1
|
Biddinger JE, Elson AET, Fathi PA, Sweet SR, Nishimori K, Ayala JE, Simerly RB. AgRP neurons mediate activity-dependent development of oxytocin connectivity and autonomic regulation. Proc Natl Acad Sci U S A 2024; 121:e2403810121. [PMID: 39585985 PMCID: PMC11626166 DOI: 10.1073/pnas.2403810121] [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: 02/28/2024] [Accepted: 10/24/2024] [Indexed: 11/27/2024] Open
Abstract
During postnatal life, leptin specifies neuronal inputs to the paraventricular nucleus of the hypothalamus (PVH) and activates agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus. Activity-dependent developmental mechanisms impact refinement of sensory circuits, but whether leptin-mediated postnatal neuronal activity specifies hypothalamic neural projections is largely unexplored. Here, we used chemogenetics to manipulate the activity of AgRP neurons during a discrete postnatal critical period and evaluated the development of AgRP inputs to the PVH and descending efferent outflow to the dorsal vagal complex (DVC). In leptin-deficient mice, targeting of AgRP neuronal outgrowth to PVH oxytocin neurons was reduced, and despite the lack of leptin receptors found on oxytocin neurons in the PVH, oxytocin-containing connections to the DVC were also impaired. Activation of AgRP neurons during early postnatal life not only normalized AgRP inputs to the PVH but also oxytocin outputs to the DVC in leptin-deficient mice. Blocking AgRP neuron activity during the same postnatal period reduced the density of AgRP inputs to the PVH of wild type mice, as well as the density of oxytocin-containing DVC fibers, and these innervation deficits were associated with dysregulated autonomic function. These findings suggest that leptin-mediated AgRP neuronal activity is required for the development of PVH connectivity and represents a unique activity-dependent mechanism for specification of neural pathways involved in the hypothalamic integration of autonomic responses.
Collapse
Affiliation(s)
- Jessica E. Biddinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Amanda E. T. Elson
- Developmental Neuroscience Program, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA90027
| | - Payam A. Fathi
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Serena R. Sweet
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Katsuhiko Nishimori
- Department of Obesity and Internal Inflammation, Fukushima Medical University, Fukushima City960-1295, Japan
| | - Julio E. Ayala
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| | - Richard B. Simerly
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN37232
| |
Collapse
|
2
|
Robertson RV, Meylakh N, Crawford LS, Tinoco Mendoza FA, Macey PM, Macefield VG, Keay KA, Henderson LA. Differential activation of lateral parabrachial nuclei and their limbic projections during head compared with body pain: A 7-Tesla functional magnetic resonance imaging study. Neuroimage 2024; 299:120832. [PMID: 39236852 DOI: 10.1016/j.neuroimage.2024.120832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Pain is a complex experience that involves sensory, emotional, and motivational components. It has been suggested that pain arising from the head and orofacial regions evokes stronger emotional responses than pain from the body. Indeed, recent work in rodents reports different patterns of activation in ascending pain pathways during noxious stimulation of the skin of the face when compared to noxious stimulation of the body. Such differences may dictate different activation patterns in higher brain regions, specifically in those areas processing the affective component of pain. We aimed to use ultra-high field functional magnetic resonance imaging (fMRI at 7-Tesla) to determine whether noxious thermal stimuli applied to the surface of the face and body evoke differential activation patterns within the ascending pain pathway in awake humans (n=16). Compared to the body, noxious heat stimulation to the face evoked more widespread signal changes in prefrontal cortical regions and numerous brainstem and subcortical limbic areas. Moreover, facial pain evoked significantly different signal changes in the lateral parabrachial nucleus, substantia nigra, paraventricular hypothalamus, and paraventricular thalamus, to those evoked by body pain. These results are consistent with recent preclinical findings of differential activation in the brainstem and subcortical limbic nuclei and associated cortices during cutaneous pain of the face when compared with the body. The findings suggest one potential mechanism by which facial pain could evoke a greater emotional impact than that evoked by body pain.
Collapse
Affiliation(s)
- Rebecca V Robertson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Noemi Meylakh
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Lewis S Crawford
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Fernando A Tinoco Mendoza
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California, 90095, USA
| | | | - Kevin A Keay
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, 2006, Australia.
| |
Collapse
|
3
|
Tang H, Xing X, Han Y, Gao D, Chan P, Zhang S, Xue H. A Retrospective Study of Brain-Heart Syndrome in Patients with Acute Cerebrovascular Diseases. Risk Manag Healthc Policy 2024; 17:2161-2168. [PMID: 39263551 PMCID: PMC11389706 DOI: 10.2147/rmhp.s467205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Objective To investigate the clinical characteristics, risk factors and outcomes of brain-heart syndrome (BHS) in patients with acute cerebrovascular diseases (ACVDs). Methods A retrospective analysis was conducted of 100 patients who were admitted to our hospital with ACVDs between January 2023 and December 2023. The demographic, clinical, laboratory and imaging data of the patients were collected, and the presence and severity of BHS were evaluated. The neurological and cardiac outcomes of the patients at discharge and at 12-month follow-up were also assessed. Results Out of the 100 patients, 38% had BHS, classified as mild (18%), moderate (12%) and severe (8%). The most prevalent ACVDs were cerebral infarction (58%), cerebral haemorrhage (32%) and subarachnoid haemorrhage (10%). Cardiac complications included arrhythmia (26%), myocardial ischaemia (18%) and heart failure (10%). Patients with BHS had higher results for blood pressure, heart rate, white blood cell count, C-reactive protein, IL-6, D-dimer and troponin, more severe neurological deficits, higher mortality and poorer functional outcomes. Multivariable analysis identified age, hypertension, diabetes, coronary artery disease, prior cardiovascular events, cerebral haemorrhage, brainstem infarction and hypothalamic or insular lesions as independent risk factors for BHS. Conclusion Brain-heart syndrome is a frequent, severe complication in patients with ACVD, linked with multiple risk factors and poor prognosis. Prompt diagnosis and treatment are crucial for improving patient outcomes.
Collapse
Affiliation(s)
- Hui Tang
- Department of Emergency Internal Medicine, Xuanwu Hospital of China Capital Medical University, Beijing, People's Republic of China
| | - Xiurong Xing
- Department of Emergency Internal Medicine, Xuanwu Hospital of China Capital Medical University, Beijing, People's Republic of China
| | - Yingna Han
- Department of Emergency Internal Medicine, Xuanwu Hospital of China Capital Medical University, Beijing, People's Republic of China
| | - Daiquan Gao
- Department of Neurology, Xuanwu Hospital of China Capital Medical University, Beijing, People's Republic of China
| | - Piu Chan
- Department of Neurobiology, Xuanwu Hospital of China Capital Medical University, Beijing, People's Republic of China
| | - Shengfang Zhang
- School of Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Huixin Xue
- School of Medicine, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
4
|
Zhang Z, Su J, Tang J, Chung L, Page JC, Winter CC, Liu Y, Kegeles E, Conti S, Zhang Y, Biundo J, Chalif JI, Hua CY, Yang Z, Yao X, Yang Y, Chen S, Schwab JM, Wang KH, Chen C, Prerau MJ, He Z. Spinal projecting neurons in rostral ventromedial medulla co-regulate motor and sympathetic tone. Cell 2024; 187:3427-3444.e21. [PMID: 38733990 PMCID: PMC11193620 DOI: 10.1016/j.cell.2024.04.022] [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: 08/28/2023] [Revised: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Many behaviors require the coordinated actions of somatic and autonomic functions. However, the underlying mechanisms remain elusive. By opto-stimulating different populations of descending spinal projecting neurons (SPNs) in anesthetized mice, we show that stimulation of excitatory SPNs in the rostral ventromedial medulla (rVMM) resulted in a simultaneous increase in somatomotor and sympathetic activities. Conversely, opto-stimulation of rVMM inhibitory SPNs decreased both activities. Anatomically, these SPNs innervate both sympathetic preganglionic neurons and motor-related regions in the spinal cord. Fiber-photometry recording indicated that the activities of rVMM SPNs correlate with different levels of muscle and sympathetic tone during distinct arousal states. Inhibiting rVMM excitatory SPNs reduced basal muscle and sympathetic tone, impairing locomotion initiation and high-speed performance. In contrast, silencing the inhibitory population abolished muscle atonia and sympathetic hypoactivity during rapid eye movement (REM) sleep. Together, these results identify rVMM SPNs as descending spinal projecting pathways controlling the tone of both the somatomotor and sympathetic systems.
