1
|
Chen D, Yang YY, Yang Y. Astrocyte Loss Augments Body Weight Through Reduction in Adipose Sympathetic Outflows. Glia 2025; 73:1068-1076. [PMID: 39780483 PMCID: PMC11922664 DOI: 10.1002/glia.24673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Emerging evidence indicates that astrocytes modulate energy metabolism and homeostasis. However, one important but poorly understood element is the necessity of astrocytes in the control of body weight. Here, we apply viral vector-assisted brain-region selective loss of astrocytes to define physiological roles played by astrocytes in the arcuate nucleus of the hypothalamus (ARH) and to elucidate the involved mechanism. We find that astrocyte loss potently augments body weight in adult mice fed chow or high-fat diet. Mechanistically, we find that the loss of astrocytes reduces adipose tissue norepinephrine (NE) contents and chemogenetic stimulation of adipose tissue sympathetic inputs reverses the astrocyte loss-induced increase in body weight. Collectively, our findings in this study suggest a crucial physiological role of astrocytes in preventing diet-induced energy surfeit and obesity by modulating adipose tissue lipid metabolism through central sympathetic outflows to adipose tissues.
Collapse
Affiliation(s)
- Dan Chen
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | - Yunlei Yang
- Department of Medicine Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
2
|
Lin Z, Xuan Y, Zhang Y, Zhou Q, Qiu W. Hypothalamus and brainstem circuits in the regulation of glucose homeostasis. Am J Physiol Endocrinol Metab 2025; 328:E588-E598. [PMID: 40047236 DOI: 10.1152/ajpendo.00474.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/03/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
The central nervous system (CNS) senses and integrates blood glucose status, regulating its levels through communication with peripheral organs. Since traditional wisdom holds that the hypothalamus primarily controls glucose homeostasis, the brainstem, although less studied, has been emerging as a key player in blood glucose metabolism. Although the brainstem is reciprocally wired with the hypothalamus, their interactions are crucial for glucose control. Here, we focus on classic discoveries and recent advancements of hypothalamic and brainstem nodes that regulate glucose homeostasis. Based on the current progress and development for central regulation of blood sugar, we propose that the circuitry and cellular mechanisms for how hypothalamus and brainstem coordinate in blood sugar regulation are crucial; hence, a deeper understanding of both nuclei could shed light on a future cure for diabetes.
Collapse
Affiliation(s)
- Zitian Lin
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yunxin Xuan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yingshi Zhang
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Qirui Zhou
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Weiwei Qiu
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| |
Collapse
|
3
|
Zhang T, Li XY, Kuang DD, Pan LH, Li QM, Luo JP, Zha XQ. Bone-brain communication mediates the amelioration of Polgonatum cyrtonema Hua polysaccharide on fatigue in chronic sleep-deprived mice. Int J Biol Macromol 2025; 296:139706. [PMID: 39793823 DOI: 10.1016/j.ijbiomac.2025.139706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
This study aimed to investigate the anti-fatigue efficacy and underlying mechanisms of Polygonatum cyrtonema Hua polysaccharide (PCP) in chronic sleep-deprived mice. Following three weeks of oral administration, PCP demonstrated significant efficacy in alleviating fatigue symptoms. This was evidenced by the prolonged swimming and rotarod time in the high-dose group of PCP, which increased by 73 % and 64 %, respectively. Additionally, serum activities of CAT, GSH-Px, and SOD enzymes rose by 53.56 %, 37.69 % and 53.67 %, respectively, while MDA, lactic acid and BUN levels decreased by 22.90 %, 17.48 % and 24.61 %. The crosstalk between bone and brain is crucial for maintaining energy homeostasis. Molecular docking studies indicated a spontaneous and strong mutual binding between PCP and the bone-promoting target protein BMPR1A. Furthermore, it was observed that PCP enhanced osteogenic differentiation via the BMP-2/Smad1 pathway, leading to an upregulation of osteocalcin expression, which in turn regulated neurotransmitter balance and improved central arousal capacity. Moreover, PCP treatment stimulated neurogenesis by activating the CREB/BDNF/Akt signaling cascade, exhibiting neurotrophic effects. Additionally, PCP increased AMPK phosphorylation and destabilized TXNIP, facilitating astrocyte glucose uptake, glycolysis, and lactate conversion to support neuronal activity. These findings suggested that PCP could effectively respond to energy demands through bone-brain crosstalk, ultimately exerting anti-fatigue properties.
Collapse
Affiliation(s)
- Ting Zhang
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Dan-Dan Kuang
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| |
Collapse
|
4
|
Bogard AT, Hembree TG, Pollet AK, Smith AC, Ryder SC, Marzloff GE, Tan AQ. Intermittent hypoxia-induced enhancements in corticospinal excitability predict gains in motor learning and metabolic efficiency. Sci Rep 2025; 15:6614. [PMID: 39994358 PMCID: PMC11850928 DOI: 10.1038/s41598-025-90890-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Acute intermittent hypoxia (AIH) enhances human motor function after incomplete spinal cord injury. Although the underlying mechanisms in humans are unknown, emerging evidence indicates that AIH facilitates corticospinal excitability to the upper limb. However, the functional relevance of this plasticity remains unexplored, and it is unclear whether similar plasticity can be induced for lower limb motor areas. We recently demonstrated that AIH improves motor adaptation, motor savings, and metabolic efficiency during split-belt walking. Thus, we hypothesized that AIH increases lower limb excitability and that these enhancements would predict the magnitude of motor learning and the corresponding reductions in net metabolic power. We assessed tibialis anterior (TA) excitability using transcranial magnetic stimulation and quantified changes in spatiotemporal asymmetries and net metabolic power in response to split-belt speed perturbations. We show that AIH enhances TA excitability, and that the magnitude of this facilitation positively correlates with greater spatiotemporal adaptation. Notably, we demonstrate a novel association between increased excitability and reduced net metabolic power during motor adaptation and motor savings. Together, our results suggest that AIH-induced gains in excitability predict both the magnitude of motor learning and the associated metabolic efficiency. Determining indices of AIH-induced improvements in motor performance is critical for optimizing its therapeutic reach.
Collapse
Affiliation(s)
- Alysha T Bogard
- Sensorimotor Recovery and Neuroplasticity Lab, Department of Integrative Physiology, University of Colorado, Boulder, 80309, USA
| | - Thomas G Hembree
- Sensorimotor Recovery and Neuroplasticity Lab, Department of Integrative Physiology, University of Colorado, Boulder, 80309, USA
| | - Aviva K Pollet
- Sensorimotor Recovery and Neuroplasticity Lab, Department of Integrative Physiology, University of Colorado, Boulder, 80309, USA
| | - Andrew C Smith
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, 80045, USA
| | - Stephanie C Ryder
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, 80045, USA
| | - George E Marzloff
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, 80045, USA
| | - Andrew Q Tan
- Sensorimotor Recovery and Neuroplasticity Lab, Department of Integrative Physiology, University of Colorado, Boulder, 80309, USA.
- Rocky Mountain Regional VA Medical Center, Aurora, 80045, USA.
- Center for Neuroscience, University of Colorado, Boulder, 80309, USA.
| |
Collapse
|
5
|
Fan YH, Zhang S, Wang Y, Wang H, Li H, Bai L. Inter-organ metabolic interaction networks in non-alcoholic fatty liver disease. Front Endocrinol (Lausanne) 2025; 15:1494560. [PMID: 39850476 PMCID: PMC11754069 DOI: 10.3389/fendo.2024.1494560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem metabolic disorder, marked by abnormal lipid accumulation and intricate inter-organ interactions, which contribute to systemic metabolic imbalances. NAFLD may progress through several stages, including simple steatosis (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and potentially liver cancer. This disease is closely associated with metabolic disorders driven by overnutrition, with key pathological processes including lipid dysregulation, impaired lipid autophagy, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and local inflammation. While hepatic lipid metabolism in NAFLD is well-documented, further research into inter-organ communication mechanisms is crucial for a deeper understanding of NAFLD progression. This review delves into intrahepatic networks and tissue-specific signaling mediators involved in NAFLD pathogenesis, emphasizing their impact on distal organs.
