a hypothalamic dysregulation of energy and glucose homeostasis has been hypothesized. Therefore, we investigated whether hypothalamic reactivity to glucose metabolism is impaired in AN.METHODSTwenty-four participants with AN, 28 normal-weight participants, and 24 healthy participants with obesity underwent 2 MRI sessions in a single-blind, randomized, case-controlled crossover study. We used an intragastric infusion of glucose and water to bypass the cephalic phase of food intake. The responsivity of the hypothalamus and the crosstalk of the hypothalamus with reward-related brain regions were investigated using high-resolution MRI.RESULTSNormal-weight control participants displayed the expected glucose-induced deactivation of hypothalamic activation, whereas patients with AN and participants with obesity showed blunted hypothalamic reactivity. Furthermore, patients with AN displayed blunted reactivity in the nucleus accumbens and amygdala. Compared with the normal-weight participants and control participants with obesity, the patients with AN failed to show functional connectivity between the hypothalamus and the reward-related brain regions during water infusion relative to glucose infusion. Finally, the patients with AN displayed typical baseline levels of peripheral appetite hormones during a negative energy balance.CONCLUSIONThese results indicate that blunted hypothalamic glucose reactivity might be related to the pathophysiology of AN. This study provides insights for future research, as it is an extended perspective of the traditional primary nonhomeostatic understanding of the disease.FUNDINGThis study was supported by a grant from the DFG (SI 2087/2-1).
Collapse
Affiliation(s)
- Joe J Simon
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marion A Stopyra
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Mönning
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Sailer
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Nora Lavandier
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Lars P Kihm
- Endocrinology and Nephrology, Department of Internal Medicine I, and
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Hubert Preissl
- fMEG Center, Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich (IDM) at the University of Tübingen, Tübingen, Germany.,German Center for Diabetes Research (DZD e.V.), Tübingen, Germany.,Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.,Department of Pharmacy and Biochemistry, Interfaculty Centre for Pharmacogenomics and Pharma Research, University of Tübingen, Tübingen, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Centre, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Wolfgang Herzog
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Christoph Friederich
- Centre for Psychosocial Medicine, Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
27
|
Tirou L, Russo M, Faure H, Pellegrino G, Demongin C, Daynac M, Sharif A, Amosse J, Le Lay S, Denis R, Luquet S, Taouis M, Benomar Y, Ruat M. Sonic Hedgehog receptor Patched deficiency in astrocytes enhances glucose metabolism in mice. Mol Metab 2021; 47:101172. [PMID: 33513436 PMCID: PMC7893488 DOI: 10.1016/j.molmet.2021.101172] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 01/06/2023] Open
Abstract
Objective Astrocytes are glial cells proposed as the main Sonic hedgehog (Shh)-responsive cells in the adult brain. Their roles in mediating Shh functions are still poorly understood. In the hypothalamus, astrocytes support neuronal circuits implicated in the regulation of energy metabolism. In this study, we investigated the impact of genetic activation of Shh signaling on hypothalamic astrocytes and characterized its effects on energy metabolism. Methods We analyzed the distribution of gene transcripts of the Shh pathway (Ptc, Gli1, Gli2, and Gli3) in astrocytes using single molecule fluorescence in situ hybridization combined with immunohistofluorescence of Shh peptides by Western blotting in the adult mouse hypothalamus. Based on the metabolic phenotype, we characterized Glast-CreERT2-YFP-Ptc−/− (YFP-Ptc−/−) mice and their controls over time and under a high-fat diet (HFD) to investigate the potential effects of conditional astrocytic deletion of the Shh receptor Patched (Ptc) on metabolic efficiency, insulin sensitivity, and systemic glucose metabolism. Molecular and biochemical assays were used to analyze the alteration of key pathways modulating energy metabolism, insulin sensitivity, glucose uptake, and inflammation. Primary astrocyte cultures were used to evaluate a potential role of Shh signaling in astrocytic glucose uptake. Results Shh peptides were the highest in the hypothalamic extracts of adult mice and a large population of hypothalamic astrocytes expressed Ptc and Gli1-3 mRNAs. Characterization of Shh signaling after conditional Ptc deletion in the YFP-Ptc−/− mice revealed heterogeneity in hypothalamic astrocyte populations. Interestingly, activation of Shh signaling in Glast+ astrocytes enhanced insulin responsiveness as evidenced by glucose and insulin tolerance tests. This effect was maintained over time and associated with lower blood insulin levels and also observed under a HFD. The YFP-Ptc−/− mice exhibited a lean phenotype with the absence of body weight gain and a marked reduction of white and brown adipose tissues accompanied by increased whole-body fatty acid oxidation. In contrast, food intake, locomotor activity, and body temperature were not altered. At the cellular level, Ptc deletion did not affect glucose uptake in primary astrocyte cultures. In the hypothalamus, activation of the astrocytic Shh pathway was associated with the upregulation of transcripts coding for the insulin receptor and liver kinase B1 (LKB1) after 4 weeks and the glucose transporter GLUT-4 after 32 weeks. Conclusions Here, we define hypothalamic Shh action on astrocytes as a novel master regulator of energy metabolism. In the hypothalamus, astrocytic Shh signaling could be critically involved in preventing both aging- and obesity-related metabolic disorders. Astrocytes exhibit differences in regulating the hedgehog signaling pathway. Deletion of Ptc in Glast+ cells prevents body weight gain and insulin resistance. Deletion of Ptc in Glast+ cells increases β oxidation. Central hedgehog signaling participates in the regulation of whole-body metabolism.