Collapse
Affiliation(s)
- Zicong Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Junfeng Su
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jing Tang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Leeyup Chung
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jessica C Page
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Carla C Winter
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Yuchu Liu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Evgenii Kegeles
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA; PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Sara Conti
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yu Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jason Biundo
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Joshua I Chalif
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Charles Y Hua
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Zhiyun Yang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Xue Yao
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yang Yang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shuqiang Chen
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Jan M Schwab
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA; Departments of Neurology and Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Kuan Hong Wang
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Chinfei Chen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Michael J Prerau
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA; Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Biddinger JE, Elson AET, Fathi PA, Sweet SR, Nishimori K, Ayala JE, Simerly RB. AgRP neurons mediate activity-dependent development of oxytocin connectivity and autonomic regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.592838. [PMID: 38895484 PMCID: PMC11185571 DOI: 10.1101/2024.06.02.592838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
During postnatal life, the adipocyte-derived hormone leptin is required for proper targeting of neural inputs to the paraventricular nucleus of the hypothalamus (PVH) and impacts the activity of neurons containing agouti-related peptide (AgRP) in the arcuate nucleus of the hypothalamus. Activity-dependent developmental mechanisms are known to play a defining role during postnatal organization of neural circuits, but whether leptin-mediated postnatal neuronal activity specifies neural projections to the PVH or impacts downstream connectivity is largely unexplored. Here, we blocked neuronal activity of AgRP neurons during a discrete postnatal period and evaluated development of AgRP inputs to defined regions in the PVH, as well as descending projections from PVH oxytocin neurons to the dorsal vagal complex (DVC) and assessed their dependence on leptin or postnatal AgRP neuronal activity. In leptin-deficient mice, AgRP inputs to PVH neurons were significantly reduced, as well as oxytocin-specific neuronal targeting by AgRP. Moreover, downstream oxytocin projections from the PVH to the DVC were also impaired, despite the lack of leptin receptors found on PVH oxytocin neurons. Blocking AgRP neuron activity specifically during early postnatal life reduced the density of AgRP inputs to the PVH, as well as the density of projections from PVH oxytocin neurons to the DVC, and these innervation deficits were associated with dysregulated autonomic function. These findings suggest that postnatal targeting of descending PVH oxytocin projections to the DVC requires leptin-mediated AgRP neuronal activity, and represents a novel activity-dependent mechanism for hypothalamic specification of metabolic circuitry, with consequences for autonomic regulation. Significance statement Hypothalamic neural circuits maintain homeostasis by coordinating endocrine signals with autonomic responses and behavioral outputs to ensure that physiological responses remain in tune with environmental demands. The paraventricular nucleus of the hypothalamus (PVH) plays a central role in metabolic regulation, and the architecture of its neural inputs and axonal projections is a defining feature of how it receives and conveys neuroendocrine information. In adults, leptin regulates multiple aspects of metabolic physiology, but it also functions during development to direct formation of circuits controlling homeostatic functions. Here we demonstrate that leptin acts to specify the input-output architecture of PVH circuits through an activity-dependent, transsynaptic mechanism, which represents a novel means of sculpting neuroendocrine circuitry, with lasting effects on how the brain controls energy balance.
Collapse
|
6
|
Yu J, Yang Z, Sun S, Sun K, Chen W, Zhang L, Xu J, Xu Q, Liu Z, Ke J, Zhang L, Zhu Y. The effect of weighted blankets on sleep and related disorders: a brief review. Front Psychiatry 2024; 15:1333015. [PMID: 38686123 PMCID: PMC11056563 DOI: 10.3389/fpsyt.2024.1333015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
Background Sleep disorders such as insomnia can lead to a range of health problems. The high risk of side effects and drug abuse of traditional pharmacotherapy calls for a safer non-pharmacotherapy. Aims To examine the use and efficacy of weighted blankets in improving sleep and related disorders in different populations and explore the possible mechanisms. Methods A literature search was conducted using PubMed, Embase, Web of Science, MEDLINE, Cochrane Library and CNKI databases. Eligible studies included an intervention with weighted blankets and outcomes covering sleep and/or related disorders (behavioral disturbance, negative emotions and daytime symptoms). Studies using other deep pressure, compression, or exercise-related interventions were excluded. Conclusions Most of the included studies showed that weighted blankets could effectively improve sleep quality and alleviate negative emotions and daytime symptoms in patients with sleep disorders, attention deficit hyperactivity disorder, autism spectrum disorder, and other related disorders, with a possible mechanism of deep pressure touch. Recommendations Weighted blankets might be a promising tool for sleep interventions among individuals with sleep disorders in clinical settings. More high-quality and large-scale randomized controlled trials are needed to further validate the safety and efficacy of weighted blankets and explore precise mechanisms.
Collapse
Affiliation(s)
- Jie Yu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenqing Yang
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Sudan Sun
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaili Sun
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiran Chen
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Liming Zhang
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiahui Xu
- Department of Neurology/Center for Sleep Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qinglin Xu
- Department of Neurology/Center for Sleep Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Ke
- Department of Internal Medicine of Traditional Chinese Medicine, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Lisan Zhang
- Department of Neurology/Center for Sleep Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yubo Zhu
- Department of Neurology, Affiliated Hospital of Shaoxing University, Shaoxing, China
| |
Collapse
|
7
|
Toussaint B, Heinzle J, Stephan KE. A computationally informed distinction of interoception and exteroception. Neurosci Biobehav Rev 2024; 159:105608. [PMID: 38432449 DOI: 10.1016/j.neubiorev.2024.105608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
While interoception is of major neuroscientific interest, its precise definition and delineation from exteroception continue to be debated. Here, we propose a functional distinction between interoception and exteroception based on computational concepts of sensor-effector loops. Under this view, the classification of sensory inputs as serving interoception or exteroception depends on the sensor-effector loop they feed into, for the control of either bodily (physiological and biochemical) or environmental states. We explain the utility of this perspective by examining the perception of skin temperature, one of the most challenging cases for distinguishing between interoception and exteroception. Specifically, we propose conceptualising thermoception as inference about the thermal state of the body (including the skin), which is directly coupled to thermoregulatory processes. This functional view emphasises the coupling to regulation (control) as a defining property of perception (inference) and connects the definition of interoception to contemporary computational theories of brain-body interactions.
