1
|
Iremonger KJ, Power EM. The paraventricular nucleus of the hypothalamus: a key node in the control of behavioural states. J Physiol 2025. [PMID: 40119815 DOI: 10.1113/jp288366] [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: 01/07/2025] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
The paraventricular nucleus (PVN) of the hypothalamus contains diverse populations of neuropeptide-producing neurons. These include neurons that synthesise oxytocin, vasopressin, corticotropin-releasing hormone, thyrotropin-releasing hormone and somatostatin. While it is well established that these neurons control the secretion of neuroendocrine hormones, there is growing evidence that they also control the expression of important homeostatic behaviours. Here we review recent data showing a critical role of PVN neurons in controlling arousal, social behaviour, defensive behaviour and pain. Collectively, this suggests that the PVN is a key node in a wider neural network controlling behavioural states.
Collapse
Affiliation(s)
- Karl J Iremonger
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Emmet M Power
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
2
|
Desmoulins LD, Molinas AJR, Dugas CM, Williams GL, Kamenetsky S, Davis RK, Derbenev AV, Zsombok A. A subset of neurons in the paraventricular nucleus of the hypothalamus directly project to liver-related premotor neurons in the ventrolateral medulla. Auton Neurosci 2025; 257:103222. [PMID: 39647176 DOI: 10.1016/j.autneu.2024.103222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/03/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
Sympathetic circuits including pre-sympathetic neurons in the ventrolateral medulla (VLM) and in the paraventricular nucleus (PVN) of the hypothalamus play an important role in the regulation of hepatic glucose metabolism. Despite the importance of central regulatory pathways, specific information regarding the circuits of liver-related neurons is limited. Here, we tested the hypothesis that PVN neurons are directly connected to spinally-projecting liver-related neurons in the VLM of mice. Pseudorabies virus (PRV) was used to identify liver-related neurons and time-dependent analyses revealed the location and distribution of neurons in the PVN and ventral brainstem. Four days following PRV injection, most liver-related neurons were found in the VLM and consist of both catecholaminergic (CA) and non-CA neurons. Furthermore, in addition to PRV inoculation, a monosynaptic viral tracer was used to identify VLM-projecting PVN neurons to specifically dissect PVN-VLM connections within the liver pathway. Five days following PRV inoculation, our anatomical findings revealed that a small population of liver-related PVN neurons projected to the VLM. In addition, photo-stimulation of axonal projections from SIM1-expressing PVN neurons resulted in evoked excitatory postsynaptic currents in a subset of spinally projecting liver-related neurons in the VLM. In summary, our data demonstrate the existence of monosynaptic, glutamatergic connections between PVN neurons and pre-sympathetic liver-related neurons in the VLM. These new findings regarding the central circuits involved in the sympathetic regulation of the liver provide further information necessary for developing new strategies to improve glucose homeostasis via modulation of the autonomic nerves.
Collapse
Affiliation(s)
- Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Adrien J R Molinas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Courtney M Dugas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Gabrielle L Williams
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Sophie Kamenetsky
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Roslyn K Davis
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA; Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, USA; Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA.
| |
Collapse
|
3
|
Martins Dos Santos K, Saunders SE, Antunes VR, Boychuk CR. Insulin activates parasympathetic hepatic-related neurons of the paraventricular nucleus of the hypothalamus through mTOR signaling. J Neurophysiol 2025; 133:320-332. [PMID: 39665212 PMCID: PMC11918334 DOI: 10.1152/jn.00284.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
Integration of autonomic and metabolic regulation, including hepatic function, is a critical role played by the brain's hypothalamic region. Specifically, the paraventricular nucleus of the hypothalamus (PVN) regulates autonomic functions related to metabolism, such as hepatic glucose production. Although insulin can act directly on hepatic tissue to inhibit hepatic glucose production, recent evidence implicates that central actions of insulin within PVN also regulate glucose metabolism. However, specific central circuits responsible for insulin signaling with relation to hepatic regulation are poorly understood. As a heterogeneous nucleus essential to controlling parasympathetic motor output with notable expression of insulin receptors, PVN is an appealing target for insulin-dependent modulation of parasympathetic activity. Here, we tested the hypothesis that insulin activates hepatic-related PVN (PVNhepatic) neurons through a parasympathetic pathway. Using transsynaptic retrograde tracing, labeling within PVN was first identified 24 h after its expression in the dorsal motor nucleus of the vagus (DMV) and 72 h after hepatic injection. Critically, nearly all labeling in medial PVN was abolished after a left vagotomy, indicating that PVNhepatic neurons in this region are part of a central circuit innervating parasympathetic motor neurons. Insulin also significantly increased the firing frequency of PVNhepatic neurons in this subregion. Mechanistically, rapamycin pretreatment inhibited insulin-dependent activation of PVNhepatic neurons. Therefore, central insulin signaling can activate a subset of PVNhepatic neurons that are part of a unique parasympathetic network in control of hepatic function. Taken together, PVNhepatic neurons related to parasympathetic output regulation could serve as a key central network in insulin's ability to control hepatic functions.NEW & NOTEWORTHY Increased peripheral insulin concentrations are known to decrease hepatic glucose production through both direct actions on hepatocytes and central autonomic networks. Despite this understanding, how (and in which brain regions) insulin exerts its action is still obscure. Here, we demonstrate that insulin activates parasympathetic hepatic-related PVN neurons (PVNhepatic) and that this effect relies on mammalian target of rapamycin (mTOR) signaling, suggesting that insulin modulates hepatic function through autonomic pathways involving insulin receptor intracellular signaling cascades.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandy E Saunders
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| | - Vagner R Antunes
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, United States
- Dalton Cardiovascular Research Center, Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
4
|
Du T, Liu S, Yu H, Hu T, Huang L, Gao L, Jia L, Hu J, Yu Y, Sun Q. Chronic sleep deprivation disturbs energy balance modulated by suprachiasmatic nucleus efferents in mice. BMC Biol 2024; 22:296. [PMID: 39710657 DOI: 10.1186/s12915-024-02097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Epidemiologic researches show that short sleep duration may affect feeding behaviors resulting in higher energy intake and increased risk of obesity, but the further mechanisms that can interpret the causality remain unclear. The circadian rhythm is fine-tuned by the suprachiasmatic nucleus (SCN) as the master clock, which is essential for driving rhythms in food intake and energy metabolism through neuronal projections to the arcuate nucleus (ARC) and paraventricular nucleus (PVN). RESULTS We showed that chronic SD-induced aberrant expressions of AgRP/NPY and POMC attributed to compromised JAK/STAT3 signals and reduced energy expenditure in the mice, which can be rescued with AAV-genetic overexpression of BMAL1 into SCN. The potential mechanism may be related to the disruptions of SCN efferent mediated by BMAL1. CONCLUSIONS Chronic SD impairs energy balance through directly dampening BMAL1 expression, probably in the transcription level, in the SCN, which in turn affects the neuron projections to ARC and PVN. Remarkably, we provide evidence that may explain the causal mechanisms associated with sleep curtailment and obesity in adolescents.
Collapse
Affiliation(s)
- Tingting Du
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Shuailing Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Honghong Yu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Tian Hu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lina Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Lanyue Gao
- Experimental Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lihong Jia
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Jiajin Hu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qi Sun
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention Ministry of Education, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
5
|
Yang X, Qiu K, Jiang Y, Huang Y, Zhang Y, Liao Y. Metabolic Crosstalk between Liver and Brain: From Diseases to Mechanisms. Int J Mol Sci 2024; 25:7621. [PMID: 39062868 PMCID: PMC11277155 DOI: 10.3390/ijms25147621] [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: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver and brain is implicated in numerous metabolic and neurodegenerative disorders. Recent insights have positioned the liver not only as a central metabolic hub but also as an endocrine organ, capable of secreting hepatokines that transmit metabolic signals throughout the body via the bloodstream. Metabolites from the liver or gut microbiota also facilitate a complex dialogue between liver and brain. In parallel to humoral factors, the neural pathways, particularly the hypothalamic nuclei and autonomic nervous system, are pivotal in modulating the bilateral metabolic interplay between the cerebral and hepatic compartments. The term "liver-brain axis" vividly portrays this interaction. At the end of this review, we summarize cutting-edge technical advancements that have enabled the observation and manipulation of these signals, including genetic engineering, molecular tracing, and delivery technologies. These innovations are paving the way for a deeper understanding of the liver-brain axis and its role in metabolic homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Speksnijder EM, Bisschop PH, Siegelaar SE, Stenvers DJ, Kalsbeek A. Circadian desynchrony and glucose metabolism. J Pineal Res 2024; 76:e12956. [PMID: 38695262 DOI: 10.1111/jpi.12956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 05/09/2024]
Abstract
The circadian timing system controls glucose metabolism in a time-of-day dependent manner. In mammals, the circadian timing system consists of the main central clock in the bilateral suprachiasmatic nucleus (SCN) of the anterior hypothalamus and subordinate clocks in peripheral tissues. The oscillations produced by these different clocks with a period of approximately 24-h are generated by the transcriptional-translational feedback loops of a set of core clock genes. Glucose homeostasis is one of the daily rhythms controlled by this circadian timing system. The central pacemaker in the SCN controls glucose homeostasis through its neural projections to hypothalamic hubs that are in control of feeding behavior and energy metabolism. Using hormones such as adrenal glucocorticoids and melatonin and the autonomic nervous system, the SCN modulates critical processes such as glucose production and insulin sensitivity. Peripheral clocks in tissues, such as the liver, muscle, and adipose tissue serve to enhance and sustain these SCN signals. In the optimal situation all these clocks are synchronized and aligned with behavior and the environmental light/dark cycle. A negative impact on glucose metabolism becomes apparent when the internal timing system becomes disturbed, also known as circadian desynchrony or circadian misalignment. Circadian desynchrony may occur at several levels, as the mistiming of light exposure or sleep will especially affect the central clock, whereas mistiming of food intake or physical activity will especially involve the peripheral clocks. In this review, we will summarize the literature investigating the impact of circadian desynchrony on glucose metabolism and how it may result in the development of insulin resistance. In addition, we will discuss potential strategies aimed at reinstating circadian synchrony to improve insulin sensitivity and contribute to the prevention of type 2 diabetes.