Collapse
Affiliation(s)
- Yu-Hong Fan
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Siyao Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ye Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongni Wang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China
- Department of Cardiology, Renmin Hospital, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Bai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
6
|
Cheng B, Du J, Tian S, Zhang Z, Chen W, Liu Y. High-intensity interval training or lactate administration combined with aerobic training enhances visceral fat loss while promoting VMH neuroplasticity in female rats. Lipids Health Dis 2024; 23:405. [PMID: 39696579 DOI: 10.1186/s12944-024-02397-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND High-intensity interval training (HIT) does not burn fat during exercise. However, it significantly reduces visceral adipose after long-term training. The underlying mechanism may be related to the elevation of fat consumption during the post-exercise recovery period, which is regulated by the hypothalamus-adipose axis. Lactate is a hallmark metabolite of high-intensity exercise, which could mediate significant neuroplasticity through the brain-derived neurotrophic factor (BDNF) pathway. However, whether HIT could enhance hypothalamus activity and adipose catabolism in the recovery period remains to be elucidated. Also, it is worth exploring whether adding lactate administration to prolonged, continuous submaximal aerobic training (AT) could simulate HIT-induced neuroplastic effects and fat loss. METHODS First, we compared the influence of 4-week HIT and aerobic training (AT) on the electrophysiology of the ventromedial hypothalamus (VMH), which is deeply involved in the regulation of lipolysis, as well as the 24-hour excess post-exercise oxygen consumption (EPOC), the fat oxidation rate and lipolysis. To further confirm whether excess lactate during AT could reproduce the effect of HIT, we also observed the effects of lactate infusion during AT (AT + Lac) on neuroplasticity and metabolism. RESULTS Four-week HIT induced higher BDNF expression and a higher neuronal spike firing rate in VMH than AT, accompanied by elevated EPOC, fat oxidation and visceral fat lipolysis. AT + Lac and HITT could induce similar hypothalamic and metabolic changes. However, power spectral density analysis of local field potentials (LFPs) showed that the AT + Lac group was affected in fewer frequency bands than the HIT group. CONCLUSION HIT-induced reduction of visceral fat was accompanied by increased VMH activity. Adding lactate administration to AT could partially reproduce hypothalamic plasticity and the metabolic effects of HIT. However, different band changes of LFPs implied that the neuronal subpopulations or pathways influenced by these two methods were not entirely consistent.
Collapse
Affiliation(s)
- Baishuo Cheng
- Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Jinchan Du
- Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Shuai Tian
- Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Zixiong Zhang
- Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Wei Chen
- Physical Education College, Hebei Normal University, Shijiazhuang, China
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio-Information, Hebei Normal University, No. 20, South Second Ring Road East, Shijiazhuang, Hebei, China
| | - Yang Liu
- Physical Education College, Hebei Normal University, Shijiazhuang, China.
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio-Information, Hebei Normal University, No. 20, South Second Ring Road East, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Zhang W, Yan Y, Yi C, Jiang X, Guo L, Huang S, Xia T, Huang F, Jiao Y, Li H, Yu B, Dai Y. Targeting ferroptosis in the neurovascular unit: A promising approach for treating diabetic cognitive impairment. Int Immunopharmacol 2024; 142:113146. [PMID: 39298819 DOI: 10.1016/j.intimp.2024.113146] [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: 05/02/2024] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
The cognitive decline associated with chronic metabolic disease diabetes has garnered extensive scrutiny, yet its pathogenesis remains incompletely understood, and the advancement of targeted therapeutics has posed a persistent challenge. Ferroptosis, a novel form of cell death characterized by intracellular lipid peroxidation and iron overload, has recently emerged as a significant factor. Numerous contemporary studies have corroborated that ferroptosis within the neurovascular unit is intimately associated with the onset of diabetes-induced cognitive impairment. Numerous contemporary studies have corroborated that ferroptosis within the neurovascular unit is intimately associated with the onset of diabetic cognitive impairment (DCI). This article initially conducts a profound analysis of the mechanism of ferroptosis, followed by a detailed elucidation of the specific manifestations of neurovascular unit ferroptosis in the context of diabetic cognitive function impairment. Furthermore, an exhaustive review of pertinent literature from April 2020 to March 2024 has been undertaken, resulting in the selection of 31 documents of significant reference value. These documents encompass studies on 11 distinct drugs, all of which are centered around investigating methods to inhibit the ferroptosis pathway as a potential treatment for DCI. Simultaneously, we conducted a review of 12 supplementary literary sources that presented 10 pharmacological agents with anti-ferroptosis properties in other neurodegenerative disorders. This article critically examines the potential influence of neurovascular unit ferroptosis on the progression of cognitive impairment in diabetes, from the three aforementioned perspectives, and organizes the existing and potential therapeutic drugs. It is our aspiration that this article will serve as a theoretical foundation for scholars in related disciplines when conceptualizing, investigating, and developing novel clinical drugs for DCI.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yijing Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chunmei Yi
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shanshan Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tong Xia
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fayin Huang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yike Jiao
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
8
|
Kosse C, Ivanov J, Knight Z, Pellegrino K, Friedman J. A subcortical feeding circuit linking an interoceptive node to jaw movement. Nature 2024; 636:151-161. [PMID: 39443799 PMCID: PMC11618074 DOI: 10.1038/s41586-024-08098-1] [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: 10/24/2022] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The brain processes an array of stimuli, enabling the selection of appropriate behavioural responses, but the neural pathways linking interoceptive inputs to outputs for feeding are poorly understood1-3. Here we delineate a subcortical circuit in which brain-derived neurotrophic factor (BDNF)-expressing neurons in the ventromedial hypothalamus (VMH) directly connect interoceptive inputs to motor centres, controlling food consumption and jaw movements. VMHBDNF neuron inhibition increases food intake by gating motor sequences of feeding through projections to premotor areas of the jaw. When food is unavailable, VMHBDNF inhibition elicits consummatory behaviours directed at inanimate objects such as wooden blocks, and inhibition of perimesencephalic trigeminal area (pMe5) projections evokes rhythmic jaw movements. The activity of these neurons is decreased during food consumption and increases when food is in proximity but not consumed. Activity is also increased in obese animals and after leptin treatment. VMHBDNF neurons receive monosynaptic inputs from both agouti-related peptide (AgRP) and proopiomelanocortin neurons in the arcuate nucleus (Arc), and constitutive VMHBDNF activation blocks the orexigenic effect of AgRP activation. These data indicate an Arc → VMHBDNF → pMe5 circuit that senses the energy state of an animal and regulates consummatory behaviours in a state-dependent manner.
Collapse
Affiliation(s)
- Christin Kosse
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jessica Ivanov
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Zachary Knight
- Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Pellegrino
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Jeffrey Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
9
|
Ma W, Sui D, Sun W, Yu P, Li Y, Guo M, Wang H, Zhang X, Yu X, Fu W, Xu H. 5,7,3',4',5'-Pentamethoxyflavone, a Flavonoid Monomer Extracted From Murraya paniculata (L.) Jack, Alleviates Anxiety Through the A 2AR/Gephyrin/GABRA2 Pathway. Phytother Res 2024; 38:5539-5548. [PMID: 39261011 DOI: 10.1002/ptr.8327] [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: 12/26/2023] [Revised: 07/28/2024] [Accepted: 08/17/2024] [Indexed: 09/13/2024]
Abstract
The sedative and hypnotic properties of 5,7,3',4',5'-pentamethoxyflavone (PMF), a monomer extracted from the leaves of Murraya paniculata (L.) Jack, have been reported. However, the role of PMFs in the development of anxiety remains uncertain. An anxiety model was developed using chronic unpredictable mild stimulation (CUMS). Kunming mice were randomly allocated to the following groups: control, CUMS, PMF (50 mg/kg), PMF (100 mg/kg), and diazepam (3 mg/kg). The anxiolytic effects of PMFs were evaluated using elevated plus maze (EPM) test and open field test (OFT). Enzyme-linked immunosorbent assay (ELISA) kits were used to analyze the serum levels of corticosterone (CORT), 5-hydroxytryptamine (5-HT), gamma-aminobutyric acid (GABA), and cyclic adenosine monophosphate (cAMP) in the hippocampus. High-throughput-16S rRNA sequencing was performed to investigate its effect on the composition of the gut microbiota. Subsequently, western blotting was performed to assess the expression of GABAergic synaptic-associated proteins. PMF effectively mitigated CUMS-induced anxiety-like behavior. Further examination revealed that PMF treatment ameliorated dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis and increased 5-HT and GABA levels in the hippocampus. Notably, the ability of PMF to maintain the stability of GABAergic synapses by enhancing the species composition of the gut microbiota and acting on the adenosine a2a receptor (A2AR)/gephyrin/gamma-aminobutyric acid A receptor alpha 2 (GABRA2) pathway revealed a previously unrecognized mechanism for the anxiolytic effect of PMF. These findings suggest that PMF enhances the expression of A2AR, preserves GABAergic synaptic stability, and reduces anxiety by modulating the microbiota composition. Thus, it holds promise as an anxiolytic agent.