Collapse
Affiliation(s)
- Birte Toussaint
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland.
| | - Jakob Heinzle
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland; Max Planck Institute for Metabolism Research, Cologne, Germany
| |
Collapse
|
8
|
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
|
9
|
Zahner MR, Brown MC, Chandley MJ. Inactivation of the paraventricular nucleus attenuates the cardiogenic sympathetic afferent reflex in the spontaneously hypertensive rat. J Hypertens 2024; 42:70-78. [PMID: 37889604 PMCID: PMC10792548 DOI: 10.1097/hjh.0000000000003542] [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] [Indexed: 10/29/2023]
Abstract
BACKGROUND Myocardial ischemia causes the release of bradykinin, which stimulates cardiac afferents, causing sympathetic excitation and chest pain. Glutamatergic activation of the paraventricular hypothalamic nucleus (PVN) in the spontaneously hypertensive rat (SHR) drives elevated basal sympathetic activity. Thus, we tested the hypothesis that inactivation of the PVN attenuates the elevated reflex response to epicardial bradykinin in the SHR and that ionotropic PVN glutamate receptors mediate the elevated reflex. METHODS We recorded the arterial pressure and renal sympathetic nerve activity (RSNA) response to epicardial bradykinin application in anesthetized SHR and Wistar Kyoto (WKY) rats before and after PVN microinjection of GABA A agonist muscimol or ionotropic glutamate receptor antagonist kynurenic acid. RESULTS Muscimol significantly decreased the arterial pressure response to bradykinin from 180.4 ± 5.8 to 119.5 ± 6.9 mmHg in the SHR and from 111.8 ± 7.0 to 84.2 ± 8.3 mmHg in the WKY and the RSNA response from 186.2 ± 7.1 to 142.7 ± 7.3% of baseline in the SHR and from 201.0 ± 11.5 to 160.2 ± 9.3% of baseline in the WKY. Kynurenic acid significantly decreased the arterial pressure response in the SHR from 164.5 ± 5.0 to 126.2 ± 7.7 mmHg and the RSNA response from 189.9 ± 13.7to 168.5 ± 12.7% of baseline but had no effect in the WKY. CONCLUSION These results suggest that tonic PVN activity is critical for the full manifestation of the CSAR in both the WKY and SHR. Glutamatergic PVN activity contributes to the augmented CSAR observed in the SHR.
Collapse
Affiliation(s)
- Matthew R Zahner
- Department of Health Sciences, East Tennessee State University College of Public Health
| | - Mary C Brown
- Department of Health Sciences, East Tennessee State University College of Public Health
| | - Michelle J Chandley
- Department of Biomedical Science, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA
| |
Collapse
|
10
|
Winter CC, Jacobi A, Su J, Chung L, van Velthoven CTJ, Yao Z, Lee C, Zhang Z, Yu S, Gao K, Duque Salazar G, Kegeles E, Zhang Y, Tomihiro MC, Zhang Y, Yang Z, Zhu J, Tang J, Song X, Donahue RJ, Wang Q, McMillen D, Kunst M, Wang N, Smith KA, Romero GE, Frank MM, Krol A, Kawaguchi R, Geschwind DH, Feng G, Goodrich LV, Liu Y, Tasic B, Zeng H, He Z. A transcriptomic taxonomy of mouse brain-wide spinal projecting neurons. Nature 2023; 624:403-414. [PMID: 38092914 PMCID: PMC10719099 DOI: 10.1038/s41586-023-06817-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023]
Abstract
The brain controls nearly all bodily functions via spinal projecting neurons (SPNs) that carry command signals from the brain to the spinal cord. However, a comprehensive molecular characterization of brain-wide SPNs is still lacking. Here we transcriptionally profiled a total of 65,002 SPNs, identified 76 region-specific SPN types, and mapped these types into a companion atlas of the whole mouse brain1. This taxonomy reveals a three-component organization of SPNs: (1) molecularly homogeneous excitatory SPNs from the cortex, red nucleus and cerebellum with somatotopic spinal terminations suitable for point-to-point communication; (2) heterogeneous populations in the reticular formation with broad spinal termination patterns, suitable for relaying commands related to the activities of the entire spinal cord; and (3) modulatory neurons expressing slow-acting neurotransmitters and/or neuropeptides in the hypothalamus, midbrain and reticular formation for 'gain setting' of brain-spinal signals. In addition, this atlas revealed a LIM homeobox transcription factor code that parcellates the reticulospinal neurons into five molecularly distinct and spatially segregated populations. Finally, we found transcriptional signatures of a subset of SPNs with large soma size and correlated these with fast-firing electrophysiological properties. Together, this study establishes a comprehensive taxonomy of brain-wide SPNs and provides insight into the functional organization of SPNs in mediating brain control of bodily functions.
Collapse
Affiliation(s)
- Carla C Winter
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Harvard-MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Anne Jacobi
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
- F. Hoffman-La Roche, pRED, Basel, Switzerland.
| | - Junfeng Su
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Leeyup Chung
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zicong Zhang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shuguang Yu
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kun Gao
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Geraldine Duque Salazar
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Evgenii Kegeles
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Yu Zhang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Makenzie C Tomihiro
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yiming Zhang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Zhiyun Yang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Junjie Zhu
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jing Tang
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Xuan Song
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ryan J Donahue
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Qing Wang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - Ning Wang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Gabriel E Romero
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Michelle M Frank
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Alexandra Krol
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riki Kawaguchi
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Yuanyuan Liu
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| | - Zhigang He
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Yoshida T, Fujitani M, Farmer S, Harada A, Shi Z, Lee JJ, Tinajero A, Singha AK, Fujikawa T. VMHdm/c SF-1 neuronal circuits regulate skeletal muscle PGC1-α via the sympathoadrenal drive. Mol Metab 2023; 77:101792. [PMID: 37633515 PMCID: PMC10491730 DOI: 10.1016/j.molmet.2023.101792] [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: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023] Open
Abstract
OBJECTIVE To adapt to metabolically challenging environments, the central nervous system (CNS) orchestrates metabolism of peripheral organs including skeletal muscle. The organ-communication between the CNS and skeletal muscle has been investigated, yet our understanding of the neuronal pathway from the CNS to skeletal muscle is still limited. Neurons in the dorsomedial and central parts of the ventromedial hypothalamic nucleus (VMHdm/c) expressing steroidogenic factor-1 (VMHdm/cSF-1 neurons) are key for metabolic adaptations to exercise, including increased basal metabolic rate and skeletal muscle mass in mice. However, the mechanisms by which VMHdm/cSF-1 neurons regulate skeletal muscle function remain unclear. Here, we show that VMHdm/cSF-1 neurons increase the sympathoadrenal activity and regulate skeletal muscle peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) in mice via multiple downstream nodes. METHODS Optogenetics was used to specifically manipulate VMHdm/cSF-1 neurons combined with genetically-engineered mice and surgical manipulation of the sympathoadrenal activity. RESULTS Optogenetic activation of VMHdm/cSF-1 neurons dramatically elevates mRNA levels of skeletal muscle Pgc-1α, which regulates a spectrum of skeletal muscle function including protein synthesis and metabolism. Mechanistically, the sympathoadrenal drive coupled with β2 adrenergic receptor (β2AdR) is essential for VMHdm/cSF-1 neurons-mediated increases in skeletal muscle PGC1-α. Specifically, both adrenalectomy and β2AdR knockout block augmented skeletal muscle PGC1-α by VMHdm/cSF-1 neuronal activation. Optogenetic functional mapping reveals that downstream nodes of VMHdm/cSF-1 neurons are functionally redundant to increase circulating epinephrine and skeletal muscle PGC1-α. CONCLUSIONS Collectively, we propose that VMHdm/cSF-1 neurons-skeletal muscle pathway, VMHdm/cSF-1 neurons→multiple downstream nodes→the adrenal gland→skeletal muscle β2AdR, underlies augmented skeletal muscle function for metabolic adaptations.