Collapse
Affiliation(s)
- Esther M Speksnijder
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Sarah E Siegelaar
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism (AGEM), Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Tsay HJ, Gan YL, Su YH, Sun YY, Yao HH, Chen HW, Hsu YT, Hsu JTA, Wang HD, Shie FS. Reducing brain Aβ burden ameliorates high-fat diet-induced fatty liver disease in APP/PS1 mice. Biomed Pharmacother 2024; 173:116404. [PMID: 38471275 DOI: 10.1016/j.biopha.2024.116404] [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: 12/06/2023] [Revised: 02/18/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aβ clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aβ antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aβ burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aβ phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aβ burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aβ immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.
Collapse
Affiliation(s)
- Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Ling Gan
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Han Su
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Yo Sun
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Heng-Hsiang Yao
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hui-Wen Chen
- Institute of Neuroscience, School of Life Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Ying-Ting Hsu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - John Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC
| | - Horng-Dar Wang
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, ROC.
| |
Collapse
|
8
|
Pan S, A.C. Souza L, Worker CJ, Reyes Mendez ME, Gayban AJB, Cooper SG, Sanchez Solano A, Bergman RN, Stefanovski D, Morton GJ, Schwartz MW, Feng Earley Y. (Pro)renin receptor signaling in hypothalamic tyrosine hydroxylase neurons is required for obesity-associated glucose metabolic impairment. JCI Insight 2024; 9:e174294. [PMID: 38349753 PMCID: PMC11063935 DOI: 10.1172/jci.insight.174294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 02/08/2024] [Indexed: 03/06/2024] Open
Abstract
Glucose homeostasis is achieved via complex interactions between the endocrine pancreas and other peripheral tissues and glucoregulatory neurocircuits in the brain that remain incompletely defined. Within the brain, neurons in the hypothalamus appear to play a particularly important role. Consistent with this notion, we report evidence that (pro)renin receptor (PRR) signaling within a subset of tyrosine hydroxylase (TH) neurons located in the hypothalamic paraventricular nucleus (PVNTH neurons) is a physiological determinant of the defended blood glucose level. Specifically, we demonstrate that PRR deletion from PVNTH neurons restores normal glucose homeostasis in mice with diet-induced obesity (DIO). Conversely, chemogenetic inhibition of PVNTH neurons mimics the deleterious effect of DIO on glucose. Combined with our finding that PRR activation inhibits PVNTH neurons, these findings suggest that, in mice, (a) PVNTH neurons play a physiological role in glucose homeostasis, (b) PRR activation impairs glucose homeostasis by inhibiting these neurons, and (c) this mechanism plays a causal role in obesity-associated metabolic impairment.
Collapse
Affiliation(s)
- Shiyue Pan
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Lucas A.C. Souza
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Caleb J. Worker
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Miriam E. Reyes Mendez
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Ariana Julia B. Gayban
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Silvana G. Cooper
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Alfredo Sanchez Solano
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Darko Stefanovski
- New Bolton Center, School of Veterinary Medicine, University of Pennsylvania Philadelphia, Pennsylvania, USA
| | - Gregory J. Morton
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Michael W. Schwartz
- University of Washington Medicine Diabetes Institute, University of Washington, Seattle, Washington, USA
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology and
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, USA
| |
Collapse
|
9
|
Chen CC, Peng SJ, Chou YH, Lee CY, Lee PH, Hu RH, Ho MC, Chung MH, Hsiao FT, Tien YW, Tang SC. Human liver afferent and efferent nerves revealed by 3-D/Airyscan super-resolution imaging. Am J Physiol Endocrinol Metab 2024; 326:E107-E123. [PMID: 38170164 DOI: 10.1152/ajpendo.00205.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/31/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Neural regulation of hepatic metabolism has long been recognized. However, the detailed afferent and efferent innervation of the human liver has not been systematically characterized. This is largely due to the liver's high lipid and pigment contents, causing false-negative (light scattering and absorption) and false-positive (autofluorescence) results in in-depth fluorescence imaging. Here, to avoid the artifacts in three-dimensional (3-D) liver neurohistology, we embed the bleached human liver in the high-refractive-index polymer for tissue clearing and antifade 3-D/Airyscan super-resolution imaging. Importantly, using the paired substance P (SP, sensory marker) and PGP9.5 (pan-neuronal marker) labeling, we detect the sensory nerves in the portal space, featuring the SP+ varicosities in the PGP9.5+ nerve bundles/fibers, confirming the afferent liver innervation. Also, using the tyrosine hydroxylase (TH, sympathetic marker) labeling, we identify 1) condensed TH+ sympathetic nerves in the portal space, 2) extension of sympathetic nerves from the portal to the intralobular space, in which the TH+ nerve density is 2.6 ± 0.7-fold higher than that of the intralobular space in the human pancreas, and 3) the TH+ nerve fibers and varicosities contacting the ballooning cells, implicating potential sympathetic influence on hepatocytes with macrovesicular fatty change. Finally, using the vesicular acetylcholine transporter (VAChT, parasympathetic marker), PGP9.5, and CK19 (epithelial marker) labeling with panoramic-to-Airyscan super-resolution imaging, we detect and confirm the parasympathetic innervation of the septal bile duct. Overall, our labeling and 3-D/Airyscan imaging approach reveal the hepatic sensory (afferent) and sympathetic and parasympathetic (efferent) innervation, establishing a clinically related setting for high-resolution 3-D liver neurohistology.NEW & NOTEWORTHY We embed the human liver (vs. pancreas, positive control) in the high-refractive-index polymer for tissue clearing and antifade 3-D/Airyscan super-resolution neurohistology. The pancreas-liver comparison reveals: 1) sensory nerves in the hepatoportal space; 2) intralobular sympathetic innervation, including the nerve fibers and varicosities contacting the ballooning hepatocytes; and 3) parasympathetic innervation of the septal bile duct. Our results highlight the sensitivity and resolving power of 3-D/Airyscan super-resolution imaging in human liver neurohistology.
Collapse
Affiliation(s)
- Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Jung Peng
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hsien Chou
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Po-Huang Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Rey-Heng Hu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital-Yunlin Branch, Yunlin, Taiwan
| | - Ming-Chih Ho
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital-Hsinchu Branch, Hsinchu, Taiwan
| | - Mei-Hsin Chung
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Pathology, National Taiwan University Hospital-Hsinchu Branch, Hsinchu, Taiwan
| | - Fu-Ting Hsiao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Shiue-Cheng Tang
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
10
|
Yu Z, Han J, Li L, Zhang Q, Chen A, Chen J, Wang K, Jin J, Li H, Chen G. Chronic triclosan exposure induce impaired glucose tolerance by altering the gut microbiota. Food Chem Toxicol 2024; 183:114305. [PMID: 38052405 DOI: 10.1016/j.fct.2023.114305] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Triclosan (TCS) is an antimicrobial compound incorporated into more than 2000 consumer products. This compound is frequently detected in the human body and causes ubiquitous contamination in the environment, thereby raising concerns about its impact on human health and environmental pollution. Here, we demonstrated that 20 weeks' exposure of TCS drove the development of glucose intolerance by inducing compositional and functional alterations in intestinal microbiota in rats. Fecal-transplantation experiments corroborated the involvement of gut microbiota in TCS-induced glucose-tolerance impairment. 16S rRNA gene-sequencing analysis of cecal contents showed that TCS disrupted the gut microbiota composition in rats and increased the ratio of Firmicutes to Bacteroidetes. Cecal metabolomic analyses detected that TCS altered host metabolic pathways that are linked to host glucose and amino acid metabolism, particularly branched-chain amino acid (BCAA) biosynthesis. BCAA measurement confirmed the increase in serum BCAAs in rats exposed to TCS. Western blot and immunostaining results further confirmed that elevated BCAAs stimulated mTOR, a nutrient-sensing complex, and following IRS-1 serine phosphorylation, resulted in insulin resistance and glucose intolerance. These results suggested that TCS may induce glucose metabolism imbalance by regulating BCAA concentration by remodeling the gut microbiota.