Collapse
Affiliation(s)
- Linda Tirou
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mariagiovanna Russo
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Helene Faure
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Giuliana Pellegrino
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Clement Demongin
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Mathieu Daynac
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog (JPARC) - Lille Neurosciences & Cognition, F-59000, Lille, France
| | - Jeremy Amosse
- Univ. Angers SFR ICAT, F-49100, Angers, France; IRSET Laboratory, Inserm, UMR, 1085, Rennes, France
| | - Soazig Le Lay
- Univ. Angers SFR ICAT, F-49100, Angers, France; Univ. Nantes, CNRS, Inserm, Thorax Institut, F-44000, Nantes, France
| | - Raphaël Denis
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Serge Luquet
- Univ. Paris, BFA, UMR 8251, CNRS, F-75013, Paris, France
| | - Mohammed Taouis
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Yacir Benomar
- CNRS, Paris-Saclay University, UMR 9197, Neuroscience Paris-Saclay Institute, Molecular Neuroendocrinology of Food Intake, Orsay, France
| | - Martial Ruat
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, F-91198, Gif-sur-Yvette, France.
| |
Collapse
|
28
|
Lin EE, Scott-Solomon E, Kuruvilla R. Peripheral Innervation in the Regulation of Glucose Homeostasis. Trends Neurosci 2020; 44:189-202. [PMID: 33229051 DOI: 10.1016/j.tins.2020.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Precise regulation of circulating glucose is crucial for human health and ensures a sufficient supply to the brain, which relies almost exclusively on glucose for metabolic energy. Glucose homeostasis is coordinated by hormone-secreting endocrine cells in the pancreas, as well as glucose utilization and production in peripheral metabolic tissues including the liver, muscle, and adipose tissue. Glucose-regulatory tissues receive dense innervation from sympathetic, parasympathetic, and sensory fibers. In this review, we summarize the functions of peripheral nerves in glucose regulation and metabolism. Dynamic changes in peripheral innervation have also been observed in animal models of obesity and diabetes. Together, these studies highlight the importance of peripheral nerves as a new therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Eugene E Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
29
|
Quenneville S, Labouèbe G, Basco D, Metref S, Viollet B, Foretz M, Thorens B. Hypoglycemia-Sensing Neurons of the Ventromedial Hypothalamus Require AMPK-Induced Txn2 Expression but Are Dispensable for Physiological Counterregulation. Diabetes 2020; 69:2253-2266. [PMID: 32839348 PMCID: PMC7576557 DOI: 10.2337/db20-0577] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/18/2020] [Indexed: 12/23/2022]
Abstract
The ventromedial nucleus of the hypothalamus (VMN) is involved in the counterregulatory response to hypoglycemia. VMN neurons activated by hypoglycemia (glucose-inhibited [GI] neurons) have been assumed to play a critical although untested role in this response. Here, we show that expression of a dominant negative form of AMPK or inactivation of AMPK α1 and α2 subunit genes in Sf1 neurons of the VMN selectively suppressed GI neuron activity. We found that Txn2, encoding a mitochondrial redox enzyme, was strongly downregulated in the absence of AMPK activity and that reexpression of Txn2 in Sf1 neurons restored GI neuron activity. In cell lines, Txn2 was required to limit glucopenia-induced reactive oxygen species production. In physiological studies, absence of GI neuron activity after AMPK suppression in the VMN had no impact on the counterregulatory hormone response to hypoglycemia or on feeding. Thus, AMPK is required for GI neuron activity by controlling the expression of the antioxidant enzyme Txn2. However, the glucose-sensing capacity of VMN GI neurons is not required for the normal counterregulatory response to hypoglycemia. Instead, it may represent a fail-safe system in case of impaired hypoglycemia sensing by peripherally located glucose detection systems that are connected to the VMN.