Collapse
Affiliation(s)
- Takuya Yoshida
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Department of Clinical Nutrition School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Mina Fujitani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA; Laboratory of Nutrition Science, Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama, Japan
| | - Scotlynn Farmer
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA
| | - Ami Harada
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Nara Medical University, Nara, Japan
| | - Zhen Shi
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Department of Plastic Surgery, Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Jenny J Lee
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - Arely Tinajero
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA
| | - Ashish K Singha
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA
| | - Teppei Fujikawa
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, USA; Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
12
|
Ma L, Liu Q, Liu X, Chang H, Jin S, Ma W, Xu F, Liu H. Paraventricular Hypothalamic Nucleus Upregulates Intraocular Pressure Via Glutamatergic Neurons. Invest Ophthalmol Vis Sci 2023; 64:43. [PMID: 37773501 PMCID: PMC10547014 DOI: 10.1167/iovs.64.12.43] [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: 02/04/2023] [Accepted: 08/11/2023] [Indexed: 10/01/2023] Open
Abstract
Purpose The neuroregulatory center of intraocular pressure (IOP) is located in the hypothalamus. An efferent neural pathway exists between the hypothalamic nuclei and the autonomic nerve endings in the anterior chamber of the eye. This study was designed to investigate whether the paraventricular hypothalamic nucleus (PVH) regulates IOP as the other nuclei do. Methods Optogenetic manipulation of PVH neurons was used in this study. Light stimulation was applied via an optical fiber embedded over the PVH to activate projection neurons after AAV2/9-CaMKIIα-hChR2-mCherry was injected into the right PVH of C57BL/6J mice. The same methods were used to inhibit projection neurons after AAV2/9-CaMKIIα-eNpHR3.0-mCherry was injected into the bilateral PVH of C57BL/6J mice. AAV2/9-EF1α-DIO-hChR2-mCherry was injected into the right PVH of Vglut2-Cre mice to elucidate the effect of glutamatergic neuron-specific activation. IOP was measured before and after light manipulation. Associated nuclei activation was clarified by c-Fos immunohistochemical staining. Only mice with accurate viral expression and fiber embedding were included in the statistical analysis. Results Activation of projection neurons in the right PVH induced significant bilateral IOP elevation (n = 11, P < 0.001); the ipsilateral IOP increased more noticeably (n = 11, P < 0.05); Bilateral inhibition of PVH projection neurons did not significantly influence IOP (n = 5, P > 0.05). Specific activation of glutamatergic neurons among PVH projection neurons also induced IOP elevation in both eyes (n = 5, P < 0.001). The dorsomedial hypothalamic nucleus, ventromedial hypothalamic nucleus, locus coeruleus and basolateral amygdaloid nucleus responded to light stimulation of PVH in AAV-ChR2 mice. Conclusions The PVH may play a role in IOP upregulation via glutamatergic neurons.
Collapse
Affiliation(s)
- Lin Ma
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Chang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sen Jin
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Wenyu Ma
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Haixia Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Meth EMS, Brandão LEM, van Egmond LT, Xue P, Grip A, Wu J, Adan A, Andersson F, Pacheco AP, Uvnäs-Moberg K, Cedernaes J, Benedict C. A weighted blanket increases pre-sleep salivary concentrations of melatonin in young, healthy adults. J Sleep Res 2023; 32:e13743. [PMID: 36184925 DOI: 10.1111/jsr.13743] [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: 07/03/2022] [Revised: 09/04/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022]
Abstract
Weighted blankets have emerged as a potential non-pharmacological intervention to ease conditions such as insomnia and anxiety. Despite a lack of experimental evidence, these alleged effects are frequently attributed to a reduced activity of the endogenous stress systems and an increased release of hormones such as oxytocin and melatonin. Thus, the aim of the present in-laboratory crossover study (26 young and healthy participants, including 15 men and 11 women) was to investigate if using a weighted blanket (~12% of body weight) at bedtime resulted in higher salivary concentrations of melatonin and oxytocin compared with a light blanket (~2.4% of body weight). We also examined possible differences in salivary concentrations of the stress hormone cortisol, salivary alpha-amylase activity (as an indicative metric of sympathetic nervous system activity), subjective sleepiness, and sleep duration. When using a weighted blanket, the 1 hour increase of salivary melatonin from baseline (i.e., 22:00) to lights off (i.e., 23:00) was about 32% higher (p = 0.011). No other significant differences were found between the blanket conditions, including subjective sleepiness and total sleep duration. Our study is the first to suggest that using a weighted blanket may result in a more significant release of melatonin at bedtime. Future studies should investigate whether the stimulatory effect on melatonin secretion is observed on a nightly basis when frequently using a weighted blanket over weeks to months. It remains to be determined whether the observed increase in melatonin may be therapeutically relevant for the previously described effects of the weighted blanket on insomnia and anxiety.
Collapse
Affiliation(s)
- Elisa M S Meth
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - Lieve T van Egmond
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Pei Xue
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Anastasia Grip
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Jiafei Wu
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ayaat Adan
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - André P Pacheco
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Kerstin Uvnäs-Moberg
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jonathan Cedernaes
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Murthy VD, Phillips K, Knipe M, Giuffrida M, Li CF. Horner syndrome as a physiological biomarker of disease in canine cervical myelopathy. J Vet Intern Med 2023; 37:598-605. [PMID: 36704850 DOI: 10.1111/jvim.16588] [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: 07/06/2022] [Accepted: 11/11/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Horner syndrome often occurs with cervical myelopathies and might provide insight into the underlying disease and prognosis. OBJECTIVES To describe the clinical and imaging features of dogs with cervical myelopathy and concurrent Horner syndrome and to determine association of Horner syndrome with diseases or magnetic resonance images (MRI). ANIMALS Ninety-three client-owned dogs with cervical myelopathy and concurrent Horner syndrome and 99 randomly selected client-owned dogs with cervical myelopathy without Horner syndrome (control cases). METHODS Retrospective study. Medical records were reviewed to identify Horner and control cases and clinical findings recorded. MRI were reviewed, and lesions characterized and recorded. Descriptive and comparative statistics were performed. RESULTS Non-compressive disease occurred more frequently in the Horner group compared with controls (58%; 95% CI: 48-68 vs 9%; 95% CI: 5-16; P < .0001). The most common diseases were fibrocartilaginous embolism in the Horner group (44/93; 47%) and intervertebral disc extrusion (76/99; 77%) amongst controls. On MRI, parenchymal hyperintensity was seen more commonly in the Horner group (95%; 95% CI: 88-98) compared with controls (51%; 95% CI: 41-60; P < .0001). In the Horner group, dogs that did not survive to discharge (N = 13) had more extensive MRI lesions relative to the adjacent vertebral length (200%; IQR 110%-575%) compared with survivors (N = 80; 110%; IQR 40%-250%; P = .02). Lateralization of Horner signs and MRI changes matched in 54% of cases. The overall survival rate was high in both Horner (80/93; 86%) and control (95/99; 96%) groups. CONCLUSIONS AND CLINICAL IMPORTANCE Horner syndrome in cervical myelopathy is commonly associated with noncompressive intraparenchymal disease.