Collapse
Affiliation(s)
- Zhen Yu
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Junyong Han
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Lisha Li
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Qiufeng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Ayun Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
| | - Jinyan Chen
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Kun Wang
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Jingjun Jin
- Fujian Provincial Key Laboratory of Medical Analysis, Fujian Academy of Medical Sciences, Fuzhou, 350001, China
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
| | - Gang Chen
- Department of Endocrinology, Fujian Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
11
|
Kim HR, Young CN. Circumventricular organ-hypothalamic circuit endoplasmic reticulum stress drives hepatic steatosis during obesity. Obesity (Silver Spring) 2024; 32:59-69. [PMID: 37794528 DOI: 10.1002/oby.23895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 10/06/2023]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD), characterized by excess liver triglyceride accumulation (hepatic steatosis), leads to an increased risk for cardiometabolic diseases and obesity-related mortality. Emerging evidence points to endoplasmic reticulum (ER) stress in the central nervous system as critical in NAFLD pathogenesis. Here, we tested the contribution of ER stress in a circumventricular organ-hypothalamic circuit in NAFLD development during obesity. METHODS C57BL/6J male mice were fed a high-fat diet (HFD) or normal chow. A combination of histological, viral tracing, intersectional viral targeting, and in vivo integrative physiological approaches were used to examine the role of ER stress in subfornical organ to hypothalamic paraventricular nucleus projecting neurons (SFO➔PVN) in NAFLD during diet-induced obesity. RESULTS Immunohistochemical analysis revealed marked unfolded protein response activation in the SFO, particularly in excitatory SFO➔PVN neurons of HFD-fed animals. Moreover, intersectional viral inhibition of ER stress in SFO➔PVN neurons resulted in a reduction in hepatomegaly, hepatic steatosis, and a blunted increase in body weight gain during diet-induced obesity, independent of changes in food intake, substrate partitioning, energy expenditure, and ambulatory activity. CONCLUSIONS These results indicate that ER stress in an SFO➔PVN neural circuit contributes to hepatic steatosis during obesity.
Collapse
Affiliation(s)
- Han Rae Kim
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Colin N Young
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
12
|
Sun M, Wan Y, Shi M, Meng ZX, Zeng W. Neural innervation in adipose tissue, gut, pancreas, and liver. LIFE METABOLISM 2023; 2:load022. [PMID: 39872245 PMCID: PMC11749697 DOI: 10.1093/lifemeta/load022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 01/30/2025]
Abstract
Efficient communication between the brain and peripheral organs is indispensable for regulating physiological function and maintaining energy homeostasis. The peripheral nervous system (PNS) in vertebrates, consisting of the autonomic and somatic nervous systems, bridges the peripheral organs and the central nervous system (CNS). Metabolic signals are processed by both vagal sensory nerves and somatosensory nerves. The CNS receives sensory inputs via ascending nerves, serves as the coordination and integration center, and subsequently controls internal organs and glands via descending nerves. The autonomic nervous system consists of sympathetic and parasympathetic branches that project peripheral nerves into various anatomical locations to regulate the energy balance. Sympathetic and parasympathetic nerves typically control the reflexive and involuntary functions in organs. In this review article, we outline the innervation of adipose tissue, gut, pancreas, and liver, to illustrate the neurobiological basis of central-peripheral interactions. We emphasize the importance of understanding the functional atlas of neural control of energy metabolism, and more importantly, provide potential avenues for further research in this area.
Collapse
Affiliation(s)
- Mengxue Sun
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yongwen Wan
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Mengjie Shi
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
13
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [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: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
14
|
Molinas AJR, Desmoulins LD, Davis RK, Gao H, Satou R, Derbenev AV, Zsombok A. High-Fat Diet Modulates the Excitability of Neurons within the Brain-Liver Pathway. Cells 2023; 12:1194. [PMID: 37190103 PMCID: PMC10137256 DOI: 10.3390/cells12081194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
Stimulation of hepatic sympathetic nerves increases glucose production and glycogenolysis. Activity of pre-sympathetic neurons in the paraventricular nucleus (PVN) of the hypothalamus and in the ventrolateral and ventromedial medulla (VLM/VMM) largely influence the sympathetic output. Increased activity of the sympathetic nervous system (SNS) plays a role in the development and progression of metabolic diseases; however, despite the importance of the central circuits, the excitability of pre-sympathetic liver-related neurons remains to be determined. Here, we tested the hypothesis that the activity of liver-related neurons in the PVN and VLM/VMM is altered in diet-induced obese mice, as well as their response to insulin. Patch-clamp recordings were conducted from liver-related PVN neurons, VLM-projecting PVN neurons, and pre-sympathetic liver-related neurons in the ventral brainstem. Our data demonstrate that the excitability of liver-related PVN neurons increased in high-fat diet (HFD)-fed mice compared to mice fed with control diet. Insulin receptor expression was detected in a population of liver-related neurons, and insulin suppressed the firing activity of liver-related PVN and pre-sympathetic VLM/VMM neurons in HFD mice; however, it did not affect VLM-projecting liver-related PVN neurons. These findings further suggest that HFD alters the excitability of pre-autonomic neurons as well as their response to insulin.
Collapse
Affiliation(s)
- Adrien J. R. Molinas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
| | - Lucie D. Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
| | - Roslyn K. Davis
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
| | - Hong Gao
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
| | - Ryousuke Satou
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
| | - Andrei V. Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
- Tulane Brain Institute, Tulane University, New Orleans, LA 70130, USA
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70130, USA; (A.J.R.M.); (L.D.D.); (R.K.D.); (R.S.); (A.V.D.)
- Tulane Brain Institute, Tulane University, New Orleans, LA 70130, USA
| |
Collapse
|
15
|
Jiang Y, Rezai-Zadeh K, Desmoulins LD, Muenzberg H, Derbenev AV, Zsombok A. GABAergic leptin receptor-expressing neurons in the dorsomedial hypothalamus project to brown adipose tissue-related neurons in the paraventricular nucleus of mice. Auton Neurosci 2023; 245:103058. [PMID: 36538864 PMCID: PMC9899324 DOI: 10.1016/j.autneu.2022.103058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/14/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Brown adipose tissue (BAT) contributes to energy homeostasis via nonshivering thermogenesis. The BAT is densely innervated by the sympathetic nervous system (SNS) and activity of pre-autonomic neurons modulates the sympathetic outflow. Leptin, an adipocyte hormone, alters energy homeostasis and thermogenesis of BAT via several neuronal circuits; however, the cellular effects of leptin on interscapular BAT (iBAT)-related neurons in the hypothalamus remain to be determined. In this study, we used pseudorabies virus (PRV) to identify iBAT-related neurons in the paraventricular nucleus (PVN) of the hypothalamus and test the hypothesis that iBAT-related PVN neurons are modulated by leptin. Inoculation of iBAT with PRV in leptin receptor reporter mice (Lepr:EGFP) demonstrated that a population of iBAT-related PVN neurons expresses Lepr receptors. Our electrophysiological findings revealed that leptin application caused hyperpolarization in some of iBAT-related PVN neurons. Bath application of leptin also modulated excitatory and inhibitory neurotransmission to most of iBAT-related PVN neurons. Using channel rhodopsin assisted circuit mapping we found that GABAergic and glutamatergic Lepr-expressing neurons in the dorsomedial hypothalamus/dorsal hypothalamic area (dDMH/DHA) project to PVN neurons; however, connected iBAT-related PVN neurons receive exclusively inhibitory signals from Lepr-expressing dDMH/DHA neurons.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Kavon Rezai-Zadeh
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America
| | - Heike Muenzberg
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America.
| |
Collapse
|
16
|
Zhou L, Han D, Wang X, Chen Z. Probiotic Formulation VSL#3 Interacts with Mesenchymal Stromal Cells To Protect Dopaminergic Neurons via Centrally and Peripherally Suppressing NOD-Like Receptor Protein 3 Inflammasome-Mediated Inflammation in Parkinson's Disease Mice. Microbiol Spectr 2023; 11:e0320822. [PMID: 36728426 PMCID: PMC10100967 DOI: 10.1128/spectrum.03208-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/08/2023] [Indexed: 02/03/2023] Open
Abstract
Systemic immunomodulation is increasingly recognized among the beneficial effects of mesenchymal stromal cells (MSCs) in treatment of Parkinson's disease (PD), while the underlying mechanism is not fully understood. With the growing popularity of using probiotics as an adjuvant approach in PD treatment, concerns about the added effects of probiotics have been raised. In addition to the molecular mechanism mediating the neuroprotective effects of MSCs, the combined effects of a probiotic formulation, VSL#3, and MSC infusion were also evaluated in PD mice. The animals were weekly treated with human MSCs (hMSCs) via the tail vein, VSL#3 via the gastrointestinal tract, or their combination six times. hMSCs, VSL#3 alone, and their combination markedly ameliorated the decreased striatal dopamine content, loss of dopaminergic neurons in the substantia nigra, increased levels of proinflammatory cytokines in serum, as well as tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β) mRNAs in striatum and peripheral tissues induced by MPTP. Furthermore, hMSCs, VSL#3, and their combination notably downregulated mRNA expression of NOD-like receptor protein 3 (NLRP3) and caspase-1 in brain and peripheral tissues of PD mice. These results suggest that hMSCs, VSL#3, and their combination prevent neurodegenerative changes in PD mice via anti-inflammatory activities in both the central and peripheral systems, possibly through suppressing the NLRP3 inflammasome. Moreover, two-way analysis of variance (ANOVA) indicated that VSL#3 interacts with hMSCs to attenuate neurodegeneration and inhibit NLRP3 inflammasome-mediated inflammation without altering the effects of hMSCs. Major findings of our study support the usage of probiotic formulation VSL#3 as an adjuvant therapy to hMSC infusion in PD treatment. IMPORTANCE This study provides evidence for the neuroprotective activities of human umbilical cord MSCs from the aspect of anti-inflammation actions. hMSCs inhibit the NLRP3 inflammasome and MPTP-induced inflammation in both brain and periphery to relieve the degenerative changes in dopaminergic neurons in PD mice. Furthermore, as an additional therapeutic agent, probiotic formulation VSL#3 interacts with hMSCs in suppressing the NLRP3 inflammasome as well as the central and peripheral anti-inflammatory effects to exert neuroprotective actions in PD mice without altering the actions of hMSCs, suggesting the potential of VSL#3 as an adjuvant therapy in PD treatment. The findings of the present study give a further understanding of the anti-inflammatory activity and the molecular mechanism for the beneficial effects of MSCs as well as the potential application of probiotic formulation as an adjuvant approach to MSC therapy in PD treatment.