Collapse
Affiliation(s)
- Wenli Ma
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Weilun Sun
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Ping Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Yuangeng Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Meiqi Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Huifeng Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiaoze Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Roszkowicz-Ostrowska K, Młotkowska P, Marciniak E, Szlis M, Barszcz M, Misztal T. Activation of BDNF-TrkB Signaling in Specific Structures of the Sheep Brain by Kynurenic Acid. Cells 2024; 13:1928. [PMID: 39682677 PMCID: PMC11639857 DOI: 10.3390/cells13231928] [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: 10/13/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Fluctuations in kynurenic acid (KYNA) and brain-derived neurotrophic factor (BDNF) levels in the brain reflect its neurological status. The aim of the study was to investigate the effect of transiently elevated KYNA concentrations in the cerebroventricular circulation on the expression of BDNF and its high-affinity tropomyosin-related kinase receptor B (TrkB) in specific structures of the sheep brain. Intracerebroventricularly cannulated anestrous sheep were subjected to a series of four 30 min infusions of KYNA: 4 × 5 μg/60 μL/30 min (KYNA20, n = 6) and 4 × 25 μg/60 μL/30 min (KYNA100, n = 6) or a control infusion (n = 6), at 30 min intervals. Sections of the hippocampal CA3 field, amygdala (AMG), prefrontal cortex (PCx), and the hypothalamic medial-basal (MBH) and preoptic (POA) areas were dissected from the brain immediately after the experiment. The highest concentration of BDNF protein was found in the CA3 field (p < 0.001), which was 8-fold higher than in the AMG and 12-fold higher than that in the PCx (MBH and POA were not analyzed). The most pronounced BDNF mRNA expression was observed in the MBH, followed by the PCx, POA, AMG and CA3, while the highest abundance of TrkB mRNA was recorded in the AMG, followed by the MBH, PCx, CA3, and POA. KYNA increased (p < 0.05-p < 0.01) BDNF protein levels and the expression of its gene in the brain structures were examined, with the effect varying by dose and brain region. KYNA, particularly at the KYNA100 dose, also increased (p < 0.01) TrkB gene expression, except for the AMG, where the lower KYNA20 dose was more effective (p < 0.01). These findings suggest a positive relationship between KYNA levels in the cerebroventricular circulation and BDNF-TrkB expression in specific brain regions in a sheep model. This indicates that a transient increase in the CSF KYNA concentration can potentially restore BDNF production, for which deficiency underlies numerous neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Tomasz Misztal
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Str., 05-110 Jabłonna, Poland; (K.R.-O.); (P.M.); (E.M.); (M.S.); (M.B.)
| |
Collapse
|
11
|
Paidlewar M, Kumari S, Dhapola R, Sharma P, HariKrishnaReddy D. Unveiling the role of astrogliosis in Alzheimer's disease Pathology: Insights into mechanisms and therapeutic approaches. Int Immunopharmacol 2024; 141:112940. [PMID: 39154532 DOI: 10.1016/j.intimp.2024.112940] [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: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is one of the most debilitating age-related disorders that affect people globally. It impacts social and cognitive behavior of the individual and is characterized by phosphorylated tau and Aβ accumulation. Astrocytesmaintain a quiescent, anti-inflammatory state on anatomical level, expressing few cytokines and exhibit phagocytic activity to remove misfolded proteins. But in AD, in response to specific stimuli, astrocytes overstimulate their phagocytic character with overexpressing cytokine gene modules. Upon interaction with generated Aβ and neurofibrillary tangle, astrocytes that are continuously activated release a large number of inflammatory cytokines. This cytokine storm leads to neuroinflammation which is also one of the recognizable features of AD. Astrogliosis eventually promotes cholinergic dysfunction, calcium imbalance, oxidative stress and excitotoxicity. Furthermore, C5aR1, Lcn2/, BDNF/TrkB and PPARα/TFEB signaling dysregulation has a major impact on the disease progression. This review clarifies numerous ways that lead to astrogliosis, which is stimulated by a variety of processes that exacerbate AD pathology and make it a suitable target for AD treatment. Drugs under clinical and preclinical investigations that target several pathways managing astrogliosis and are efficacious in ameliorating the pathology of the disease are also included in this study. D-ALA2GIP, TRAM-34, Genistein, L-serine, MW150 and XPro1595 are examples of few drugs targeting astrogliosis. Therefore, this study may aid in the development of a potent therapeutic agent for ameliorating astrogliosis mediated AD progression.
Collapse
Affiliation(s)
- Mohit Paidlewar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda-151401, Punjab, India.
| |
Collapse
|
12
|
Valladolid-Acebes I. Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines 2024; 12:2422. [PMID: 39594988 PMCID: PMC11591892 DOI: 10.3390/biomedicines12112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Leptin, an adipokine essential for regulating energy balance, exerts important effects on brain function, notably within the hippocampus, a region integral to learning and memory. Leptin resistance, characterized by diminished responsiveness to elevated leptin levels, disrupts hippocampal function and exacerbates both obesity and cognitive impairments. Scope: This review critically examines how leptin resistance impairs hippocampal synaptic plasticity processes, specifically affecting long-term potentiation (LTP) and long-term depression (LTD), which are crucial for cognitive performance. Findings: Recent research highlights that leptin resistance disrupts N-methyl-D-aspartate (NMDA) receptor dynamics and hippocampal structure, leading to deficits in spatial learning and memory. Additionally, high-fat diets (HFDs), which contribute to leptin resistance, further deteriorate hippocampal function. Potential therapeutic strategies, including leptin sensitizers, show promise in mitigating brain disorders associated with leptin resistance. Complementary interventions such as caloric restriction and physical exercise also enhance leptin sensitivity and offer potential benefits to alleviating cognitive impairments. Aims of the review: This review synthesizes recent findings on the molecular pathways underlying leptin resistance and its impact on synaptic transmission and plasticity in the hippocampus. By identifying potential therapeutic targets, this work aims to provide an integrated approach for addressing cognitive deficits in obesity, ultimately improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
| |
Collapse
|
13
|
Pinkston BTC, Browning JL, Olsen ML. Astrocyte TrkB.T1 deficiency disrupts glutamatergic synaptogenesis and astrocyte-synapse interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619696. [PMID: 39484608 PMCID: PMC11526899 DOI: 10.1101/2024.10.22.619696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Perisynaptic astrocyte processes (PAPs) contact pre- and post-synaptic elements to provide structural and functional support to synapses. Accumulating research demonstrates that the cradling of synapses by PAPs is critical for synapse formation, stabilization, and plasticity. The specific signaling pathways that govern these astrocyte-synapse interactions, however, remain to be elucidated. Herein, we demonstrate a role for the astrocyte TrkB.T1 receptor, a truncated isoform of the canonical receptor for brain derived neurotrophic factor (BDNF), in modulating astrocyte-synapse interactions and excitatory synapse development. Neuron-astrocyte co-culture studies revealed that loss of astrocyte TrkB.T1 disrupts the formation of PAPs. To elucidate the role of TrkB.T1 in synapse development, we conditionally deleted TrkB.T1 in astrocytes in mice. Synaptosome preparations were employed to probe for TrkB.T1 localization at the PAP, and confocal three-dimensional microscopy revealed a significant reduction in synapse density and astrocyte-synapse interactions across development in the absence of astrocytic TrkB.T1. These findings suggest that BDNF/TrkB.T1 signaling in astrocytes is critical for normal excitatory synapse formation in the cortex and that astrocyte TrkB.T1 serves a requisite role in astrocyte synapse interactions. Overall, this work provides new insights into the molecular mechanisms of astrocyte-mediated synaptogenesis and may have implications for understanding neurodevelopmental disorders and developing potential therapeutic targets.
Collapse
|
14
|
Ferrario CR, Münzberg-Gruening H, Rinaman L, Betley JN, Borgland SL, Dus M, Fadool DA, Medler KF, Morton GJ, Sandoval DA, de La Serre CB, Stanley SA, Townsend KL, Watts AG, Maruvada P, Cummings D, Cooke BM. Obesity- and diet-induced plasticity in systems that control eating and energy balance. Obesity (Silver Spring) 2024; 32:1425-1440. [PMID: 39010249 PMCID: PMC11269035 DOI: 10.1002/oby.24060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024]
Abstract
In April 2023, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), in partnership with the National Institute of Child Health and Human Development, the National Institute on Aging, and the Office of Behavioral and Social Sciences Research, hosted a 2-day online workshop to discuss neural plasticity in energy homeostasis and obesity. The goal was to provide a broad view of current knowledge while identifying research questions and challenges regarding neural systems that control food intake and energy balance. This review includes highlights from the meeting and is intended both to introduce unfamiliar audiences with concepts central to energy homeostasis, feeding, and obesity and to highlight up-and-coming research in these areas that may be of special interest to those with a background in these fields. The overarching theme of this review addresses plasticity within the central and peripheral nervous systems that regulates and influences eating, emphasizing distinctions between healthy and disease states. This is by no means a comprehensive review because this is a broad and rapidly developing area. However, we have pointed out relevant reviews and primary articles throughout, as well as gaps in current understanding and opportunities for developments in the field.