Collapse
Affiliation(s)
- Vishal D Murthy
- Department of Surgical and Radiological Sciences, College of Veterinary Medicine, University of California, Davis, California, USA
| | - Kathryn Phillips
- Department of Surgical and Radiological Sciences, College of Veterinary Medicine, University of California, Davis, California, USA
| | - Marguerite Knipe
- Department of Surgical and Radiological Sciences, College of Veterinary Medicine, University of California, Davis, California, USA
| | - Michelle Giuffrida
- Department of Surgical and Radiological Sciences, College of Veterinary Medicine, University of California, Davis, California, USA
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, College of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
15
|
Baumer-Harrison C, Breza JM, Sumners C, Krause EG, de Kloet AD. Sodium Intake and Disease: Another Relationship to Consider. Nutrients 2023; 15:535. [PMID: 36771242 PMCID: PMC9921152 DOI: 10.3390/nu15030535] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/14/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023] Open
Abstract
Sodium (Na+) is crucial for numerous homeostatic processes in the body and, consequentially, its levels are tightly regulated by multiple organ systems. Sodium is acquired from the diet, commonly in the form of NaCl (table salt), and substances that contain sodium taste salty and are innately palatable at concentrations that are advantageous to physiological homeostasis. The importance of sodium homeostasis is reflected by sodium appetite, an "all-hands-on-deck" response involving the brain, multiple peripheral organ systems, and endocrine factors, to increase sodium intake and replenish sodium levels in times of depletion. Visceral sensory information and endocrine signals are integrated by the brain to regulate sodium intake. Dysregulation of the systems involved can lead to sodium overconsumption, which numerous studies have considered causal for the development of diseases, such as hypertension. The purpose here is to consider the inverse-how disease impacts sodium intake, with a focus on stress-related and cardiometabolic diseases. Our proposition is that such diseases contribute to an increase in sodium intake, potentially eliciting a vicious cycle toward disease exacerbation. First, we describe the mechanism(s) that regulate each of these processes independently. Then, we highlight the points of overlap and integration of these processes. We propose that the analogous neural circuitry involved in regulating sodium intake and blood pressure, at least in part, underlies the reciprocal relationship between neural control of these functions. Finally, we conclude with a discussion on how stress-related and cardiometabolic diseases influence these circuitries to alter the consumption of sodium.
Collapse
Affiliation(s)
- Caitlin Baumer-Harrison
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Joseph M. Breza
- Department of Psychology, College of Arts and Sciences, Eastern Michigan University, Ypsilanti, MI 48197, USA
| | - Colin Sumners
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Eric G. Krause
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Annette D. de Kloet
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL 32610, USA
- Evelyn F. and William L. McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
16
|
Silva TM, Wasinski F, Flor KC, List EO, Kopchick JJ, Takakura AC, Donato J, Moreira TS. The effect of central growth hormone action on hypoxia ventilatory response in conscious mice. Brain Res 2022; 1791:147995. [PMID: 35779583 PMCID: PMC10938300 DOI: 10.1016/j.brainres.2022.147995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
Growth hormone (GH)-responsive neurons regulate several homeostatic behaviors including metabolism, energy balance, arousal, and stress response. Therefore, it is possible that GH-responsive neurons play a role in other responses such as CO2/H+-dependent breathing behaviors. Here, we investigated whether central GH receptor (GHR) modulates respiratory activity in conscious unrestrained mice. First, we detected clusters of GH-responsive neurons in the tyrosine hydroxylase-expressing cells in the rostroventrolateral medulla (C1 region) and within the locus coeruleus (LC). No significant expression was detected in phox2b-expressing cells in the retrotrapezoid nucleus. Whole body plethysmography revealed a reduction in the tachypneic response to hypoxia (FiO2 = 0.08) without changing baseline breathing and the hypercapnic ventilatory response. Contrary to the physiological findings, we did not find significant differences in the number of fos-activated cells in the nucleus of the solitary tract (NTS), C1, LC and paraventricular nucleus of the hypothalamus (PVH). Our finding suggests a possible secondary role of central GH action in the tachypneic response to hypoxia in conscious mice.
Collapse
Affiliation(s)
- Talita M Silva
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Frederick Wasinski
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Karine C Flor
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil.
| |
Collapse
|
17
|
Chen X, Xu L, Li Z. Autonomic Neural Circuit and Intervention for Comorbidity Anxiety and Cardiovascular Disease. Front Physiol 2022; 13:852891. [PMID: 35574459 PMCID: PMC9092179 DOI: 10.3389/fphys.2022.852891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/21/2022] [Indexed: 11/28/2022] Open
Abstract
Anxiety disorder is a prevalent psychiatric disease and imposes a significant influence on cardiovascular disease (CVD). Numerous evidence support that anxiety contributes to the onset and progression of various CVDs through different physiological and behavioral mechanisms. However, the exact role of nuclei and the association between the neural circuit and anxiety disorder in CVD remains unknown. Several anxiety-related nuclei, including that of the amygdala, hippocampus, bed nucleus of stria terminalis, and medial prefrontal cortex, along with the relevant neural circuit are crucial in CVD. A strong connection between these nuclei and the autonomic nervous system has been proven. Therefore, anxiety may influence CVD through these autonomic neural circuits consisting of anxiety-related nuclei and the autonomic nervous system. Neuromodulation, which can offer targeted intervention on these nuclei, may promote the development of treatment for comorbidities of CVD and anxiety disorders. The present review focuses on the association between anxiety-relevant nuclei and CVD, as well as discusses several non-invasive neuromodulations which may treat anxiety and CVD.
Collapse
Affiliation(s)
- Xuanzhao Chen
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Li Xu
- Department of Rheumatology and Immunology, General Hospital of Central Theater Command, Wuhan, China
| | - Zeyan Li
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
18
|
Schaeuble D, Myers B. Cortical–Hypothalamic Integration of Autonomic and Endocrine Stress Responses. Front Physiol 2022; 13:820398. [PMID: 35222086 PMCID: PMC8874315 DOI: 10.3389/fphys.2022.820398] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/19/2022] [Indexed: 12/18/2022] Open
Abstract
The prevalence and severity of cardiovascular disease (CVD) are exacerbated by chronic stress exposure. While stress-induced sympathetic activity and elevated glucocorticoid secretion impair cardiovascular health, the mechanisms by which stress-responsive brain regions integrate autonomic and endocrine stress responses remain unclear. This review covers emerging literature on how specific cortical and hypothalamic nuclei regulate cardiovascular and neuroendocrine stress responses. We will also discuss the current understanding of the cellular and circuit mechanisms mediating physiological stress responses. Altogether, the reviewed literature highlights the current state of stress integration research, as well unanswered questions about the brain basis of CVD risk.