Collapse
Affiliation(s)
- Liping Zhou
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Deqiang Han
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Xingzhe Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson’s Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
17
|
Blackmore K, Young CN. Central Feminization of Obese Male Mice Reduces Metabolic Syndrome. Brain Sci 2022; 12:1324. [PMID: 36291259 PMCID: PMC9599293 DOI: 10.3390/brainsci12101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 11/16/2022] Open
Abstract
Metabolic syndrome encompasses a spectrum of conditions that increases the risk for cardiovascular and metabolic diseases. It is widely accepted that the sex hormone estrogen plays a protective metabolic role in premenopausal women, in part through central nervous system (CNS) mechanisms. However, most work to date has focused on the loss of estrogen in females (e.g., menopause). Interestingly, transgender individuals receiving feminizing gender affirming therapy (i.e., estrogen) are relatively protected from metabolic syndrome conditions, pointing to a role for CNS estrogen in the development of metabolic syndrome in men. Here, we show that estrogen signaling in the brain protects males from metabolic syndrome and obesity related complications. First, short-term CNS specific supplementation of low-dose 17-β-estradiol in diet-induced obese male mice resulted in a significant reduction in body weight in parallel with a decrease in food intake without alterations in energy expenditure. In conjunction, central supplementation of estrogen reduced visceral adiposity, including epididymal and abdominal regions, with slighter decreases in subcutaneous inguinal and thermogenic brown adipose tissue. Furthermore, central estrogen administration reduced the liver manifestation of metabolic syndrome including hepatomegaly and hepatic steatosis. Collectively, these findings indicate that a lack of estrogen action in the brain may predispose males to metabolic syndrome pathogenesis.
Collapse
Affiliation(s)
- Katherine Blackmore
- School of Medicine and Health Sciences, George Washington University, 2300 I Street NW, Washington, DC 20037, USA
| | - Colin N. Young
- School of Medicine and Health Sciences, George Washington University, 2300 I Street NW, Washington, DC 20037, USA
| |
Collapse
|
18
|
Neuroprotective effects of human umbilical cord mesenchymal stromal cells in PD mice via centrally and peripherally suppressing NLRP3 inflammasome-mediated inflammatory responses. Biomed Pharmacother 2022; 153:113535. [DOI: 10.1016/j.biopha.2022.113535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/20/2022] Open
|
19
|
Cotero V, Graf J, Miwa H, Hirschstein Z, Qanud K, Huerta TS, Tai N, Ding Y, Jimenez-Cowell K, Tomaio JN, Song W, Devarajan A, Tsaava T, Madhavan R, Wallace K, Loghin E, Morton C, Fan Y, Kao TJ, Akhtar K, Damaraju M, Barenboim L, Maietta T, Ashe J, Tracey KJ, Coleman TR, Di Carlo D, Shin D, Zanos S, Chavan SS, Herzog RI, Puleo C. Stimulation of the hepatoportal nerve plexus with focused ultrasound restores glucose homoeostasis in diabetic mice, rats and swine. Nat Biomed Eng 2022; 6:683-705. [PMID: 35361935 PMCID: PMC10127248 DOI: 10.1038/s41551-022-00870-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
Abstract
Peripheral neurons that sense glucose relay signals of glucose availability to integrative clusters of neurons in the brain. However, the roles of such signalling pathways in the maintenance of glucose homoeostasis and their contribution to disease are unknown. Here we show that the selective activation of the nerve plexus of the hepatic portal system via peripheral focused ultrasound stimulation (pFUS) improves glucose homoeostasis in mice and rats with insulin-resistant diabetes and in swine subject to hyperinsulinemic-euglycaemic clamps. pFUS modulated the activity of sensory projections to the hypothalamus, altered the concentrations of metabolism-regulating neurotransmitters, and enhanced glucose tolerance and utilization in the three species, whereas physical transection or chemical blocking of the liver-brain nerve pathway abolished the effect of pFUS on glucose tolerance. Longitudinal multi-omic profiling of metabolic tissues from the treated animals confirmed pFUS-induced modifications of key metabolic functions in liver, pancreas, muscle, adipose, kidney and intestinal tissues. Non-invasive ultrasound activation of afferent autonomic nerves may represent a non-pharmacologic therapy for the restoration of glucose homoeostasis in type-2 diabetes and other metabolic diseases.
Collapse
Affiliation(s)
- Victoria Cotero
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - John Graf
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Hiromi Miwa
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Khaled Qanud
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tomás S Huerta
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Yuyan Ding
- Yale School of Medicine, New Haven, CT, USA
| | - Kevin Jimenez-Cowell
- Yale School of Medicine, New Haven, CT, USA
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Weiguo Song
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Alex Devarajan
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Tea Tsaava
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Radhika Madhavan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kirk Wallace
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Evelina Loghin
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Christine Morton
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Ying Fan
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Tzu-Jen Kao
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | | | | | | | | | - Jeffrey Ashe
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | - Dino Di Carlo
- University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Stavros Zanos
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | - Chris Puleo
- General Electric (GE) Research, 1 Research Circle, Niskayuna, NY, USA.
| |
Collapse
|
20
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
21
|
Torres H, Huesing C, Burk DH, Molinas AJR, Neuhuber WL, Berthoud HR, Münzberg H, Derbenev AV, Zsombok A. Sympathetic innervation of the mouse kidney and liver arising from prevertebral ganglia. Am J Physiol Regul Integr Comp Physiol 2021; 321:R328-R337. [PMID: 34231420 PMCID: PMC8530761 DOI: 10.1152/ajpregu.00079.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/11/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
The sympathetic nervous system (SNS) plays a crucial role in the regulation of renal and hepatic functions. Although sympathetic nerves to the kidney and liver have been identified in many species, specific details are lacking in the mouse. In the absence of detailed information of sympathetic prevertebral innervation of specific organs, selective manipulation of a specific function will remain challenging. Despite providing major postganglionic inputs to abdominal organs, limited data are available about the mouse celiac-superior mesenteric complex. We used tyrosine hydroxylase (TH) and dopamine β-hydroxylase (DbH) reporter mice to visualize abdominal prevertebral ganglia. We found that both the TH and DbH reporter mice are useful models for identification of ganglia and nerve bundles. We further tested if the celiac-superior mesenteric complex provides differential inputs to the mouse kidney and liver. The retrograde viral tracer, pseudorabies virus (PRV)-152 was injected into the cortex of the left kidney or the main lobe of the liver to identify kidney-projecting and liver-projecting neurons in the celiac-superior mesenteric complex. iDISCO immunostaining and tissue clearing were used to visualize unprecedented anatomical detail of kidney-related and liver-related postganglionic neurons in the celiac-superior mesenteric complex and aorticorenal and suprarenal ganglia compared with TH-positive neurons. Kidney-projecting neurons were restricted to the suprarenal and aorticorenal ganglia, whereas only sparse labeling was observed in the celiac-superior mesenteric complex. In contrast, liver-projecting postganglionic neurons were observed in the celiac-superior mesenteric complex and aorticorenal and suprarenal ganglia, suggesting spatial separation between the sympathetic innervation of the mouse kidney and liver.
Collapse
Affiliation(s)
- Hayden Torres
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Clara Huesing
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - David H Burk
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Adrien J R Molinas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | | | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Heike Münzberg
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
- Brain Institute, Tulane University, New Orleans, Louisiana
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
- Brain Institute, Tulane University, New Orleans, Louisiana
| |
Collapse
|
22
|
Dos Santos KM, Moraes DJDA, da Silva MP, Antunes VR. Exercise training rescues the electrical activity of liver-projecting DMNV neurones in response to oxytocin in spontaneously hypertensive rats. J Neuroendocrinol 2021; 33:e12977. [PMID: 33942389 DOI: 10.1111/jne.12977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
A neural circuit between the paraventricular nucleus of the hypothalamus (PVN) and the dorsal motor nucleus of the vagus (DMNV) constitutes part of an important parasympathetic autonomic pathway that controls hepatic glucose production. Intracerebroventricular injection of insulin activates oxytocinergic neurones in the PVN and elicits the release of oxytocin into the circulation, which plays an important role in the metabolism of glucose. Moreover, the central action of insulin can reduce the concentration of glucose in blood taken from the hepatic vein of Wistar rats via activation of vagal efferent nerves to the liver. This mechanism is impaired in sedentary spontaneously hypertensive rats (SHR). Because aerobic exercise increases vagal tone, partly mediated by increasing the oxytocinergic connections between the PVN and DMNV, we hypothesised that oxytocin (OT) might alter the excitability of liver-projecting DMNV neurones. Thus, we investigated the effects of OT on electrical properties of the liver-projecting DMNV neurones from Wistar, SHR subjected to 4 weeks of exercise training, as well sedentary controls, using whole cell patch-clamping. The results show that OT increased the resting membrane potential of DMNV neurones in Wistar rats, as well as the firing frequency of these cells, but not in sedentary SHR. However, in SHR subjected to 4 weeks of exercise training, the effects of OT on liver-projecting DMNV neurones of were similar to those seen in Wistar rats. These findings show that OT elicits similar changes in the electrophysiological properties of liver-projecting DMNV neurones of Wistar and exercise-trained but not sedentary SHR. These results indicate that exercise training can restore the sensitivity of liver-projecting DMNV neurones of exercise-trained SHR to OT.