Collapse
Affiliation(s)
- Simon Quenneville
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Salima Metref
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
30
|
Faber CL, Deem JD, Campos CA, Taborsky GJ, Morton GJ. CNS control of the endocrine pancreas. Diabetologia 2020; 63:2086-2094. [PMID: 32894319 PMCID: PMC7983553 DOI: 10.1007/s00125-020-05204-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that, although pancreatic islets can function autonomously to detect and respond to changes in the circulating glucose level, the brain cooperates with the islet to maintain glycaemic control. Here, we review the role of the central and autonomic nervous systems in the control of the endocrine pancreas, including mechanisms whereby the brain senses circulating blood glucose levels. We also examine whether dysfunction in these systems might contribute to complications of type 1 diabetes and the pathogenesis of type 2 diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Chelsea L Faber
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer D Deem
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Carlos A Campos
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gerald J Taborsky
- Department of Medicine, University of Washington, Seattle, WA, USA
- Veterans Affairs Puget Sound Health Care System, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Gregory J Morton
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
31
|
Reilly AM, Tsai AP, Lin PB, Ericsson AC, Oblak AL, Ren H. Metabolic Defects Caused by High-Fat Diet Modify Disease Risk through Inflammatory and Amyloidogenic Pathways in a Mouse Model of Alzheimer's Disease. Nutrients 2020; 12:nu12102977. [PMID: 33003412 PMCID: PMC7600118 DOI: 10.3390/nu12102977] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
High-fat diet (HFD) has been shown to accelerate Alzheimer’s disease (AD) pathology, but the exact molecular and cellular mechanisms remain incompletely understood. Moreover, it is unknown whether AD mice are more susceptible to HFD-induced metabolic dysfunctions. To address these questions, we used 5xFAD mice as an Alzheimer’s disease model to study the physiological and molecular underpinning between HFD-induced metabolic defects and AD pathology. We systematically profiled the metabolic parameters, the gut microbiome composition, and hippocampal gene expression in 5xFAD and wild type (WT) mice fed normal chow diet and HFD. HFD feeding impaired energy metabolism in male 5xFAD mice, leading to increased locomotor activity, energy expenditure, and food intake. 5xFAD mice on HFD had elevated circulating lipids and worsened glucose intolerance. HFD caused profound changes in gut microbiome compositions, though no difference between genotype was detected. We measured hippocampal mRNAs related to AD neuropathology and neuroinflammation and showed that HFD elevated the expression of apoptotic, microglial, and amyloidogenic genes in 5xFAD mice. Pathway analysis revealed that differentially regulated genes were involved in insulin signaling, cytokine signaling, cellular stress, and neurotransmission. Collectively, our results showed that 5xFAD mice were more susceptible to HFD-induced metabolic dysregulation and suggest that targeting metabolic dysfunctions can ameliorate AD symptoms via effects on insulin signaling and neuroinflammation in the hippocampus.