Collapse
|
19
|
Ultrastructural Evidence for Oxytocin and Oxytocin Receptor at the Spinal Dorsal Horn: Mechanism of Nociception Modulation. Neuroscience 2021; 475:117-126. [PMID: 34530103 DOI: 10.1016/j.neuroscience.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/07/2023]
Abstract
Oxytocin is a hypothalamic neuropeptide involved in the inhibition of nociception transmission at spinal dorsal horn (SDH) level (the first station where the incoming peripheral signals is modulated). Electrophysiological, behavioral, and pharmacological data strongly support the role of this neuropeptide and its receptor (the oxytocin receptor, OTR) as a key endogenous molecule with analgesic properties. Briefly, current data showed that oxytocin release from the hypothalamus induces OTR activation at the SDH, inducing selective inhibition of the nociceptive Aδ- and C-fibers (probably peptidergic) activity, but not the activity of proprioceptive fibers (i.e. Aβ-fibers). The above inhibition could be a direct presynaptic mechanism, or a mechanism mediated by GABAergic interneurons. However, the exact anatomical localization of oxytocin and OTR remains unclear. In this context, the present study set out to analyze the role of OTRs, GABAergic cells and CGRP fibers in the SDH in rats by using electron microscopy. Ultrastructural analyses of the SDH tissue show that: (i) oxytocin and OTR are found in asymmetrical synapsis; (ii) OTR is found in GABAergic interneurons (near unmyelinated fibers), CGRPergic fibers and glial cells; (iii) whereas oxytocin is present in supraspinal descending projection fibers. These anatomical data strongly support the notion that oxytocin released at the SDH could presynaptically inhibit the nociceptive input from the peripheral primary afferent fibers. This inhibitory action could be direct or use a GABA interneuron. Furthermore, our findings that OTR is exhibited in glial tissue at the SDH requires further exploration in nociception assays.
Collapse
|
20
|
Pittman QJ. Vasopressin and central control of the cardiovascular system: A 40-year retrospective. J Neuroendocrinol 2021; 33:e13011. [PMID: 34235812 DOI: 10.1111/jne.13011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/25/2021] [Indexed: 01/24/2023]
Abstract
In the 40 years since vasopressin (AVP) was reported to have a central action with respect to raising blood pressure, the finding has been repeatedly replicated using a variety of complimentary approaches. The role of AVP as a central neurotransmitter involved in control of the cardiovascular system is now textbook material. However, it is evident that brain AVP plays, at best, a minor role in regulation of normal blood pressure. However, it appears to be an important player in a several cardiovascular-associated pathologies, ranging from hypertension to neural changes associated with heart failure. There are many interventions that have been shown to affect neural function, many of which are associated with alterations in behaviour. Possible alterations in neuronal AVP actions relevant to cardiovascular control in the setting of chronic inflammatory disease, early-life stress and inflammation are suggested areas for future research.
Collapse
Affiliation(s)
- Quentin J Pittman
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
Cano G, Hernan SL, Sved AF. Centrally Projecting Edinger-Westphal Nucleus in the Control of Sympathetic Outflow and Energy Homeostasis. Brain Sci 2021; 11:1005. [PMID: 34439626 PMCID: PMC8392615 DOI: 10.3390/brainsci11081005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
The centrally projecting Edinger-Westphal nucleus (EWcp) is a midbrain neuronal group, adjacent but segregated from the preganglionic Edinger-Westphal nucleus that projects to the ciliary ganglion. The EWcp plays a crucial role in stress responses and in maintaining energy homeostasis under conditions that require an adjustment of energy expenditure, by virtue of modulating heart rate and blood pressure, thermogenesis, food intake, and fat and glucose metabolism. This modulation is ultimately mediated by changes in the sympathetic outflow to several effector organs, including the adrenal gland, heart, kidneys, brown and white adipose tissues and pancreas, in response to environmental conditions and the animal's energy state, providing for appropriate energy utilization. Classic neuroanatomical studies have shown that the EWcp receives inputs from forebrain regions involved in these functions and projects to presympathetic neuronal populations in the brainstem. Transneuronal tracing with pseudorabies virus has demonstrated that the EWcp is connected polysynaptically with central circuits that provide sympathetic innervation to all these effector organs that are critical for stress responses and energy homeostasis. We propose that EWcp integrates multimodal signals (stress, thermal, metabolic, endocrine, etc.) and modulates the sympathetic output simultaneously to multiple effector organs to maintain energy homeostasis under different conditions that require adjustments of energy demands.
Collapse
Affiliation(s)
- Georgina Cano
- Department of Neuroscience, A210 Langley Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA; (S.L.H.); (A.F.S.)
| | | | | |
Collapse
|
22
|
Piñol RA, Mogul AS, Hadley CK, Saha A, Li C, Škop V, Province HS, Xiao C, Gavrilova O, Krashes MJ, Reitman ML. Preoptic BRS3 neurons increase body temperature and heart rate via multiple pathways. Cell Metab 2021; 33:1389-1403.e6. [PMID: 34038711 PMCID: PMC8266747 DOI: 10.1016/j.cmet.2021.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/03/2021] [Accepted: 05/03/2021] [Indexed: 11/15/2022]
Abstract
The preoptic area (POA) is a key brain region for regulation of body temperature (Tb), dictating thermogenic, cardiovascular, and behavioral responses that control Tb. Previously characterized POA neuronal populations all reduced Tb when activated. Using mice, we now identify POA neurons expressing bombesin-like receptor 3 (POABRS3) as a population whose activation increased Tb; inversely, acute inhibition of these neurons reduced Tb. POABRS3 neurons that project to either the paraventricular nucleus of the hypothalamus or the dorsomedial hypothalamus increased Tb, heart rate, and blood pressure via the sympathetic nervous system. Long-term inactivation of POABRS3 neurons caused increased Tb variability, overshooting both increases and decreases in Tb set point, with RNA expression profiles suggesting multiple types of POABRS3 neurons. Thus, POABRS3 neuronal populations regulate Tb and heart rate, contribute to cold defense, and fine-tune feedback control of Tb. These findings advance understanding of homeothermy, a defining feature of mammalian biology.