Collapse
Affiliation(s)
- Karoline Martins Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Davi José de Almeida Moraes
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Melina Pires da Silva
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vagner Roberto Antunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
23
|
Luo S, Ezrokhi M, Cominos N, Tsai TH, Stoelzel CR, Trubitsyna Y, Cincotta AH. Experimental dopaminergic neuron lesion at the area of the biological clock pacemaker, suprachiasmatic nuclei (SCN) induces metabolic syndrome in rats. Diabetol Metab Syndr 2021; 13:11. [PMID: 33485386 PMCID: PMC7825247 DOI: 10.1186/s13098-021-00630-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The daily peak in dopaminergic neuronal activity at the area of the biological clock (hypothalamic suprachiasmatic nuclei [SCN]) is diminished in obese/insulin resistant vs lean/insulin sensitive animals. The impact of targeted lesioning of dopamine (DA) neurons specifically at the area surrounding (and that communicate with) the SCN (but not within the SCN itself) upon glucose metabolism, adipose and liver lipid gene expression, and cardiovascular biology in normal laboratory animals has not been investigated and was the focus of this study. METHODS Female Sprague-Dawley rats received either DA neuron neurotoxic lesion by bilateral intra-cannula injection of 6-hydroxydopamine (2-4 μg/side) or vehicle treatment at the area surrounding the SCN at 20 min post protriptyline ip injection (20 mg/kg) to protect against damage to noradrenergic and serotonergic neurons. RESULTS At 16 weeks post-lesion relative to vehicle treatment, peri-SCN area DA neuron lesioning increased weight gain (34.8%, P < 0.005), parametrial and retroperitoneal fat weight (45% and 90% respectively, P < 0.05), fasting plasma insulin, leptin and norepinephrine levels (180%, 71%, and 40% respectively, P < 0.05), glucose tolerance test area under the curve (AUC) insulin (112.5%, P < 0.05), and insulin resistance (44%-Matsuda Index, P < 0.05) without altering food consumption during the test period. Such lesion also induced the expression of several lipid synthesis genes in adipose and liver and the adipose lipolytic gene, hormone sensitive lipase in adipose (P < 0.05 for all). Liver monocyte chemoattractant protein 1 (a proinflammatory protein associated with metabolic syndrome) gene expression was also significantly elevated in peri-SCN area dopaminergic lesioned rats. Peri-SCN area dopaminergic neuron lesioned rats were also hypertensive (systolic BP rose from 157 ± 5 to 175 ± 5 mmHg, P < 0.01; diastolic BP rose from 109 ± 4 to 120 ± 3 mmHg, P < 0.05 and heart rate increase from 368 ± 12 to 406 ± 12 BPM, P < 0.05) and had elevated plasma norepinephrine levels (40% increased, P < 0.05) relative to controls. CONCLUSIONS These findings indicate that reduced dopaminergic neuronal activity in neurons at the area of and communicating with the SCN contributes significantly to increased sympathetic tone and the development of metabolic syndrome, without effect on feeding.
Collapse
Affiliation(s)
- Shuqin Luo
- VeroScience LLC, 1334 Main Road, Tiverton, RI, 02878, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Kwon E, Joung HY, Liu SM, Chua SC, Schwartz GJ, Jo YH. Optogenetic stimulation of the liver-projecting melanocortinergic pathway promotes hepatic glucose production. Nat Commun 2020; 11:6295. [PMID: 33293550 PMCID: PMC7722761 DOI: 10.1038/s41467-020-20160-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 11/18/2020] [Indexed: 12/18/2022] Open
Abstract
The central melanocortin system plays a fundamental role in the control of feeding and body weight. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) also regulate overall glucose homeostasis via insulin-dependent and -independent pathways. Here, we report that a subset of ARC POMC neurons innervate the liver via preganglionic parasympathetic acetylcholine (ACh) neurons in the dorsal motor nucleus of the vagus (DMV). Optogenetic stimulation of this liver-projecting melanocortinergic pathway elevates blood glucose levels that is associated with increased expression of hepatic gluconeogenic enzymes in female and male mice. Pharmacological blockade and knockdown of the melanocortin-4 receptor gene in the DMV abolish this stimulation-induced effect. Activation of melanocortin-4 receptors inhibits DMV cholinergic neurons and optogenetic inhibition of liver-projecting parasympathetic cholinergic fibers increases blood glucose levels. This elevated blood glucose is not due to altered pancreatic hormone release. Interestingly, insulin-induced hypoglycemia increases ARC POMC neuron activity. Hence, this liver-projecting melanocortinergic circuit that we identified may play a critical role in the counterregulatory response to hypoglycemia.
Collapse
Affiliation(s)
- Eunjin Kwon
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hye-Young Joung
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shun-Mei Liu
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Streamson C Chua
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary J Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA.
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
25
|
Wang J, Liang XF, He S, Zhang YP, Li J, Huang K, Shi LJ, Ren P. Valine acts as a nutritional signal in brain to activate TORC1 and attenuate postprandial ammonia-N excretion in Chinese perch (Siniperca chuatsi). FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:2015-2025. [PMID: 32749664 DOI: 10.1007/s10695-020-00767-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/13/2020] [Indexed: 06/11/2023]
Abstract
An emerging concept is that the hypothalamic nutrient sensor can regulate peripheral energy metabolism via a brain-liver circuit. Valine is an essential branched-chain amino acid (BCAA) that drives intracellular signaling cascades by the activation of target of rapamycin complex 1 (TORC1) which is critical to protein metabolism in mammals. However, in teleost fish, it remains scarce in this area especially about how the intraventricular (ICV) injection of valine can mediate the protein metabolism in peripheral organs. This study would tentatively explore the effects of ICV injection of valine on protein metabolism in peripheral organs through evaluating the postprandial ammonia-N excretion rate in Chinese perch. The control group was injected with 5-μL PBS, and the Val group was injected with 20-μg L valine dissolved into 5-μL PBS. The ammonia-N excretion rate of Val group was lower than control group at 4-, 12-, and 24-h postinjection, while the concertation of plasma glucose was increased sharply at 0.5-, 4-, 12-, and 24-h postinjection. We further checked both mRNA level and the enzyme activity of glutamate dehydrogenase (GDH) in the liver and adenosine monophosphate deaminase (AMPD) in muscle, and we found that they were obviously decreased in Val group at 4-, 12-, and 24-h postinjection. The phosphorylation level of ribosomal protein S6, a downstream target protein of TORC1, was markedly enhanced in the liver of Val group at 4-, 12-, and 24-h postinjection. Collectively, these results illustrated that ICV injection of valine can attenuate protein degradation in peripheral organs by depressing the GDH and AMPD enzyme activity; on the other hand, the injected valine can trigger the activation of TORC1 in the liver via a brain-liver circuit and then interdict proteolysis.
Collapse
Affiliation(s)
- Jie Wang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China.
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China.
| | - Shan He
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Yan-Peng Zhang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Jiao Li
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Kang Huang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Lin-Jie Shi
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| | - Ping Ren
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, China
- Innovation Base for Chinese Perch Breeding, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, China
| |
Collapse
|
26
|
Bruce KD, Dobrinskikh E, Wang H, Rudenko I, Gao H, Libby AE, Gorkhali S, Yu T, Zsombok A, Eckel RH. Neuronal Lipoprotein Lipase Deficiency Alters Neuronal Function and Hepatic Metabolism. Metabolites 2020; 10:metabo10100385. [PMID: 32998280 PMCID: PMC7600143 DOI: 10.3390/metabo10100385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/21/2020] [Indexed: 12/31/2022] Open
Abstract
The autonomic regulation of hepatic metabolism offers a novel target for the treatment of non-alcoholic fatty liver disease (NAFLD). However, the molecular characteristics of neurons that regulate the brain-liver axis remain unclear. Since mice lacking neuronal lipoprotein lipase (LPL) develop perturbations in neuronal lipid-sensing and systemic energy balance, we reasoned that LPL might be a component of pre-autonomic neurons involved in the regulation of hepatic metabolism. Here, we show that, despite obesity, mice with reduced neuronal LPL (NEXCreLPLflox (LPL KD)) show improved glucose tolerance and reduced hepatic lipid accumulation with aging compared to wilt type (WT) controls (LPLflox). To determine the effect of LPL deficiency on neuronal physiology, liver-related neurons were identified in the paraventricular nucleus (PVN) of the hypothalamus using the transsynaptic retrograde tracer PRV-152. Patch-clamp studies revealed reduced inhibitory post-synaptic currents in liver-related neurons of LPL KD mice. Fluorescence lifetime imaging microscopy (FLIM) was used to visualize metabolic changes in LPL-depleted neurons. Quantification of free vs. bound nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) revealed increased glucose utilization and TCA cycle flux in LPL-depleted neurons compared to controls. Global metabolomics from hypothalamic cell lines either deficient in or over-expressing LPL recapitulated these findings. Our data suggest that LPL is a novel feature of liver-related preautonomic neurons in the PVN. Moreover, LPL loss is sufficient to cause changes in neuronal substrate utilization and function, which may precede changes in hepatic metabolism.