Collapse
Grants
- P30 DK048520 NIDDK NIH HHS
- NSF1949989 National Science Foundation
- T32 DC000044 NIDCD NIH HHS
- R01 DK089056 NIDDK NIH HHS
- R01 DK124801 NIDDK NIH HHS
- R01 DK100685 NIDDK NIH HHS
- R01 DK130875 NIDDK NIH HHS
- R01 DK133464 NIDDK NIH HHS
- R01 DK125890 NIDDK NIH HHS
- Z99 DK999999 Intramural NIH HHS
- R01 DK124461 NIDDK NIH HHS
- K26 DK138368 NIDDK NIH HHS
- R01 DK121995 NIDDK NIH HHS
- R01 DK121531 NIDDK NIH HHS
- P30 DK089503 NIDDK NIH HHS
- P01 DK119130 NIDDK NIH HHS
- R01 DK118910 NIDDK NIH HHS
- R01 AT011683 NCCIH NIH HHS
- Reported research was supported by DK130246, DK092587, AT011683, MH059911, DK100685, DK119130, DK124801, DK133399, AG079877, DK133464, T32DC000044, F31DC016817, NSF1949989, DK089056, DK124238, DK138368, DK121995, DK125890, DK118910, DK121531, DK124461, DK130875; Canada Research Chair: 950-232211, CIHRFDN148473, CIHRPJT185886; USDA Predoctoral Fellowship; Endowment from the Robinson Family and Tallahassee Memorial Hospital; Department of Defense W81XWH-20-1-0345 and HT9425-23-1-0244; American Diabetes Association #1-17-ACE-31; W.M. Keck Foundation Award; National Science Foundation CAREER 1941822
- R01 DK133399 NIDDK NIH HHS
- HT9425-23-1-0244 Department of Defense
- R01 DK092587 NIDDK NIH HHS
- W81XWH-20-1-0345 Department of Defense
- 1941822 National Science Foundation
- R01 MH059911 NIMH NIH HHS
- F31 DC016817 NIDCD NIH HHS
- R01 AG079877 NIA NIH HHS
- R01 DK130246 NIDDK NIH HHS
- P30 DK017047 NIDDK NIH HHS
- R01 DK124238 NIDDK NIH HHS
Collapse
Affiliation(s)
- Carrie R Ferrario
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Heike Münzberg-Gruening
- Laboratory of Central Leptin Signaling, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - J Nicholas Betley
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephanie L Borgland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Debra A Fadool
- Department of Biological Science, Program in Neuroscience, Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Kathryn F Medler
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute at South Lake Union, Seattle, Washington, USA
| | - Darleen A Sandoval
- Department of Pediatrics, Section of Nutrition, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Alan G Watts
- Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Padma Maruvada
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Diana Cummings
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Bradley M Cooke
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
16
|
Frago LM, Gómez-Romero A, Collado-Pérez R, Argente J, Chowen JA. Synergism Between Hypothalamic Astrocytes and Neurons in Metabolic Control. Physiology (Bethesda) 2024; 39:0. [PMID: 38530221 DOI: 10.1152/physiol.00009.2024] [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: 02/01/2024] [Revised: 03/05/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
Astrocytes are no longer considered as passive support cells. In the hypothalamus, these glial cells actively participate in the control of appetite, energy expenditure, and the processes leading to obesity and its secondary complications. Here we briefly review studies supporting this conclusion and the advances made in understanding the underlying mechanisms.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Gómez-Romero
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
17
|
Bogard AT, Hembree TG, Pollet AK, Smith AC, Ryder SC, Marzloff G, Tan AQ. Intermittent Hypoxia-Induced Enhancements in Corticospinal Excitability Predict Gains in Motor Learning and Metabolic Efficiency. RESEARCH SQUARE 2024:rs.3.rs-4259378. [PMID: 38746438 PMCID: PMC11092812 DOI: 10.21203/rs.3.rs-4259378/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Acute intermittent hypoxia (AIH) enhances human motor function after incomplete spinal cord injury. Although the underlying mechanisms in humans are unknown, emerging evidence indicates that AIH facilitates corticospinal excitability to the upper limb. However, the functional relevance of this plasticity remains unexplored, and it is unclear whether similar plasticity can be induced for lower limb motor areas. We recently demonstrated that AIH improves motor learning and metabolic efficiency during split-belt walking. Thus, we hypothesized that AIH increases lower limb excitability and that these enhancements would predict the magnitude of motor learning and the corresponding reductions in net metabolic power. We assessed tibialis anterior (TA) excitability using transcranial magnetic stimulation and quantified changes in spatiotemporal asymmetries and net metabolic power in response to split-belt speed perturbations. We show that AIH enhances TA excitability, and that the magnitude of this facilitation positively correlates with greater spatiotemporal adaptation. Notably, we demonstrate a novel association between increased excitability and reduced net metabolic power during motor learning and savings. Together, our results suggest that AIH-induced gains in excitability predict both the magnitude of motor learning and the associated metabolic efficiency. Determining indices of AIH-induced improvements in motor performance is critical for optimizing its therapeutic reach.
Collapse
Affiliation(s)
- Alysha T. Bogard
- Sensorimotor Recovery and Neuroplasticity Lab at the University of Colorado, Boulder, Dept. of Integrative Physiology, 80309, USA
| | - Thomas G. Hembree
- Sensorimotor Recovery and Neuroplasticity Lab at the University of Colorado, Boulder, Dept. of Integrative Physiology, 80309, USA
| | - Aviva K. Pollet
- Sensorimotor Recovery and Neuroplasticity Lab at the University of Colorado, Boulder, Dept. of Integrative Physiology, 80309, USA
| | - Andrew C. Smith
- University of Colorado School of Medicine, Dept. of Physical Medicine and Rehabilitation, Aurora, 80045, USA
| | | | - George Marzloff
- Rocky Mountain Regional VA Medical Center, Aurora, 80045, USA
| | - Andrew Q. Tan
- Sensorimotor Recovery and Neuroplasticity Lab at the University of Colorado, Boulder, Dept. of Integrative Physiology, 80309, USA
- Rocky Mountain Regional VA Medical Center, Aurora, 80045, USA
- Center for Neuroscience, University of Colorado, Boulder, 80309, USA
| |
Collapse
|
18
|
Lu M, Shi J, Li X, Liu Y, Liu Y. Long-term intake of thermo-induced oxidized oil results in anxiety-like and depression-like behaviors: involvement of microglia and astrocytes. Food Funct 2024; 15:4037-4050. [PMID: 38533894 DOI: 10.1039/d3fo05302d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Frequent consumption of fried foods has been strongly associated with a higher risk of anxiety and depression, particularly among young individuals. The existing evidence has indicated that acrylamide produced from starchy foods at high temperatures can induce anxious behavior. However, there is limited research on the nerve damage caused by thermo-induced oxidized oil (TIOO). In this study, we conducted behavioral tests on mice and found that prolonged consumption of TIOO led to significant anxiety behavior and a tendency toward depression. TIOO primarily induced these two emotional disorders by affecting the differentiation of microglia, the level of inflammatory factors, the activation of astrocytes, and glutamate circulation in brain tissue. By promoting the over-differentiation of microglia into M1 microglia, TIOO disrupted their differentiation balance, resulting in an up-regulation of inflammatory factors (IL-1β, IL-6, TNF-α, NOS2) in M1 microglia and a down-regulation of neuroprotective factors IL-4/IL-10 in M2 microglia, leading to nerve damage. Moreover, TIOO activated astrocytes, accelerating their proliferation and causing GFAP precipitation, which damaged astrocytes. Meanwhile, TIOO stimulates the secretion of the BDNF and reduces the level of the glutamate receptor GLT-1 in astrocytes, leading to a disorder in the glutamate-glutamine cycle, further exacerbating nerve damage. In conclusion, this study suggests that long-term intake of thermo-induced oxidized oil can trigger symptoms of anxiety and depression.
Collapse
Affiliation(s)
- Meishan Lu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China.
| | - Jiachen Shi
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China.
| | - Xue Li
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yanjun Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China.
- Laboratory of Food Science and Human Health, College of Food Science and Engineering, Ocean University of China, 5 Yushan Road, Qingdao, 266003, China
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China.