Collapse
Affiliation(s)
- Ramón A Piñol
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Allison S Mogul
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Colleen K Hadley
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Atreyi Saha
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chia Li
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vojtěch Škop
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haley S Province
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cuiying Xiao
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Tenorio-Lopes L, Kinkead R. Sex-Specific Effects of Stress on Respiratory Control: Plasticity, Adaptation, and Dysfunction. Compr Physiol 2021; 11:2097-2134. [PMID: 34107062 DOI: 10.1002/cphy.c200022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As our understanding of respiratory control evolves, we appreciate how the basic neurobiological principles of plasticity discovered in other systems shape the development and function of the respiratory control system. While breathing is a robust homeostatic function, there is growing evidence that stress disrupts respiratory control in ways that predispose to disease. Neonatal stress (in the form of maternal separation) affects "classical" respiratory control structures such as the peripheral O2 sensors (carotid bodies) and the medulla (e.g., nucleus of the solitary tract). Furthermore, early life stress disrupts the paraventricular nucleus of the hypothalamus (PVH), a structure that has emerged as a primary determinant of the intensity of the ventilatory response to hypoxia. Although underestimated, the PVH's influence on respiratory function is a logical extension of the hypothalamic control of metabolic demand and supply. In this article, we review the functional and anatomical links between the stress neuroendocrine axis and the medullary network regulating breathing. We then present the persistent and sex-specific effects of neonatal stress on respiratory control in adult rats. The similarities between the respiratory phenotype of stressed rats and clinical manifestations of respiratory control disorders such as sleep-disordered breathing and panic attacks are remarkable. These observations are in line with the scientific consensus that the origins of adult disease are often found among developmental and biological disruptions occurring during early life. These observations bring a different perspective on the structural hierarchy of respiratory homeostasis and point to new directions in our understanding of the etiology of respiratory control disorders. © 2021 American Physiological Society. Compr Physiol 11:1-38, 2021.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
24
|
Kiss A, Osacka J. The effect of amisulpride, olanzapine, quetiapine, and aripiprazole single administration on c-Fos expression in vasopressinergic and oxytocinergic neurons of the rat hypothalamic paraventricular nucleus. Neuropeptides 2021; 87:102148. [PMID: 33887540 DOI: 10.1016/j.npep.2021.102148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/07/2020] [Accepted: 01/21/2021] [Indexed: 11/25/2022]
Abstract
Antipsychotics, including amisulpride (AMI), quetiapine (QUE), aripiprazole (ARI), and olanzapine (OLA), are used to treat mental illnesses associated with psychotic symptoms. The effect of these drugs on c-Fos expression in vasopressinergic (AVP) and oxytocinergic (OXY) neurons was studied in the hypothalamic paraventricular nucleus (PVN) of rats. The presence of c-Fos in AVP and OXY perikarya was investigated in seven PVN cells segregations: the anterior (Ant), dorsal cup (Dc), wing-shaped (Wi), periventricular zone (Pe), circle-shaped core (Co) and shell of core (Sh), and the posterior (pPVN) after an acute treatment with AMI-20 mg/kg, QUE-15 mg/kg, ARI-10 mg/kg, and OLA-5 mg/kg/bw in rats. Ninety min after treatments, the animals were sacrificed by transcardial perfusion with fixative and the PVN area sliced into 35 μm thick coronal sections for immunohistochemistry. The c-Fos was processed by avidin-biotin-peroxidase complex intensified with nickel-enhanced 3,3'-diaminobenzidine tetrahydrochloride. Visualization of AVP- and OXY-synthesizing neurons was achieved by a fluorescent marker Alexa Flour 568. The c-Fos-AVP and c-Fos-OXY colocalizations were evaluated from c-Fos stained sections merged with AVP or OXY ones. AMI, QUE, ARI, and OLA, single administration distinctly increased the c-Fos expression in each of the PVN cells segregations. QUE induced the highest magnitude of activation of AVP and OXY neurons, while OLA and AMI had only moderate effects. Incontestable variabilities detected in c-Fos expression in PVN AVP and OXY neurons extend the knowledge of selected antipsychotics extra-striatal actions and may also be helpful in a presumption of their possible functional impact.
Collapse
Affiliation(s)
- Alexander Kiss
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia.
| | - Jana Osacka
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia
| |
Collapse
|
25
|
Ueno M. Restoring neuro-immune circuitry after brain and spinal cord injuries. Int Immunol 2021; 33:311-325. [PMID: 33851981 DOI: 10.1093/intimm/dxab017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Neuro-immune interactions are essential for our body's defense and homeostasis. Anatomical and physiological analyses have shown that the nervous system comprises multiple pathways that regulate the dynamics and functions of immune cells, which are mainly mediated by the autonomic nervous system and adrenal signals. These are disturbed when the neurons and circuits are damaged by diseases of the central nervous system (CNS). Injuries caused by stroke or trauma often cause immune dysfunction by abrogation of the immune-regulating neural pathways, which leads to an increased risk of infections. Here, I review the structures and functions of the neural pathways connecting the brain and the immune system, and the neurogenic mechanisms of immune dysfunction that emerge after CNS injuries. Recent technological advances in manipulating specific neural circuits have added mechanistic aspects of neuro-immune interactions and their dysfunctions. Understanding the neural bases of immune control and their pathological processes will deepen our knowledge of homeostasis and lead to the development of strategies to cure immune deficiencies observed in various CNS disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Niigata 951-8585, Japan
| |
Collapse
|
26
|
Barbier M, González JA, Houdayer C, Burdakov D, Risold P, Croizier S. Projections from the dorsomedial division of the bed nucleus of the stria terminalis to hypothalamic nuclei in the mouse. J Comp Neurol 2021; 529:929-956. [PMID: 32678476 PMCID: PMC7891577 DOI: 10.1002/cne.24988] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022]
Abstract
As stressful environment is a potent modulator of feeding, we seek in the present work to decipher the neuroanatomical basis for an interplay between stress and feeding behaviors. For this, we combined anterograde and retrograde tracing with immunohistochemical approaches to investigate the patterns of projections between the dorsomedial division of the bed nucleus of the stria terminalis (BNST), well connected to the amygdala, and hypothalamic structures such as the paraventricular (PVH) and dorsomedial (DMH), the arcuate (ARH) nuclei and the lateral hypothalamic areas (LHA) known to control feeding and motivated behaviors. We particularly focused our study on afferences to proopiomelanocortin (POMC), agouti-related peptide (AgRP), melanin-concentrating-hormone (MCH) and orexin (ORX) neurons characteristics of the ARH and the LHA, respectively. We found light to intense innervation of all these hypothalamic nuclei. We particularly showed an innervation of POMC, AgRP, MCH and ORX neurons by the dorsomedial and dorsolateral divisions of the BNST. Therefore, these results lay the foundation for a better understanding of the neuroanatomical basis of the stress-related feeding behaviors.
Collapse
Affiliation(s)
- Marie Barbier
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
- Department of PsychiatrySeaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - J. Antonio González
- The Francis Crick InstituteLondonUK
- The Rowett Institute, School of MedicineMedical Sciences and Nutrition, University of AberdeenAberdeenUK
| | - Christophe Houdayer
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Denis Burdakov
- The Francis Crick InstituteLondonUK
- Neurobehavioural Dynamics Lab, Institute for Neuroscience, D‐HESTSwiss Federal Institute of Technology / ETH ZürichZürichSwitzerland
| | - Pierre‐Yves Risold
- EA481, Neurosciences Intégratives et Cliniques, UFR SantéUniversité Bourgogne Franche‐ComtéBesançonFrance
| | - Sophie Croizier
- University of LausanneCenter for Integrative GenomicsLausanneSwitzerland
| |
Collapse
|
27
|
Browning KN, Carson KE. Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 2021; 13:nu13030908. [PMID: 33799575 PMCID: PMC7998662 DOI: 10.3390/nu13030908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of energy balance requires the complex integration of homeostatic and hedonic pathways, but sensory inputs from the gastrointestinal (GI) tract are increasingly recognized as playing critical roles. The stomach and small intestine relay sensory information to the central nervous system (CNS) via the sensory afferent vagus nerve. This vast volume of complex sensory information is received by neurons of the nucleus of the tractus solitarius (NTS) and is integrated with responses to circulating factors as well as descending inputs from the brainstem, midbrain, and forebrain nuclei involved in autonomic regulation. The integrated signal is relayed to the adjacent dorsal motor nucleus of the vagus (DMV), which supplies the motor output response via the efferent vagus nerve to regulate and modulate gastric motility, tone, secretion, and emptying, as well as intestinal motility and transit; the precise coordination of these responses is essential for the control of meal size, meal termination, and nutrient absorption. The interconnectivity of the NTS implies that many other CNS areas are capable of modulating vagal efferent output, emphasized by the many CNS disorders associated with dysregulated GI functions including feeding. This review will summarize the role of major CNS centers to gut-related inputs in the regulation of gastric function with specific reference to the regulation of food intake.