Collapse
Affiliation(s)
- Kimberley D. Bruce
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
- Correspondence:
| | - Evgenia Dobrinskikh
- Department of Medicine, University of Colorado, Denver Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Hong Wang
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
| | - Ivan Rudenko
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
| | - Hong Gao
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70112, USA; (H.G.); (A.Z.)
| | - Andrew E. Libby
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Sachi Gorkhali
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
| | - Tian Yu
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA 70112, USA; (H.G.); (A.Z.)
| | - Robert H. Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Denver Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (H.W.); (I.R.); (S.G.); (T.Y.); (R.H.E.)
| |
Collapse
|
27
|
Méndez-Hernández R, Escobar C, Buijs RM. Suprachiasmatic Nucleus-Arcuate Nucleus Axis: Interaction Between Time and Metabolism Essential for Health. Obesity (Silver Spring) 2020; 28 Suppl 1:S10-S17. [PMID: 32538539 DOI: 10.1002/oby.22774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
In mammals, time and metabolism are tightly coupled variables; this relationship can be illustrated by numerous examples, such as the circadian variation in food intake or the circadian response to a glucose bolus. We review evidence that the interaction between the suprachiasmatic nucleus and the arcuate nucleus plays a key role in the execution of these functions. The nuclei are reciprocally connected via different projections, and this interaction provides an ideal anatomical framework to modify the temporal output of the hypothalamus to metabolic organs as a consequence of the feedback from the periphery. The suprachiasmatic nucleus-arcuate nucleus relationship is essential to integrate metabolic information into the circadian system and thus adapt circadian rhythms in core body temperature, locomotor activity, food intake, and circulating molecules such as glucose and corticosterone. With the rise in obesity-associated diseases in the world population, gaining knowledge about this relationship, and the consequences of disturbing this liaison, is essential to understand the pathogenesis of obesity.
Collapse
Affiliation(s)
- Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Carolina Escobar
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Ruud M Buijs
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
28
|
Yosten GLC. AJP-Regulatory, Integrative and Comparative Physiology: Looking Toward the Future. Am J Physiol Regul Integr Comp Physiol 2020; 319:R82-R86. [DOI: 10.1152/ajpregu.00104.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Parker CG, Dailey MJ, Phillips H, Davis EA. Central sensory-motor crosstalk in the neural gut-brain axis. Auton Neurosci 2020; 225:102656. [PMID: 32151980 DOI: 10.1016/j.autneu.2020.102656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/27/2020] [Accepted: 02/14/2020] [Indexed: 12/15/2022]
Abstract
The neural gut-brain axis consists of viscerosensory and autonomic motor neurons innervating the gastrointestinal (GI) tract. Sensory neurons transmit nutrient-related and non-nutrient-related information to the brain, while motor neurons regulate GI motility and secretion. Previous research provides an incomplete picture of the brain nuclei that are directly connected with the neural gut-brain axis, and no studies have thoroughly assessed sensory-motor overlap in those nuclei. Our goal in this study was to comprehensively characterize the central sensory and motor circuitry associated with the neural gut-brain axis linked to a segment of the small intestine. We injected a retrograde (pseudorabies; PRV) and anterograde (herpes simplex virus 1; HSV) transsynaptic viral tracer into the duodenal wall of adult male rats. Immunohistochemical processing revealed single- and double-labeled cells that were quantified per nucleus. We found that across nearly all brain regions assessed, PRV + HSV immunoreactive neurons comprised the greatest percentage of labeled cells compared with single-labeled PRV or HSV neurons. These results indicate that even though sensory and motor information can be processed by separate neuronal populations, there is neuroanatomical evidence of direct sensory-motor feedback in the neural gut-brain axis throughout the entire caudal-rostral extent of the brain. This is the first study to exhaustively investigate the sensory-motor organization of the neural gut-brain axis, and is a step toward phenotyping the many central neuronal populations involved in GI control.
Collapse
Affiliation(s)
- Coltan G Parker
- Neuroscience Program, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana 61801, IL, USA
| | - Megan J Dailey
- Neuroscience Program, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana 61801, IL, USA; Department of Food Sciences and Human Nutrition, University of Illinois at Urbana-Champaign, 905 South Goodwin Avenue, Urbana 61801, IL, USA
| | - Heidi Phillips
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 2001 S Lincoln Avenue, Urbana 61802, IL, USA
| | - Elizabeth A Davis
- Neuroscience Program, University of Illinois at Urbana-Champaign, 405 North Mathews Avenue, Urbana 61801, IL, USA
| |
Collapse
|
30
|
Caba M, Huerta C, Meza E, Hernández M, Rovirosa-Hernández MJ. Oxytocinergic Cells of the Hypothalamic Paraventricular Nucleus Are Involved in Food Entrainment. Front Neurosci 2020; 14:49. [PMID: 32082116 PMCID: PMC7005215 DOI: 10.3389/fnins.2020.00049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022] Open
Abstract
When food is presented at a specific time of day subjects develop intense locomotor behavior before food presentation, termed food anticipatory activity (FAA). Metabolic and hormonal parameters, as well as neural structures also shift their rhythm according to mealtime. Food-entrained activity rhythms are thought to be driven by a distributed system of central and peripheral oscillators sensitive to food cues, but it is not well understood how they are organized for the expression of FAA. The hormone Oxytocin plays an important role in food intake, satiety and homeostatic glucose metabolism and although it is recognized that food is the main cue for food entrainment this hormone has not been implicated in FAA. Here we investigated the activity of oxytocinergic (OTergic) cells of the hypothalamus in relation to the timing of feeding in rabbit pups, a natural model of food entrainment. We found that OTergic cells of the supraoptic nucleus and the main body of the paraventricular nucleus (PVN) are activated after feeding which suggests that OT may be an entraining signal for food synchronization. Moreover, a detailed analysis of the PVN revealed that OTergic cells of the caudal PVN and a subpopulation in the dorsal part of the main body of this nucleus shows activation before the time of food but not 12 h later. Moreover this pattern persists in fasted subjects at the time of the previous scheduled time of nursing. The fact that those OTergic cells of the dorsal and caudal part of the PVN contain preautonomic cells that project to the adrenal, pancreas and liver perhaps may be related to the physiological changes in preparation for food ingestion, and synchronization of peripheral oscillators, which remains to be determined; perhaps they play a main role in the central oscillatory mechanism of FAA as their activity persists in fasted subjects at the time of the next feeding time.
Collapse
Affiliation(s)
- Mario Caba
- Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Mexico
| | - César Huerta
- Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Mexico
| | - Enrique Meza
- Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Mexico
| | - Manuel Hernández
- Doctorado en Ciencias Biomédicas, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Mexico
| | | |
Collapse
|
31
|
Influence of intranasal exposure of MPTP in multiple doses on liver functions and transition from non-motor to motor symptoms in a rat PD model. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:147-165. [PMID: 31468077 DOI: 10.1007/s00210-019-01715-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/15/2019] [Indexed: 12/14/2022]
Abstract
Besides the effects on the striatum, the impairment of visceral organs including liver functions has been reported in Parkinson's disease (PD) patients. However, it is yet unclear if liver functions are affected in the early stage of the disease before the motor phase has appeared. The aim of our present study was thus to assess the effect of intranasal administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in different doses on striatum and liver functions. Deterioration of non-motor activities appeared on single exposure to MPTP along with rise in striatum oxidative stress and decline in antioxidant levels. Decreases in dopamine, noradrenaline, and GABA and increase in serotonin were detected in striatum. Motor coordination was impaired with a single dose of MPTP, and with repeated MPTP exposure, there was further significant impairment. Locomotor activity was affected from second exposure of MPTP, and the impairment increased with third MPTP exposure. Impairment of liver function through increase in serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels was observed after first MPTP insult, and it worsened with second and third administrations. First administration of MPTP triggered systemic inflammation showing significant increase in inflammatory markers in the liver. Our data shows for the first time that an intranasal route of entry of MPTP affects liver from the non-motor phase of PD itself, occurring concomitantly with the reduction of striatal dopamine. It also suggests that a single dose is not enough to bring about progression of the disease from non-motor to locomotor deficiency, and a repeated dose is needed to establish the motor severity phase in the rat intranasal MPTP model.