- Future Food (Bai Ma) Research Institute, 111 Baima Road, Lishui District, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Ohira K. Localization of truncated TrkB and co-expression with full-length TrkB in the cerebral cortex of adult mice. Neuropeptides 2024; 104:102411. [PMID: 38335799 DOI: 10.1016/j.npep.2024.102411] [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: 12/12/2023] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024]
Abstract
Brain-derived neurotrophic factor (BDNF), one of the neurotrophins, and its specific receptor TrkB, are abundantly distributed in the central nervous system (CNS) and have a variety of biological effects, such as neural survival, neurite elongation, neural differentiation, and enhancing synaptic functions. Currently, there are two TrkB subtypes: full-length TrkB (TrkB-FL), which has a tyrosine kinase in the intracellular domain, and TrkB-T1, which is a tyrosine kinase-deficient form. While TrkB-FL is a typical tyrosine kinase receptor, TrkB-T1 is a main form expressed in the CNS of adult mammals, but its function is unknown. In this study, we performed fluorescent staining of the cerebral cortex of adult mice, by using TrkB-T1 antiserum and various antibodies of marker molecules for neurons and glial cells. We found that TrkB-T1 was expressed not only in neurons but also in astrocytes. In contrast, little expression of TrkB-T1 was found in oligodendrocytes and microglia. TrkB-T1 was expressed in almost all of the cells expressing TrkB-FL, indicating the direct interaction between TrkB subtypes. These findings suggest that a part of various functions of BDNF-TrkB signaling might be due to the interaction and cellular localization of TrkB subtypes in the cerebral cortex.
Collapse
Affiliation(s)
- Koji Ohira
- Laboratory of Nutritional Brain Science, Department of Food Science and Nutrition, 6-46 Ikebiraki, Nishinomiya, Hyogo 663-8558, Japan.
| |
Collapse
|
20
|
Harvey T, Rios M. The Role of BDNF and TrkB in the Central Control of Energy and Glucose Balance: An Update. Biomolecules 2024; 14:424. [PMID: 38672441 PMCID: PMC11048226 DOI: 10.3390/biom14040424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The global rise in obesity and related health issues, such as type 2 diabetes and cardiovascular disease, is alarming. Gaining a deeper insight into the central neural pathways and mechanisms that regulate energy and glucose homeostasis is crucial for developing effective interventions to combat this debilitating condition. A significant body of evidence from studies in humans and rodents indicates that brain-derived neurotrophic factor (BDNF) signaling plays a key role in regulating feeding, energy expenditure, and glycemic control. BDNF is a highly conserved neurotrophin that signals via the tropomyosin-related kinase B (TrkB) receptor to facilitate neuronal survival, differentiation, and synaptic plasticity and function. Recent studies have shed light on the mechanisms through which BDNF influences energy and glucose balance. This review will cover our current understanding of the brain regions, neural circuits, and cellular and molecular mechanisms underlying the metabolic actions of BDNF and TrkB.
Collapse
Affiliation(s)
- Theresa Harvey
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
21
|
Liu S, Lu Y, Tian D, Zhang T, Zhang C, Hu CY, Chen P, Meng Y. Hydroxytyrosol Alleviates Obesity-Induced Cognitive Decline by Modulating the Expression Levels of Brain-Derived Neurotrophic Factors and Inflammatory Factors in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6250-6264. [PMID: 38491001 DOI: 10.1021/acs.jafc.3c08319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
Hydroxytyrosol (HT; 3,4-dihydroxyphenyl ethanol) is an important functional polyphenol in olive oil. Our study sought to evaluate the protective effects and underlying mechanisms of HT on obesity-induced cognitive impairment. A high-fat and high-fructose-diet-induced obese mice model was treated with HT for 14 weeks. The results show that HT improved the learning and memory abilities and enhanced the expressions of brain-derived neurotrophic factors (BDNFs) and postsynaptic density proteins, protecting neuronal and synaptic functions in obese mice. Transcriptomic results further confirmed that HT improved cognitive impairment by regulating gene expression in neural system development and synaptic function-related pathways. Moreover, HT treatment alleviated neuroinflammation in the brain of obese mice. To sum up, our results indicated that HT can alleviate obesity-induced cognitive dysfunction by enhancing BDNF expression and alleviating neuroinflammation in the brain, which also means that HT may become a potentially useful nutritional supplement to alleviate obesity-induced cognitive decline.
Collapse
Affiliation(s)
- Shenlin Liu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Yalong Lu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Dan Tian
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Tingting Zhang
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Chaoqun Zhang
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| | - Ching Yuan Hu
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, 1955 East-West Road, AgSci. 415J, Honolulu, Hawaii 96822, United States
| | - Ping Chen
- Shaanxi Provincial Center for Disease Control and Prevention, Xian, Shaanxi 710054, P. R. China
| | - Yonghong Meng
- The Engineering Research Center of High-Valued Utilization of Fruit Resources in Western China, Ministry of Education; National Research & Development Center of Apple Processing Technology; College of Food Engineering and Nutritional Science, Shaanxi Normal University, 620 West Changan Avenue, Xian, Shaanxi 710119, P. R. China
| |
Collapse
|
22
|
Harkany T, Tretiakov E, Varela L, Jarc J, Rebernik P, Newbold S, Keimpema E, Verkhratsky A, Horvath T, Romanov R. Molecularly stratified hypothalamic astrocytes are cellular foci for obesity. RESEARCH SQUARE 2024:rs.3.rs-3748581. [PMID: 38405925 PMCID: PMC10889077 DOI: 10.21203/rs.3.rs-3748581/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Astrocytes safeguard the homeostasis of the central nervous system1,2. Despite their prominent morphological plasticity under conditions that challenge the brain's adaptive capacity3-5, the classification of astrocytes, and relating their molecular make-up to spatially devolved neuronal operations that specify behavior or metabolism, remained mostly futile6,7. Although it seems unexpected in the era of single-cell biology, the lack of a major advance in stratifying astrocytes under physiological conditions rests on the incompatibility of 'neurocentric' algorithms that rely on stable developmental endpoints, lifelong transcriptional, neurotransmitter, and neuropeptide signatures for classification6-8 with the dynamic functional states, anatomic allocation, and allostatic plasticity of astrocytes1. Simplistically, therefore, astrocytes are still grouped as 'resting' vs. 'reactive', the latter referring to pathological states marked by various inducible genes3,9,10. Here, we introduced a machine learning-based feature recognition algorithm that benefits from the cumulative power of published single-cell RNA-seq data on astrocytes as a reference map to stepwise eliminate pleiotropic and inducible cellular features. For the healthy hypothalamus, this walk-back approach revealed gene regulatory networks (GRNs) that specified subsets of astrocytes, and could be used as landmarking tools for their anatomical assignment. The core molecular censuses retained by astrocyte subsets were sufficient to stratify them by allostatic competence, chiefly their signaling and metabolic interplay with neurons. Particularly, we found differentially expressed mitochondrial genes in insulin-sensing astrocytes and demonstrated their reciprocal signaling with neurons that work antagonistically within the food intake circuitry. As a proof-of-concept, we showed that disrupting Mfn2 expression in astrocytes reduced their ability to support dynamic circuit reorganization, a time-locked feature of satiety in the hypothalamus, thus leading to obesity in mice. Overall, our results suggest that astrocytes in the healthy brain are fundamentally more heterogeneous than previously thought and topologically mirror the specificity of local neurocircuits.
Collapse
Affiliation(s)
- Tibor Harkany
- Center for Brain Research, Medical University of Vienna
| | | | | | - Jasna Jarc
- Center for Brain Research, Medical University of Vienna
| | | | | | - Erik Keimpema
- Medical University of Vienna, Center for Brain Research
| | | | | | | |
Collapse
|
23
|
Niu C, Yue X, An JJ, Bass R, Xu H, Xu B. Genetic Dissection of BDNF and TrkB Expression in Glial Cells. Biomolecules 2024; 14:91. [PMID: 38254691 PMCID: PMC10813193 DOI: 10.3390/biom14010091] [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: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL) as well as a lower level of truncated TrkB (TrkB.T). However, there are conflicting reports regarding the expression of BDNF and TrkB in glial cells, particularly microglia. In this study, we employed a sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent reporter mice to label BDNF- or TrkB-expressing cells with soma-localized EGFP. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB-expressing microglia in examined CNS regions, including the somatomotor cortex, hippocampal CA1, and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the hippocampus of adult brains. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF in the hippocampus, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the hippocampus of adult mice. These results not only clarify the expression of BDNF and TrkB in glial cells but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in astrocytes and oligodendrocytes.
Collapse
Affiliation(s)
- Changran Niu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Xinpei Yue
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Juan Ji An
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
| | - Robert Bass
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Haifei Xu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
| | - Baoji Xu
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL 33458, USA; (C.N.); (X.Y.); (J.J.A.); (R.B.); (H.X.)