Collapse
Affiliation(s)
- Austin M. Reilly
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Andy P. Tsai
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Peter B. Lin
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Aaron C. Ericsson
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA;
| | - Adrian L. Oblak
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Hongxia Ren
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-274-1567
| |
Collapse
|
32
|
Bentsen MA, Rausch DM, Mirzadeh Z, Muta K, Scarlett JM, Brown JM, Herranz-Pérez V, Baquero AF, Thompson J, Alonge KM, Faber CL, Kaiyala KJ, Bennett C, Pyke C, Ratner C, Egerod KL, Holst B, Meek TH, Kutlu B, Zhang Y, Sparso T, Grove KL, Morton GJ, Kornum BR, García-Verdugo JM, Secher A, Jorgensen R, Schwartz MW, Pers TH. Transcriptomic analysis links diverse hypothalamic cell types to fibroblast growth factor 1-induced sustained diabetes remission. Nat Commun 2020; 11:4458. [PMID: 32895383 PMCID: PMC7477234 DOI: 10.1038/s41467-020-17720-5] [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: 03/05/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
In rodent models of type 2 diabetes (T2D), sustained remission of hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1), and the mediobasal hypothalamus (MBH) was recently implicated as the brain area responsible for this effect. To better understand the cellular response to FGF1 in the MBH, we sequenced >79,000 single-cell transcriptomes from the hypothalamus of diabetic Lepob/ob mice obtained on Days 1 and 5 after icv injection of either FGF1 or vehicle. A wide range of transcriptional responses to FGF1 was observed across diverse hypothalamic cell types, with glial cell types responding much more robustly than neurons at both time points. Tanycytes and ependymal cells were the most FGF1-responsive cell type at Day 1, but astrocytes and oligodendrocyte lineage cells subsequently became more responsive. Based on histochemical and ultrastructural evidence of enhanced cell-cell interactions between astrocytes and Agrp neurons (key components of the melanocortin system), we performed a series of studies showing that intact melanocortin signaling is required for the sustained antidiabetic action of FGF1. These data collectively suggest that hypothalamic glial cells are leading targets for the effects of FGF1 and that sustained diabetes remission is dependent on intact melanocortin signaling.
Collapse
MESH Headings
- Agouti-Related Protein/metabolism
- Animals
- Astrocytes/drug effects
- Astrocytes/metabolism
- Blood Glucose/analysis
- Cell Communication
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/diet therapy
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Dietary Sucrose/administration & dosage
- Dietary Sucrose/adverse effects
- Fibroblast Growth Factor 1/administration & dosage
- Humans
- Hypoglycemic Agents/administration & dosage
- Hypothalamus/cytology
- Hypothalamus/drug effects
- Hypothalamus/pathology
- Injections, Intraventricular
- Leptin/genetics
- Male
- Melanocortins/metabolism
- Melanocyte-Stimulating Hormones/administration & dosage
- Mice
- Mice, Knockout
- Neurons/drug effects
- Neurons/metabolism
- Oligodendroglia/drug effects
- Oligodendroglia/metabolism
- RNA-Seq
- Receptor, Melanocortin, Type 4/genetics
- Receptors, Melanocortin/antagonists & inhibitors
- Receptors, Melanocortin/metabolism
- Recombinant Proteins/administration & dosage
- Remission Induction/methods
- Signal Transduction/drug effects
- Single-Cell Analysis
- Stereotaxic Techniques
- Transcriptome/drug effects
Collapse
Affiliation(s)
- Marie A Bentsen
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dylan M Rausch
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Kenjiro Muta
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jarrad M Scarlett
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Jenny M Brown
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Vicente Herranz-Pérez
- Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
- Predepartamental Unit of Medicine, Jaume I University, Castelló de la Plana, Spain
| | - Arian F Baquero
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Jonatan Thompson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kimberly M Alonge
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Chelsea L Faber
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Karl J Kaiyala
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, USA
| | - Camdin Bennett
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Charles Pyke
- Pathology & Imaging, Global Discovery and Development Sciences, Novo Nordisk A/S, Maaloev, Denmark
| | - Cecilia Ratner
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas H Meek
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Burak Kutlu
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Yu Zhang
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Thomas Sparso
- Bioinformatics and Data Mining, Global Research Technologies, Novo Nordisk A/S, Maaloev, Denmark
| | - Kevin L Grove
- Obesity Research Unit, Novo Nordisk Research Center Seattle, Inc., Seattle, WA, USA
| | - Gregory J Morton
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Birgitte R Kornum
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Anna Secher
- Diabetes Research, Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| | - Rasmus Jorgensen
- Diabetes Research, Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
- Cytoki Pharma, Copenhagen, Denmark
| | - Michael W Schwartz
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA.