Collapse
|
32
|
Noninvasive sub-organ ultrasound stimulation for targeted neuromodulation. Nat Commun 2019; 10:952. [PMID: 30862827 PMCID: PMC6414607 DOI: 10.1038/s41467-019-08750-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 01/22/2019] [Indexed: 12/20/2022] Open
Abstract
Tools for noninvasively modulating neural signaling in peripheral organs will advance the study of nerves and their effect on homeostasis and disease. Herein, we demonstrate a noninvasive method to modulate specific signaling pathways within organs using ultrasound (U/S). U/S is first applied to spleen to modulate the cholinergic anti-inflammatory pathway (CAP), and US stimulation is shown to reduce cytokine response to endotoxin to the same levels as implant-based vagus nerve stimulation (VNS). Next, hepatic U/S stimulation is shown to modulate pathways that regulate blood glucose and is as effective as VNS in suppressing the hyperglycemic effect of endotoxin exposure. This response to hepatic U/S is only found when targeting specific sub-organ locations known to contain glucose sensory neurons, and both molecular (i.e. neurotransmitter concentration and cFOS expression) and neuroimaging results indicate US induced signaling to metabolism-related hypothalamic sub-nuclei. These data demonstrate that U/S stimulation within organs provides a new method for site-selective neuromodulation to regulate specific physiological functions. Stimulation of peripheral nerve activity may be used to treat metabolic and inflammatory disorders, but current approaches need implanted devices. Here, the authors present a non-invasive approach, and show that ultrasound-mediated stimulation can be targeted to specific sub-organ locations in preclinical models and alter the response of metabolic and inflammatory neural pathways.
Collapse
|
33
|
Feetham CH, O'Brien F, Barrett-Jolley R. Ion Channels in the Paraventricular Hypothalamic Nucleus (PVN); Emerging Diversity and Functional Roles. Front Physiol 2018; 9:760. [PMID: 30034342 PMCID: PMC6043726 DOI: 10.3389/fphys.2018.00760] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVN) is critical for the regulation of homeostatic function. Although also important for endocrine regulation, it has been referred to as the "autonomic master controller." The emerging consensus is that the PVN is a multifunctional nucleus, with autonomic roles including (but not limited to) coordination of cardiovascular, thermoregulatory, metabolic, circadian and stress responses. However, the cellular mechanisms underlying these multifunctional roles remain poorly understood. Neurones from the PVN project to and can alter the function of sympathetic control regions in the medulla and spinal cord. Dysfunction of sympathetic pre-autonomic neurones (typically hyperactivity) is linked to several diseases including hypertension and heart failure and targeting this region with specific pharmacological or biological agents is a promising area of medical research. However, to facilitate future medical exploitation of the PVN, more detailed models of its neuronal control are required; populated by a greater compliment of constituent ion channels. Whilst the cytoarchitecture, projections and neurotransmitters present in the PVN are reasonably well documented, there have been fewer studies on the expression and interplay of ion channels. In this review we bring together an up to date analysis of PVN ion channel studies and discuss how these channels may interact to control, in particular, the activity of the sympathetic system.
Collapse
Affiliation(s)
- Claire H Feetham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Fiona O'Brien
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Richard Barrett-Jolley
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
34
|
Harrison K, Sisley S. Vitamin D and the paraventricular nucleus: Relevance for type 2 diabetes. J Steroid Biochem Mol Biol 2018; 177:125-128. [PMID: 28993246 DOI: 10.1016/j.jsbmb.2017.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
Abstract
Vitamin D deficiency is linked to type 2 diabetes and we recently showed this may be through action of vitamin D in the paraventricular nuclei (PVN) in the hypothalamus of the brain. This review focuses on the known roles of the PVN in glucose control and how previously discovered actions of vitamin D in other tissues may translate to action in the PVN. Specifically, we focus on the role of insulin and inflammation in the hypothalamus and how these may be modified through vitamin D action.
Collapse
Affiliation(s)
- Keisha Harrison
- Department of Pediatrics, Baylor College of Medicine, 1100 Bates Avenue, Houston, TX 77030, USA.
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, 1100 Bates Avenue, Houston, TX 77030, USA.
| |
Collapse
|
35
|
Udoh US, Valcin JA, Swain TM, Filiano AN, Gamble KL, Young ME, Bailey SM. Genetic deletion of the circadian clock transcription factor BMAL1 and chronic alcohol consumption differentially alter hepatic glycogen in mice. Am J Physiol Gastrointest Liver Physiol 2018; 314:G431-G447. [PMID: 29191941 PMCID: PMC5899240 DOI: 10.1152/ajpgi.00281.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/27/2017] [Accepted: 11/27/2017] [Indexed: 01/31/2023]
Abstract
Multiple metabolic pathways exhibit time-of-day-dependent rhythms that are controlled by the molecular circadian clock. We have shown that chronic alcohol is capable of altering the molecular clock and diurnal oscillations in several elements of hepatic glycogen metabolism ( 19 , 44 ). Herein, we sought to determine whether genetic disruption of the hepatocyte clock differentially impacts hepatic glycogen content in chronic alcohol-fed mice. Male hepatocyte-specific BMAL1 knockout (HBK) and littermate controls were fed control or alcohol-containing diets for 5 wk to alter hepatic glycogen content. Glycogen displayed a significant diurnal rhythm in livers of control genotype mice fed the control diet. While rhythmic, alcohol significantly altered the diurnal oscillation of glycogen in livers of control genotype mice. The glycogen rhythm was mildly altered in livers of control-fed HBK mice. Importantly, glycogen content was arrhythmic in livers of alcohol-fed HBK mice. Consistent with these changes in hepatic glycogen content, we observed that some glycogen and glucose metabolism genes were differentially altered by chronic alcohol consumption in livers of HBK and littermate control mice. Diurnal rhythms in glycogen synthase (mRNA and protein) were significantly altered by alcohol feeding and clock disruption. Alcohol consumption significantly altered Gck, Glut2, and Ppp1r3g rhythms in livers of control genotype mice, with diurnal rhythms of Pklr, Glut2, Ppp1r3c, and Ppp1r3g further disrupted (dampened or arrhythmic) in livers of HBK mice. Taken together, these findings show that chronic alcohol consumption and hepatocyte clock disruption differentially influence the diurnal rhythm of glycogen and various key glycogen metabolism-related genes in the liver. NEW & NOTEWORTHY We report that circadian clock disruption exacerbates alcohol-mediated alterations in hepatic glycogen. We observed differential responsiveness in diurnal rhythms of glycogen and glycogen metabolism genes and proteins in livers of hepatocyte-specific BMAL1 knockout and littermate control mice fed alcohol. Our findings provide new insights into potential mechanisms by which alcohol alters glycogen, an important energy source for liver and other organs.
Collapse
Affiliation(s)
- Uduak S Udoh
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jennifer A Valcin
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Telisha M Swain
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Ashley N Filiano
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Karen L Gamble
- Department of Psychiatry, Division of Behavioral Neurobiology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham , Birmingham, Alabama
| | - Shannon M Bailey
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
36
|
Luo S, Zhang Y, Ezrokhi M, Li Y, Tsai T, Cincotta AH. Circadian peak dopaminergic activity response at the biological clock pacemaker (suprachiasmatic nucleus) area mediates the metabolic responsiveness to a high-fat diet. J Neuroendocrinol 2018; 30:e12563. [PMID: 29224246 PMCID: PMC5817247 DOI: 10.1111/jne.12563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/24/2022]
Abstract
Among vertebrate species of the major vertebrate classes in the wild, a seasonal rhythm of whole body fuel metabolism, oscillating from a lean to obese condition, is a common biological phenomenon. This annual cycle is driven in part by annual changes in the circadian dopaminergic signalling at the suprachiasmatic nuclei (SCN), with diminution of circadian peak dopaminergic activity at the SCN facilitating development of the seasonal obese insulin-resistant condition. The present study investigated whether such an ancient circadian dopamine-SCN activity system for expression of the seasonal obese, insulin-resistant phenotype may be operative in animals made obese amd insulin resistant by high-fat feeding and, if so, whether reinstatement of the circadian dopaminergic peak at the SCN would be sufficient to reverse the adverse metabolic impact of the high-fat diet without any alteration of caloric intake. First, we identified the supramammillary nucleus as a novel site providing the majority of dopaminergic neuronal input to the SCN. We further identified dopamine D2 receptors within the peri-SCN region as being functional in mediating SCN responsiveness to local dopamine. In lean, insulin-sensitive rats, the peak in the circadian rhythm of dopamine release at the peri-SCN coincided with the daily peak in SCN electrophysiological responsiveness to local dopamine administration. However, in rats made obese and insulin resistant by high-fat diet (HFD) feeding, these coincident circadian peak activities were both markedly attenuated or abolished. Reinstatement of the circadian peak in dopamine level at the peri-SCN by its appropriate circadian-timed daily microinjection to this area (but not outside this circadian time-interval) abrogated the obese, insulin-resistant condition without altering the consumption of the HFD. These findings suggest that the circadian peak of dopaminergic activity at the peri-SCN/SCN is a key modulator of metabolism and the responsiveness to adverse metabolic consequences of HFD consumption.