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
24
|
Ameroso D, Rios M. Synaptic plasticity and the role of astrocytes in central metabolic circuits. WIREs Mech Dis 2024; 16:e1632. [PMID: 37833830 PMCID: PMC10842964 DOI: 10.1002/wsbm.1632] [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: 07/01/2023] [Revised: 09/08/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023]
Abstract
Neural circuits in the brain, primarily in the hypothalamus, are paramount to the homeostatic control of feeding and energy utilization. They integrate hunger, satiety, and body adiposity cues from the periphery and mediate the appropriate behavioral and physiological responses to satisfy the energy demands of the animal. Notably, perturbations in central homeostatic circuits have been linked to the etiology of excessive feeding and obesity. Considering the ever-changing energy requirements of the animal and required adaptations, it is not surprising that brain-feeding circuits remain plastic in adulthood and are subject to changes in synaptic strength as a consequence of nutritional status. Indeed, synapse density, probability of presynaptic transmitter release, and postsynaptic responses in hypothalamic energy balance centers are tailored to behavioral and physiological responses required to sustain survival. Mounting evidence supports key roles of astrocytes facilitating some of this plasticity. Here we discuss these synaptic plasticity mechanisms and the emerging roles of astrocytes influencing energy and glucose balance control in health and disease. This article is categorized under: Cancer > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
25
|
Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res 2023; 197:42-51. [PMID: 36780947 DOI: 10.1016/j.neures.2023.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023]
Abstract
Astrocytes are emerging in the neuroscience field as crucial modulators of brain functions, from the molecular control of synaptic plasticity to orchestrating brain-wide circuit activity for cognitive processes. The cellular pathways through which astrocytes modulate neuronal activity and plasticity are quite diverse. In this review, we focus on neurotrophic pathways, mostly those mediated by brain-derived neurotrophic factor (BDNF). Neurotrophins are a well-known family of trophic factors with pleiotropic functions in neuronal survival, maturation and activity. Within the brain, BDNF is the most abundantly expressed and most studied of all neurotrophins. While we have detailed knowledge of the effect of BDNF on neurons, much less is known about its physiology on astroglia. However, over the last years new findings emerged demonstrating that astrocytes take an active part into BDNF physiology. In this work, we discuss the state-of-the-art knowledge about astrocytes and BDNF. Indeed, astrocytes sense extracellular BDNF through its specific TrkB receptors and activate intracellular responses that greatly vary depending on the brain area, stage of development and receptors expressed. Astrocytes also uptake and recycle BDNF / proBDNF at synapses contributing to synaptic plasticity. Finally, experimental evidence is now available describing deficits in astrocytic BDNF in several neuropathologies, suggesting that astrocytic BDNF may represent a promising target for clinical translation.
Collapse
Affiliation(s)
- Martina Albini
- Department of Experimental Medicine, University of Genova, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy
| | - Fabrizia Cesca
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy.
| |
Collapse
|
26
|
Murphy-Royal C, Ching S, Papouin T. A conceptual framework for astrocyte function. Nat Neurosci 2023; 26:1848-1856. [PMID: 37857773 PMCID: PMC10990637 DOI: 10.1038/s41593-023-01448-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/01/2023] [Indexed: 10/21/2023]
Abstract
The participation of astrocytes in brain computation was hypothesized in 1992, coinciding with the discovery that these cells display a form of intracellular Ca2+ signaling sensitive to neuroactive molecules. This finding fostered conceptual leaps crystalized around the idea that astrocytes, once thought to be passive, participate actively in brain signaling and outputs. A multitude of disparate roles of astrocytes has since emerged, but their meaningful integration has been muddied by the lack of consensus and models of how we conceive the functional position of these cells in brain circuitry. In this Perspective, we propose an intuitive, data-driven and transferable conceptual framework we coin 'contextual guidance'. It describes astrocytes as 'contextual gates' that shape neural circuitry in an adaptive, state-dependent fashion. This paradigm provides fresh perspectives on principles of astrocyte signaling and its relevance to brain function, which could spur new experimental avenues, including in computational space.
Collapse
Affiliation(s)
- Ciaran Murphy-Royal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) & Département de Neurosciences, Université de Montréal, Montréal, Quebec, Canada
| | - ShiNung Ching
- Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
27
|
Liu Z, Xiao T, Liu H. Leptin signaling and its central role in energy homeostasis. Front Neurosci 2023; 17:1238528. [PMID: 38027481 PMCID: PMC10644276 DOI: 10.3389/fnins.2023.1238528] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Leptin plays a critical role in regulating appetite, energy expenditure and body weight, making it a key factor in maintaining a healthy balance. Despite numerous efforts to develop therapeutic interventions targeting leptin signaling, their effectiveness has been limited, underscoring the importance of gaining a better understanding of the mechanisms through which leptin exerts its functions. While the hypothalamus is widely recognized as the primary site responsible for the appetite-suppressing and weight-reducing effects of leptin, other brain regions have also been increasingly investigated for their involvement in mediating leptin's action. In this review, we summarize leptin signaling pathways and the neural networks that mediate the effects of leptin, with a specific emphasis on energy homeostasis.
Collapse
Affiliation(s)
- Zhaoxun Liu
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Xiao
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hailan Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
28
|
Meng A, Ameroso D, Rios M. mGluR5 in Astrocytes in the Ventromedial Hypothalamus Regulates Pituitary Adenylate Cyclase-Activating Polypeptide Neurons and Glucose Homeostasis. J Neurosci 2023; 43:5918-5935. [PMID: 37507231 PMCID: PMC10436691 DOI: 10.1523/jneurosci.0193-23.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/09/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The ventromedial hypothalamus (VMH) is a functionally heterogeneous nucleus critical for systemic energy, glucose, and lipid balance. We showed previously that the metabotropic glutamate receptor 5 (mGluR5) plays essential roles regulating excitatory and inhibitory transmission in SF1+ neurons of the VMH and facilitating glucose and lipid homeostasis in female mice. Although mGluR5 is also highly expressed in VMH astrocytes in the mature brain, its role there influencing central metabolic circuits is unknown. In contrast to the glucose intolerance observed only in female mice lacking mGluR5 in VMH SF1 neurons, selective depletion of mGluR5 in VMH astrocytes enhanced glucose tolerance without affecting food intake or body weight in both adult female and male mice. The improved glucose tolerance was associated with elevated glucose-stimulated insulin release. Astrocytic mGluR5 male and female mutants also exhibited reduced adipocyte size and increased sympathetic tone in gonadal white adipose tissue. Diminished excitatory drive and synaptic inputs onto VMH Pituitary adenylate cyclase-activating polypeptide (PACAP+) neurons and reduced activity of these cells during acute hyperglycemia underlie the observed changes in glycemic control. These studies reveal an essential role of astrocytic mGluR5 in the VMH regulating the excitatory drive onto PACAP+ neurons and activity of these cells facilitating glucose homeostasis in male and female mice.SIGNIFICANCE STATEMENT Neuronal circuits within the VMH play chief roles in the regulation of whole-body metabolic homeostasis. It remains unclear how astrocytes influence neurotransmission in this region to facilitate energy and glucose balance control. Here, we explored the role of the metabotropic glutamate receptor, mGluR5, using a mouse model with selective depletion of mGluR5 from VMH astrocytes. We show that astrocytic mGluR5 critically regulates the excitatory drive and activity of PACAP-expressing neurons in the VMH to control glucose homeostasis in both female and male mice. Furthermore, mGluR5 in VMH astrocytes influences adipocyte size and sympathetic tone in white adipose tissue. These studies provide novel insight toward the importance of hypothalamic astrocytes participating in central circuits regulating peripheral metabolism.
Collapse
Affiliation(s)
- Alice Meng
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Dominique Ameroso
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
| | - Maribel Rios
- Graduate Program in Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| |
Collapse
|
29
|
Niu C, Yue X, An JJ, Xu H, Xu B. Genetic dissection of BDNF and TrkB expression in glial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549007. [PMID: 37503044 PMCID: PMC10370033 DOI: 10.1101/2023.07.14.549007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase receptor B (TrkB) are widely expressed in the central nervous system. It is well documented that neurons express BDNF and full-length TrkB (TrkB.FL), and a lower level of truncated TrkB (TrkB.T). With conflicting results, glial cells also have been reported to express BDNF and TrkB. In the current study, we employed a more sensitive and reliable genetic method to characterize the expression of BDNF and TrkB in glial cells in the mouse brain. We utilized three Cre mouse strains in which Cre recombinase is expressed in the same cells as BDNF, TrkB.FL, or all TrkB isoforms, and crossed them to Cre-dependent EGFP reporter mice to label BDNF- or TrkB- expressing cells. We performed immunohistochemistry with glial cell markers to examine the expression of BDNF and TrkB in microglia, astrocytes, and oligodendrocytes. Surprisingly, we found no BDNF- or TrkB- expressing microglia in the brain and spinal cord. Consistent with previous studies, most astrocytes only express TrkB.T in the adult brain. Moreover, there are a small number of astrocytes and oligodendrocytes that express BDNF, the function of which is to be determined. We also found that oligodendrocyte precursor cells, but not mature oligodendrocytes, express both TrkB.FL and TrkB.T in the adult brain. These results not only clarify the expression of BDNF and TrkB in glial cells, but also open opportunities to investigate previously unidentified roles of BDNF and TrkB in glial cells.