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
33
|
Carey M, Lontchi-Yimagou E, Mitchell W, Reda S, Zhang K, Kehlenbrink S, Koppaka S, Maginley SR, Aleksic S, Bhansali S, Huffman DM, Hawkins M. Central K ATP Channels Modulate Glucose Effectiveness in Humans and Rodents. Diabetes 2020; 69:1140-1148. [PMID: 32217610 PMCID: PMC7243288 DOI: 10.2337/db19-1256] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022]
Abstract
Hyperglycemia is a potent regulator of endogenous glucose production (EGP). Loss of this "glucose effectiveness" is a major contributor to elevated plasma glucose concentrations in type 2 diabetes (T2D). KATP channels in the central nervous system have been shown to regulate EGP in humans and rodents. We examined the contribution of central KATP channels to glucose effectiveness. Under fixed hormonal conditions (studies using a pancreatic clamp), hyperglycemia suppressed EGP by ∼50% in both humans without diabetes and normal Sprague-Dawley rats. By contrast, antagonism of KATP channels with glyburide significantly reduced the EGP-lowering effect of hyperglycemia in both humans and rats. Furthermore, the effects of glyburide on EGP and gluconeogenic enzymes were abolished in rats by intracerebroventricular administration of the KATP channel agonist diazoxide. These findings indicate that about half of the suppression of EGP by hyperglycemia is mediated by central KATP channels. These central mechanisms may offer a novel therapeutic target for improving glycemic control in subjects with T2D.
Collapse
Affiliation(s)
- Michelle Carey
- Albert Einstein College of Medicine, Bronx, NY
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD
| | | | | | - Sarah Reda
- Albert Einstein College of Medicine, Bronx, NY
| | - Kehao Zhang
- Albert Einstein College of Medicine, Bronx, NY
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Interaction of glucose sensing and leptin action in the brain. Mol Metab 2020; 39:101011. [PMID: 32416314 PMCID: PMC7267726 DOI: 10.1016/j.molmet.2020.101011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023] Open
Abstract
Background In response to energy abundant or deprived conditions, nutrients and hormones activate hypothalamic pathways to maintain energy and glucose homeostasis. The underlying CNS mechanisms, however, remain elusive in rodents and humans. Scope of review Here, we first discuss brain glucose sensing mechanisms in the presence of a rise or fall of plasma glucose levels, and highlight defects in hypothalamic glucose sensing disrupt in vivo glucose homeostasis in high-fat fed, obese, and/or diabetic conditions. Second, we discuss brain leptin signalling pathways that impact glucose homeostasis in glucose-deprived and excessed conditions, and propose that leptin enhances hypothalamic glucose sensing and restores glucose homeostasis in short-term high-fat fed and/or uncontrolled diabetic conditions. Major conclusions In conclusion, we believe basic studies that investigate the interaction of glucose sensing and leptin action in the brain will address the translational impact of hypothalamic glucose sensing in diabetes and obesity.
Collapse
|
35
|
Schwartsburd P. A View on Pathogenesis of ≪Vicious Cancer Progression Cycle≫. Front Oncol 2020; 10:690. [PMID: 32426290 PMCID: PMC7204907 DOI: 10.3389/fonc.2020.00690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022] Open
Abstract
Unrestricted tumor growth requires a permanent supply of glucose that can be obtained from cancer-stimulated hepatic glucose production and/or glucose redirecting from host insulin resistant tissues to cancer cells. This study proposes a mechanism based on metabolic and hormonal changes that may provoke glucose delivery to cancer cells through two interconnected "vicious cycles" whose continuous activity drives cancer progression. As follows from the proposed here feedback model, these "vicious cycles" result from cancer-mediated manipulation of host glucose sensors. The derived conclusions contribute to a better understanding of cancer pathogenesis and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Polina Schwartsburd
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
36
|
Lizarbe B, Fernández-Pérez A, Caz V, Largo C, Vallejo M, López-Larrubia P, Cerdán S. Systemic Glucose Administration Alters Water Diffusion and Microvascular Blood Flow in Mouse Hypothalamic Nuclei - An fMRI Study. Front Neurosci 2019; 13:921. [PMID: 31551685 PMCID: PMC6733885 DOI: 10.3389/fnins.2019.00921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/16/2019] [Indexed: 12/23/2022] Open
Abstract
The hypothalamus is the principal regulator of global energy balance, enclosing additionally essential neuronal centers for glucose-sensing and osmoregulation. Disturbances in these tightly regulated neuronal networks are thought to underlie the development of severe pandemic syndromes, including obesity and diabetes. In this work, we investigate in vivo the response of individual hypothalamic nuclei to the i.p. administration of glucose or vehicle solutions, using two groups of adult male C57BL6/J fasted mice and a combination of non-invasive T2∗-weighted and diffusion-weighted functional magnetic resonance imaging (fMRI) approaches. MRI parameters were assessed in both groups of animals before, during and in a post-stimulus phase, following the administration of glucose or vehicle solutions. Hypothalamic nuclei depicted different patterns of activation characterized by: (i) generalized glucose-induced increases of neuronal activation and perfusion-markers in the lateral hypothalamus, arcuate and dorsomedial nuclei, (ii) cellular shrinking events and decreases in microvascular blood flow in the dorsomedial, ventromedial and lateral hypothalamus, following the administration of vehicle solutions and (iii) increased neuronal activity markers and decreased microperfusion parameters in the ARC nuclei of vehicle-administered animals. Immunohistochemical studies performed after the post-stimulus phase confirmed the presence of c-Fos immunoreactive neurons in the arcuate nucleus (ARC) from both animal groups, with significantly higher numbers in the glucose-treated animals. Together, our results reveal that fMRI methods are able to detect in vivo diverse patterns of glucose or vehicle-induced effects in the different hypothalamic nuclei.