Collapse
Affiliation(s)
- S. Luo
- VeroScience LLCTivertonRIUSA
| | | | | | - Y. Li
- VeroScience LLCTivertonRIUSA
| | | | | |
Collapse
|
37
|
Silva AG, Frizzo ACF, Chagas EFB, Garner DM, Raimundo RD, de Alcantara Sousa LV, Valenti VE. A relationship between brainstem auditory evoked potential and vagal control of heart rate in adult women. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Zhang Z, Machado F, Zhao L, Heinen CA, Foppen E, Ackermans MT, Zhou J, Bisschop PH, Boelen A, Fliers E, Kalsbeek A. Administration of Thyrotropin-Releasing Hormone in the Hypothalamic Paraventricular Nucleus of Male Rats Mimics the Metabolic Cold Defense Response. Neuroendocrinology 2018; 107:267-279. [PMID: 30092582 PMCID: PMC6390456 DOI: 10.1159/000492785] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cold exposure increases thyrotropin-releasing hormone (TRH) expression primarily in the hypothalamic paraventricular nucleus (PVN). The PVN is a well-known hypothalamic hub in the control of energy metabolism. TRH terminals and receptors are found on PVN neurons. We hypothesized that TRH release in the PVN plays an important role in the control of thermogenesis and energy mobilization during cold exposure. METHODS Male Wistar rats were exposed to a cold environment (4°C) or TRH retrodialysis in the PVN for 2 h. We compared the effects of cold exposure and TRH administration in the PVN on plasma glucose, corticosterone, and thyroid hormone concentrations, body temperature, locomotor activity, as well as metabolic gene expression in the liver and brown adipose tissue. RESULTS Cold exposure increased body temperature, locomotor activity, and plasma corticosterone concentrations, but blood glucose concentrations were similar to that of room temperature control animals. TRH administration in the PVN also promptly increased body temperature, locomotor activity and plasma corticosterone concentrations. However, TRH administration in the PVN markedly increased blood glucose concentrations and endogenous glucose production (EGP) compared to saline controls. Selective hepatic sympathetic or parasympathetic denervation reduced the TRH-induced increase in glucose concentrations and EGP. Gene expression data indicated increased gluconeogenesis in liver and lipolysis in brown adipose tissue, both after cold exposure and TRH administration. CONCLUSIONS We conclude that TRH administration in the rat PVN largely mimics the metabolic and behavioral changes induced by cold exposure indicating a potential link between TRH release in the PVN and cold defense.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Frederico Machado
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Li Zhao
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the Netherlands
| | - Charlotte A Heinen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Ewout Foppen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jiangning Zhou
- CAS Key Laboratory of Brain Function and Disease, School of Life Science, University of Science and Technology of China, Hefei, China
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anita Boelen
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, the
| |
Collapse
|
39
|
Overactivity of Liver-Related Neurons in the Paraventricular Nucleus of the Hypothalamus: Electrophysiological Findings in db/db Mice. J Neurosci 2017; 37:11140-11150. [PMID: 29038244 DOI: 10.1523/jneurosci.1706-17.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/11/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022] Open
Abstract
Preautonomic neurons in the paraventricular nucleus (PVN) of the hypothalamus play a large role in the regulation of hepatic functions via the autonomic nervous system. Activation of hepatic sympathetic nerves increases glucose and lipid metabolism and contributes to the elevated hepatic glucose production observed in the type 2 diabetic condition. This augmented sympathetic output could originate from altered activity of liver-related PVN neurons. Remarkably, despite the importance of the brain-liver pathway, the cellular properties of liver-related neurons are not known. In this study, we provide the first evidence of overall activity of liver-related PVN neurons. Liver-related PVN neurons were identified with a retrograde, trans-synaptic, viral tracer in male lean and db/db mice and whole-cell patch-clamp recordings were conducted. In db/db mice, the majority of liver-related PVN neurons fired spontaneously; whereas, in lean mice the majority of liver-related PVN neurons were silent, indicating that liver-related PVN neurons are more active in db/db mice. Persistent, tonic inhibition was identified in liver-related PVN neurons; although, the magnitude of tonic inhibitory control was not different between lean and db/db mice. In addition, our study revealed that the transient receptor potential vanilloid type 1-dependent increase of excitatory neurotransmission was reduced in liver-related PVN neurons of db/db mice. These findings demonstrate plasticity of liver-related PVN neurons and a shift toward excitation in a diabetic mouse model. Our study suggests altered autonomic circuits at the level of the PVN, which can contribute to autonomic dysfunction and dysregulation of neural control of hepatic functions including glucose metabolism.SIGNIFICANCE STATEMENT A growing body of evidence suggests the importance of the autonomic control in the regulation of hepatic metabolism, which plays a major role in the development and progression of type 2 diabetes mellitus. Despite the importance of the brain-liver pathway, the overall activity of liver-related neurons in control and diabetic conditions is not known. This is a significant gap in knowledge, which prevents developing strategies to improve glucose homeostasis via altering the brain-liver pathway. One of the key findings of our study is the overall shift toward excitation in liver-related hypothalamic neurons in the diabetic condition. This overactivity may be one of the underlying mechanisms of elevated sympathetic activity known in metabolically compromised patients and animal models.
Collapse
|
40
|
Mukerjee S, Zhu Y, Zsombok A, Mauvais-Jarvis F, Zhao J, Lazartigues E. Perinatal Exposure to Western Diet Programs Autonomic Dysfunction in the Male Offspring. Cell Mol Neurobiol 2017; 38:233-242. [PMID: 28478572 DOI: 10.1007/s10571-017-0502-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/03/2017] [Indexed: 12/15/2022]
Abstract
Although the deleterious influence of protein deficiency on fetal programming is well documented, the impact of a Western diet on epigenetic mechanisms is less clear. We hypothesized that high-fat high-sucrose diet (HFHSD) consumption during pregnancy leads to epigenetic modifications within the progeny's compensatory renin-angiotensin system (RAS), affecting autonomic and metabolic functions. Dams were fed HFHSD (45% fat and 30% sucrose) or regular chow (RD) from mating until weaning of the pups (~7 weeks). Offspring from both groups were then maintained on chow and studied in adulthood (3-7 months). Offspring from HFHSD-exposed dams (OH) exhibited no difference in body weight or fasting blood glucose compared to controls (OR). In 3-month-old offspring, DNA methylation was significantly lower for the ACE2 gene (P < 0.05) in the brainstem, kidney and cecum. Moreover, ACE2 activity in the hypothalamus was increased at 7 months (OH: 91 ± 1 vs. OR: 74 ± 4 AFU/mg/min, P < 0.05). Although baseline blood pressure was not different between groups, vagal tone in OH was significantly impaired compared to OR. At the same time, OH offspring had a 1.7-fold increase in AT1a receptor expression and a 1.3-fold increase in ADAM17 mRNA. DOCA-salt treatment further revealed and exacerbated hypertensive response in the OH progeny (OH: 130 ± 6 vs. OR: 108 ± 3 mmHg, P < 0.05). Taken together, our data suggest that perinatal exposure to HFHSD resulted in epigenetic modifications of the compensatory brain RAS, potentially affecting plasticity of neuronal networks leading to autonomic dysfunction in the male offspring.
Collapse
Affiliation(s)
- Snigdha Mukerjee
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Yun Zhu
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Andrea Zsombok
- Department of Physiology, Tulane University, New Orleans, LA, USA
| | | | - Jinying Zhao
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA. .,Department of Pharmacology and Experimental Therapeutics, School of Medicine, Louisiana State University Health Sciences Center, 1901 Perdido Street, Room 5218, New Orleans, LA, 70112, USA.
| |
Collapse
|
41
|
Bruce KD, Zsombok A, Eckel RH. Lipid Processing in the Brain: A Key Regulator of Systemic Metabolism. Front Endocrinol (Lausanne) 2017; 8:60. [PMID: 28421037 PMCID: PMC5378716 DOI: 10.3389/fendo.2017.00060] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
Metabolic disorders, particularly aberrations in lipid homeostasis, such as obesity, type 2 diabetes mellitus, and hypertriglyceridemia often manifest together as the metabolic syndrome (MetS). Despite major advances in our understanding of the pathogenesis of these disorders, the prevalence of the MetS continues to rise. It is becoming increasingly apparent that intermediary metabolism within the central nervous system is a major contributor to the regulation of systemic metabolism. In particular, lipid metabolism within the brain is tightly regulated to maintain neuronal structure and function and may signal nutrient status to modulate metabolism in key peripheral tissues such as the liver. There is now a growing body of evidence to suggest that fatty acid (FA) sensing in hypothalamic neurons via accumulation of FAs or FA metabolites may signal nutritional sufficiency and may decrease hepatic glucose production, lipogenesis, and VLDL-TG secretion. In addition, recent studies have highlighted the existence of liver-related neurons that have the potential to direct such signals through parasympathetic and sympathetic nervous system activity. However, to date whether these liver-related neurons are FA sensitive remain to be determined. The findings discussed in this review underscore the importance of the autonomic nervous system in the regulation of systemic metabolism and highlight the need for further research to determine the key features of FA neurons, which may serve as novel therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Kimberley D. Bruce
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
- *Correspondence: Kimberley D. Bruce,
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Robert H. Eckel
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
| |
Collapse
|
42
|
Han C, Rice MW, Cai D. Neuroinflammatory and autonomic mechanisms in diabetes and hypertension. Am J Physiol Endocrinol Metab 2016; 311:E32-41. [PMID: 27166279 PMCID: PMC4967151 DOI: 10.1152/ajpendo.00012.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
Interdisciplinary studies in the research fields of endocrinology and immunology show that obesity-associated overnutrition leads to neuroinflammatory molecular changes, in particular in the hypothalamus, chronically causing various disorders known as elements of metabolic syndrome. In this process, neural or hypothalamic inflammation impairs the neuroendocrine and autonomic regulation of the brain over blood pressure and glucose homeostasis as well as insulin secretion, and elevated sympathetic activation has been appreciated as a critical mediator. This review describes the involved physiology and mechanisms, with a focus on glucose and blood pressure balance, and suggests that neuroinflammation employs the autonomic nervous system to mediate the development of diabetes and hypertension.
Collapse
Affiliation(s)
- Cheng Han
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York
| | - Matthew W Rice
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, Institute of Aging, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|