Collapse
|
30
|
Çerçi B, Gök A, Akyol A. Brain-derived neurotrophic factor: Its role in energy balance and cancer cachexia. Cytokine Growth Factor Rev 2023; 71-72:105-116. [PMID: 37500391 DOI: 10.1016/j.cytogfr.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/29/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in the development of the central and peripheral nervous system during embryogenesis. In the mature central nervous system, BDNF is required for the maintenance and enhancement of synaptic transmissions and the survival of neurons. Particularly, it is involved in the modulation of neurocircuits that control energy balance through food intake, energy expenditure, and locomotion. Regulation of BDNF in the central nervous system is complex and environmental factors affect its expression in murine models which may reflect to phenotype dramatically. Furthermore, BDNF and its high-affinity receptor tropomyosin receptor kinase B (TrkB), as well as pan-neurotrophin receptor (p75NTR) is expressed in peripheral tissues in adulthood and their signaling is associated with regulation of energy balance. BDNF/TrkB signaling is exploited by cancer cells as well and BDNF expression is increased in tumors. Intriguingly, previously demonstrated roles of BDNF in regulation of food intake, adipose tissue and muscle overlap with derangements observed in cancer cachexia. However, data about the involvement of BDNF in cachectic cancer patients and murine models are scarce and inconclusive. In the future, knock-in and/or knock-out experiments with murine cancer models could be helpful to explore potential new roles for BDNF in the development of cancer cachexia.
Collapse
Affiliation(s)
- Barış Çerçi
- Medical School, Hacettepe University, Ankara, Turkey.
| | - Ayşenur Gök
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| | - Aytekin Akyol
- Departmant of Pathology, Medical School, Hacettepe University, Ankara, Turkey; Hacettepe University Transgenic Animal Technologies Research and Application Center, Sıhhiye, Ankara 06100, Turkey
| |
Collapse
|
31
|
Li Z, Jiang Y, Long C, Peng Q, Yue R. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci Ther 2023; 29 Suppl 1:59-73. [PMID: 36601656 PMCID: PMC10314112 DOI: 10.1111/cns.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) is one of the most insidious complications of type 2 diabetes mellitus, which can seriously affect the ability to self-monitoring of blood glucose and the quality of life in the elderly. Previous pathological studies of cognitive dysfunction have focused on neuronal dysfunction, characterized by extracellular beta-amyloid deposition and intracellular tau hyperphosphorylation. In recent years, astrocytes have been recognized as a potential therapeutic target for cognitive dysfunction and important participants in the central control of metabolism. The disorder of gut microbiota and their metabolites have been linked to a series of metabolic diseases such as diabetes mellitus. The imbalance of intestinal flora has the effect of promoting the occurrence and deterioration of several diabetes-related complications. Gut microbes and their metabolites can drive astrocyte activation. AIMS We reviewed the pathological progress of DCD related to the "gut microbiota-astrocyte" axis in terms of peripheral and central inflammation, intestinal and blood-brain barrier (BBB) dysfunction, systemic and brain energy metabolism disorders to deepen the pathological research progress of DCD and explore the potential therapeutic targets. CONCLUSION "Gut microbiota-astrocyte" axis, unique bidirectional crosstalk in the brain-gut axis, mediates the intermediate pathological process of neurocognitive dysfunction secondary to metabolic disorders in diabetes mellitus.
Collapse
Affiliation(s)
- Zi‐Han Li
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ya‐Yi Jiang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Cai‐Yi Long
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Qian Peng
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ren‐Song Yue
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
32
|
Anderson JM, Boardman AA, Bates R, Zou X, Huang W, Cao L. Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism. PLoS One 2023; 18:e0282566. [PMID: 36893171 PMCID: PMC9997972 DOI: 10.1371/journal.pone.0282566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outcomes. Brain-derived neurotrophic factor (Bdnf) and its receptor tropomyosin kinase receptor B (Ntrk2) were upregulated in the hypothalamus, hippocampus, and amygdala by implementing EE in BTBR mice, suggesting that BDNF-TrkB signaling plays a role in the EE-BTBR phenotype. Here, we used an adeno-associated virus (AAV) vector to overexpress the TrkB full-length (TrkB.FL) BDNF receptor in the BTBR mouse hypothalamus in order to assess whether hypothalamic BDNF-TrkB signaling is responsible for the improved metabolic and behavioral phenotypes associated with EE. Normal chow diet (NCD)-fed and high fat diet (HFD)-fed BTBR mice were randomized to receive either bilateral injections of AAV-TrkB.FL or AAV-YFP as control, and were subjected to metabolic and behavioral assessments up to 24 weeks post-injection. Both NCD and HFD TrkB.FL overexpressing mice displayed improved metabolic outcomes, characterized as reduced percent weight gain and increased energy expenditure. NCD TrkB.FL mice showed improved glycemic control, reduced adiposity, and increased lean mass. In NCD mice, TrkB.FL overexpression altered the ratio of TrkB.FL/TrkB.T1 protein expression and increased phosphorylation of PLCγ in the hypothalamus. TrkB.FL overexpression also upregulated expression of hypothalamic genes involved in energy regulation and altered expression of genes involved in thermogenesis, lipolysis, and energy expenditure in white adipose tissue and brown adipose tissue. In HFD mice, TrkB.FL overexpression increased phosphorylation of PLCγ. TrkB.FL overexpression in the hypothalamus did not improve behavioral deficits in either NCD or HFD mice. Together, these results suggest that enhancing hypothalamic TrkB.FL signaling improves metabolic health in BTBR mice.
Collapse
Affiliation(s)
- Jacqueline M. Anderson
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Amber A. Boardman
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Rhiannon Bates
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Xunchang Zou
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Wei Huang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
33
|
Sewaybricker LE, Huang A, Chandrasekaran S, Melhorn SJ, Schur EA. The Significance of Hypothalamic Inflammation and Gliosis for the Pathogenesis of Obesity in Humans. Endocr Rev 2023; 44:281-296. [PMID: 36251886 DOI: 10.1210/endrev/bnac023] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/12/2022] [Indexed: 11/19/2022]
Abstract
Accumulated preclinical literature demonstrates that hypothalamic inflammation and gliosis are underlying causal components of diet-induced obesity in rodent models. This review summarizes and synthesizes available translational data to better understand the applicability of preclinical findings to human obesity and its comorbidities. The published literature in humans includes histopathologic analyses performed postmortem and in vivo neuroimaging studies measuring indirect markers of hypothalamic tissue microstructure. Both support the presence of hypothalamic inflammation and gliosis in children and adults with obesity. Findings predominantly point to tissue changes in the region of the arcuate nucleus of the hypothalamus, although findings of altered tissue characteristics in whole hypothalamus or other hypothalamic regions also emerged. Moreover, the severity of hypothalamic inflammation and gliosis has been related to comorbid conditions, including glucose intolerance, insulin resistance, type 2 diabetes, and low testosterone levels in men, independent of elevated body adiposity. Cross-sectional findings are augmented by a small number of prospective studies suggesting that a greater degree of hypothalamic inflammation and gliosis may predict adiposity gain and worsening insulin sensitivity in susceptible individuals. In conclusion, existing human studies corroborate a large preclinical literature demonstrating that hypothalamic neuroinflammatory responses play a role in obesity pathogenesis. Extensive or permanent hypothalamic tissue remodeling may negatively affect the function of neuroendocrine regulatory circuits and promote the development and maintenance of elevated body weight in obesity and/or comorbid endocrine disorders.