Collapse
Affiliation(s)
- Blanca Lizarbe
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Antonio Fernández-Pérez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (Ciberdem), Instituto de Salud Carlos III, Madrid, Spain
| | - Victor Caz
- Departamento de Cirugía Experimental, Instituto de Investigación Hospital Universitario La Paz - IdiPAZ, Madrid, Spain
| | - Carlota Largo
- Departamento de Cirugía Experimental, Instituto de Investigación Hospital Universitario La Paz - IdiPAZ, Madrid, Spain
| | - Mario Vallejo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (Ciberdem), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Sebastián Cerdán
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| |
Collapse
|
37
|
Brown JM, Scarlett JM, Matsen ME, Nguyen HT, Secher A, Jorgensen R, Morton GJ, Schwartz MW. The Hypothalamic Arcuate Nucleus-Median Eminence Is a Target for Sustained Diabetes Remission Induced by Fibroblast Growth Factor 1. Diabetes 2019; 68:1054-1061. [PMID: 30796029 PMCID: PMC6477902 DOI: 10.2337/db19-0025] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
Abstract
In rodent models of type 2 diabetes (T2D), sustained remission of diabetic hyperglycemia can be induced by a single intracerebroventricular (icv) injection of fibroblast growth factor 1 (FGF1). To identify the brain areas responsible for this effect, we first used immunohistochemistry to map the hypothalamic distribution of phosphorylated extracellular signal-related kinase 1/2 (pERK1/2), a marker of mitogen-activated protein kinase-ERK signal transduction downstream of FGF receptor activation. Twenty minutes after icv FGF1 injection in adult male Wistar rats, pERK1/2 staining was detected primarily in two hypothalamic areas: the arcuate nucleus-median eminence (ARC-ME) and the paraventricular nucleus (PVN). To determine whether an action of FGF1 localized to either the ARC-ME or the PVN is capable of mimicking the sustained antidiabetic effect elicited by icv FGF1, we microinjected either saline vehicle or a low dose of FGF1 (0.3 µg/side) bilaterally into either the ARC-ME area or PVN of Zucker Diabetic Fatty rats, a model of T2D, and monitored daily food intake, body weight, and blood glucose levels over a 3-week period. Whereas bilateral intra-arcuate microinjection of saline vehicle was without effect, remission of hyperglycemia lasting >3 weeks was observed following bilateral microinjection of FGF1 into the ARC-ME. This antidiabetic effect cannot be attributed to leakage of FGF1 into cerebrospinal fluid and subsequent action on other brain areas, since icv injection of the same total dose was without effect. Combined with our finding that bilateral microinjection of the same dose of FGF1 into the PVN was without effect on glycemia or other parameters, we conclude that the ARC-ME area (but not the PVN) is a target for sustained remission of diabetic hyperglycemia induced by FGF1.
Collapse
Affiliation(s)
- Jenny M Brown
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
- Department of Pediatric Gastroenterology and Hepatology, University of Washington, Seattle, WA
| | - Miles E Matsen
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Hong T Nguyen
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Anna Secher
- Diabetes Research, Global Drug Discovery, Novo Nordisk, Måløv, Denmark
| | - Rasmus Jorgensen
- Diabetes Research, Global Drug Discovery, Novo Nordisk, Måløv, Denmark
| | - Gregory J Morton
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Michael W Schwartz
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| |
Collapse
|