Collapse
Affiliation(s)
| | - Alyssa Huang
- Department of Pediatrics, University of Washington, Division of Endocrinology and Diabetes, Seattle Children's Hospital, Seattle, WA 98015, USA
| | | | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
34
|
The immediate effect of overnutrition and fluoxetine treatment during the critical period of development on the hippocampus. Neurochem Int 2023; 162:105454. [PMID: 36462683 DOI: 10.1016/j.neuint.2022.105454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/17/2022] [Indexed: 12/02/2022]
Abstract
It is well known that overnutrition, overweight, and obesity in children can modulate brain mechanisms of plasticity, monoaminergic systems, and mitochondrial function. The immediate effect of overnutrition during the developmental period has not been thoroughly examined in rats until the present. This study sought to evaluate the impact on adult rats of early life overfeeding and fluoxetine treatment from post-natal day 1 (PND1) to post-natal day 21 (PND21) relative to mitochondrial function, oxidative balance, and expression of specific monoaminergic genes in the hippocampus. The following were evaluated: mitochondrial function markers, oxidative stress biomarkers, dopamine-and serotonin-related genes, and BDNF mRNA levels. Overfeeding during the lactation period deregulates cellular metabolism and the monoaminergic systems in the hippocampus. Strikingly, serotonin modulation by fluoxetine treatment protected against some of the effects of early overnutrition. We conclude that overfeeding during brain development induce detrimental effects in mitochondria and in the genes that regulate homeostatic status that can be the molecular mechanisms related to neurological diseases.
Collapse
|
35
|
Autry AE. Function of brain-derived neurotrophic factor in the hypothalamus: Implications for depression pathology. Front Mol Neurosci 2022; 15:1028223. [PMID: 36466807 PMCID: PMC9708894 DOI: 10.3389/fnmol.2022.1028223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Depression is a prevalent mental health disorder and is the number one cause of disability worldwide. Risk factors for depression include genetic predisposition and stressful life events, and depression is twice as prevalent in women compared to men. Both clinical and preclinical research have implicated a critical role for brain-derived neurotrophic factor (BDNF) signaling in depression pathology as well as therapeutics. A preponderance of this research has focused on the role of BDNF and its primary receptor tropomyosin-related kinase B (TrkB) in the cortex and hippocampus. However, much of the symptomatology for depression is consistent with disruptions in functions of the hypothalamus including changes in weight, activity levels, responses to stress, and sociability. Here, we review evidence for the role of BDNF and TrkB signaling in the regions of the hypothalamus and their role in these autonomic and behavioral functions associated with depression. In addition, we identify areas for further research. Understanding the role of BDNF signaling in the hypothalamus will lead to valuable insights for sex- and stress-dependent neurobiological underpinnings of depression pathology.
Collapse
Affiliation(s)
- Anita E. Autry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
- *Correspondence: Anita E. Autry,
| |
Collapse
|
36
|
Shao M, Yuan F, Liu J, Luo H. Mast Cell Specific Receptor Mrgprb2 Regulating Experimental Colitis is Associated with the Microbiota-Gut-Brain Axis. J Inflamm Res 2022; 15:6137-6151. [PMID: 36386594 PMCID: PMC9656444 DOI: 10.2147/jir.s383812] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
Purpose Ulcerative colitis (UC) patients have disturbances in the microbiota-gut-brain axis, and mast cells are important components of this axis. The mast cell-specific receptor Mrgprb2 has effects on host defense against bacterial infection and neurogenic inflammation, which may help mast cells act on the axis. This study analyzed how Mrgprb2 participates in the pathogenesis of UC by affecting the microbiota-gut-brain axis. Materials and Methods Mrgprb2 knockout (b2KO) mice and wild-type (WT) mice were fed 2% (w/v) dextran sulfate sodium (DSS) in drinking water for 7 days, which was then replaced with normal water for 14 days. This cycle was repeated three times. Feces were collected on Days 21, 42, and 63 for intestinal microbiota analysis, and mice were euthanized on Day 64. Hypothalamus, amygdala and colon tissues were removed and analyzed. Results Compared with WT mice, B2KO mice exhibited increased weight loss, colon shortening and colonic pathological damage after colitis induction. Analysis of the intestinal microbiota showed that b2KO mice with colitis had a significant decrease in the abundance and diversity, as well as an increase in Allobaculum and a decrease in norank_f__Muribaculaceae and Ileibacterium. In colon tissues, the expression of mucin 2 (MUC2) and junctional adhesion molecule A (JAM-A) in b2KO mice was reduced, and oxidative stress levels were higher. B2KO mice with colitis had higher corticotropin-releasing hormone (CRH), corticotropin-releasing hormone receptor 1 (CRHR1), neuropeptide Y (NPY) and brain-derived neurotrophic factor (BDNF) mRNA levels in hypothalamus tissues and glucocorticoid receptor mRNA levels in the amygdala. Conclusion In the microbiota-gut-brain axis, Mrgprb2 was involved in regulating the intestinal microbiota composition, intestinal barrier and oxidative stress levels, and was related to stress regulation, which might help to explain the pathogenesis of UC.
Collapse
Affiliation(s)
- Ming Shao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, 430060, People’s Republic of China
| | - Fangting Yuan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, 430060, People’s Republic of China
| | - Jingwen Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, 430060, People’s Republic of China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
- Hubei Key Laboratory of Digestive Diseases, Wuhan, 430060, People’s Republic of China
- Correspondence: Hesheng Luo, Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China, Email
| |
Collapse
|
37
|
Mapps AA, Boehm E, Beier C, Keenan WT, Langel J, Liu M, Thomsen MB, Hattar S, Zhao H, Tampakakis E, Kuruvilla R. Satellite glia modulate sympathetic neuron survival, activity, and autonomic function. eLife 2022; 11:74295. [PMID: 35997251 PMCID: PMC9433091 DOI: 10.7554/elife.74295] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Satellite glia are the major glial cells in sympathetic ganglia, enveloping neuronal cell bodies. Despite this intimate association, the extent to which sympathetic functions are influenced by satellite glia in vivo remains unclear. Here, we show that satellite glia are critical for metabolism, survival, and activity of sympathetic neurons and modulate autonomic behaviors in mice. Adult ablation of satellite glia results in impaired mTOR signaling, soma atrophy, reduced noradrenergic enzymes, and loss of sympathetic neurons. However, persisting neurons have elevated activity, and satellite glia-ablated mice show increased pupil dilation and heart rate, indicative of enhanced sympathetic tone. Satellite glia-specific deletion of Kir4.1, an inward-rectifying potassium channel, largely recapitulates the cellular defects observed in glia-ablated mice, suggesting that satellite glia act in part via K+-dependent mechanisms. These findings highlight neuron–satellite glia as functional units in regulating sympathetic output, with implications for disorders linked to sympathetic hyper-activity such as cardiovascular disease and hypertension.
Collapse
Affiliation(s)
- Aurelia A Mapps
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Erica Boehm
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Corinne Beier
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - William T Keenan
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Jennifer Langel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Michael Liu
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Michael B Thomsen
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | | | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
38
|
Bazzari AH, Bazzari FH. BDNF Therapeutic Mechanisms in Neuropsychiatric Disorders. Int J Mol Sci 2022; 23:ijms23158417. [PMID: 35955546 PMCID: PMC9368938 DOI: 10.3390/ijms23158417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain and functions as both a primary neurotrophic signal and a neuromodulator. It serves essential roles in neuronal development, maintenance, transmission, and plasticity, thereby influencing aging, cognition, and behavior. Accumulating evidence associates reduced central and peripheral BDNF levels with various neuropsychiatric disorders, supporting its potential utilization as a biomarker of central pathologies. Subsequently, extensive research has been conducted to evaluate restoring, or otherwise augmenting, BDNF transmission as a potential therapeutic approach. Promising results were indeed observed for genetic BDNF upregulation or exogenous administration using a multitude of murine models of neurological and psychiatric diseases. However, varying mechanisms have been proposed to underlie the observed therapeutic effects, and many findings indicate the engagement of disease-specific and other non-specific mechanisms. This is because BDNF essentially affects all aspects of neuronal cellular function through tropomyosin receptor kinase B (TrkB) receptor signaling, the disruptions of which vary between brain regions across different pathologies leading to diversified consequences on cognition and behavior. Herein, we review the neurophysiology of BDNF transmission and signaling and classify the converging and diverging molecular mechanisms underlying its therapeutic potentials in neuropsychiatric disorders. These include neuroprotection, synaptic maintenance, immunomodulation, plasticity facilitation, secondary neuromodulation, and preservation of neurovascular unit integrity and cellular viability. Lastly, we discuss several findings suggesting BDNF as a common mediator of the therapeutic actions of centrally acting pharmacological agents used in the treatment of neurological and psychiatric illness.
Collapse
Affiliation(s)
- Amjad H. Bazzari
- Faculty of Medicine, Arab American University, 13 Zababdeh, Jenin 240, Palestine
- Correspondence:
| | - Firas H. Bazzari
- Faculty of Pharmacy, Arab American University, 13 Zababdeh, Jenin 240, Palestine;
| |
Collapse
|
39
|
Gruber T, García-Cáceres C. Astroglial clean-up of satiety synapses. Nat Metab 2022; 4:505-506. [PMID: 35501600 DOI: 10.1038/s42255-022-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München & German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München & German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
40
|
Affiliation(s)
- Anne F McGettrick
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland.
| |
Collapse
|