Collapse
|
28
|
Gautron L. The Phantom Satiation Hypothesis of Bariatric Surgery. Front Neurosci 2021; 15:626085. [PMID: 33597843 PMCID: PMC7882491 DOI: 10.3389/fnins.2021.626085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/06/2021] [Indexed: 01/26/2023] Open
Abstract
The excitation of vagal mechanoreceptors located in the stomach wall directly contributes to satiation. Thus, a loss of gastric innervation would normally be expected to result in abrogated satiation, hyperphagia, and unwanted weight gain. While Roux-en-Y-gastric bypass (RYGB) inevitably results in gastric denervation, paradoxically, bypassed subjects continue to experience satiation. Inspired by the literature in neurology on phantom limbs, I propose a new hypothesis in which damage to the stomach innervation during RYGB, including its vagal supply, leads to large-scale maladaptive changes in viscerosensory nerves and connected brain circuits. As a result, satiation may continue to arise, sometimes at exaggerated levels, even in subjects with a denervated or truncated stomach. The same maladaptive changes may also contribute to dysautonomia, unexplained pain, and new emotional responses to eating. I further revisit the metabolic benefits of bariatric surgery, with an emphasis on RYGB, in the light of this phantom satiation hypothesis.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
29
|
Resilience in the suprachiasmatic nucleus: Implications for aging and Alzheimer's disease. Exp Gerontol 2021; 147:111258. [PMID: 33516909 DOI: 10.1016/j.exger.2021.111258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/27/2020] [Accepted: 01/24/2021] [Indexed: 12/13/2022]
Abstract
Many believe that the circadian impairments associated with aging and Alzheimer's disease are, simply enough, a byproduct of tissue degeneration within the central pacemaker, the suprachiasmatic nucleus (SCN). However, the findings that have accumulated to date examining the SCNs obtained postmortem from the brains of older individuals, or those diagnosed with Alzheimer's disease upon autopsy, suggest only limited atrophy. We review this literature as well as a complementary one concerning fetal-donor SCN transplant, which established that many circadian timekeeping functions can be maintained with rudimentary (structurally limited) representations of the SCN. Together, these corpora of data suggest that the SCN is a resilient brain region that cannot be directly (or solely) implicated in the behavioral manifestations of circadian disorganization often witnessed during aging as well as early and late progression of Alzheimer's disease. We complete our review by suggesting future directions of research that may bridge this conceptual divide and briefly discuss the implications of it for improving health outcomes in later adulthood.
Collapse
|
30
|
Kreier F, Swaab DF. History of hypothalamic research: "The spring of primitive existence". HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:7-43. [PMID: 34225985 DOI: 10.1016/b978-0-12-819975-6.00031-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The central brain region of interest for neuroendocrinology is the hypothalamus, a name coined by Wilhelm His in 1893. Neuroendocrinology is the discipline that studies hormone production by neurons, the sensitivity of neurons for hormones, as well as the dynamic, bidirectional interactions between neurons and endocrine glands. These interactions do not only occur through hormones, but are also partly accomplished by the autonomic nervous system that is regulated by the hypothalamus and that innervates the endocrine glands. A special characteristic of the hypothalamus is that it contains neuroendocrine neurons projecting either to the neurohypophysis or to the portal vessels of the anterior lobe of the pituitary in the median eminence, where they release their neuropeptides or other neuroactive compounds into the bloodstream, which subsequently act as neurohormones. In the 1970s it was found that vasopressin and oxytocin not only are released as hormones in the circulation but that their neurons project to other neurons within and outside the hypothalamus and function as neurotransmitters or neuromodulators that regulate central functions, including the autonomic innervation of all our body organs. Recently magnocellular oxytocin neurons were shown to send not only an axon to the neurohypophysis, but also axon collaterals of the same neuroendocrine neuron to a multitude of brain areas. In this way, the hypothalamus acts as a central integrator for endocrine, autonomic, and higher brain functions. The history of neuroendocrinology is described in this chapter from the descriptions in De humani corporis fabrica by Vesalius (1537) to the present, with a timeline of the scientists and their findings.
Collapse
Affiliation(s)
- Felix Kreier
- Department Pediatrics, OLVG Hospitals, Amsterdam, The Netherlands.
| | - Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Dudás B. Anatomy and cytoarchitectonics of the human hypothalamus. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:45-66. [PMID: 34225981 DOI: 10.1016/b978-0-12-819975-6.00001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Due to the complexity of hypothalamic functions, the organization of the hypothalamus is extremely intricate. This relatively small brain area contains several nuclei, most of them are ill-defined regions without distinct boundaries; these nuclei are often connected with each other and other distant brain regions with similarly indistinct pathways. These hypothalamic centers control numerous key physiological functions including reproduction, growth, food intake, circadian rhythm, behavior, and autonomic balance via neural and endocrine signals. To understand the morphology of the hypothalamus is therefore extremely important, though it remains a stupendous task due to the complex organization of neuronal networks formed by the various neurotransmitter and neuromodulator systems.
Collapse
Affiliation(s)
- Bertalan Dudás
- Neuroendocrine Organization Laboratory, Lake Erie College of Osteopathic Medicine, Erie, PA, United States; Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary.
| |
Collapse
|
32
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
33
|
Martínez-Lorenzana G, Gamal-Eltrabily M, Tello-García IA, Martínez-Torres A, Becerra-González M, González-Hernández A, Condés-Lara M. CLARITY with neuronal tracing and immunofluorescence to study the somatosensory system in rats. J Neurosci Methods 2020; 350:109048. [PMID: 33359224 DOI: 10.1016/j.jneumeth.2020.109048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND The CLARITY technique enables researchers to visualize different neuronal connections along the nervous system including the somatosensory system. NEW METHOD The present work describes the antero-lateral and dorsal column pathways until the thalamic and cortical stations, as well as descending oxytocinergic and vasopressinergic innervations by means of combined CLARITY, neuronal tracing, and immunofluorescence techniques. We used male Sprague-Dawley rats of 13, 30, and 60 days. RESULTS The main results are as follows: A) CLARITY is a reliable technique that can be combined with fluorescent neuronal tracers and immunofluorescence techniques without major procedure modifications; B) at spinal level, some primary afferent fibers were labeled by CGRP, as well as the presence of neuronal populations that simultaneously project to the gracile and ventral posterolateral thalamic nuclei; C) corticothalamic connections were visible when retrograde tracers were injected at thalamic level; D) oxytocin receptors were expressed in the spinal dorsal horn by GABAergic-positive neurons, reinforcing previous outcomes about the possible mechanism for oxytocin blocking the primary afferent sensory input. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS The CLARITY technique lets us observe in a transparent way the entire processed tissue compared with classical histological methods. CLARITY is a potentially useful tool to describe neuroanatomical structures and their neurochemical stratus.
Collapse
Affiliation(s)
- Guadalupe Martínez-Lorenzana
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Mohammed Gamal-Eltrabily
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Irma Alejandra Tello-García
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Ataulfo Martínez-Torres
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Marymar Becerra-González
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Abimael González-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Miguel Condés-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico.
| |
Collapse
|
34
|
Agostinelli LJ, Bassuk AG. Novel inhibitory brainstem neurons with selective projections to spinal lamina I reduce both pain and itch. J Comp Neurol 2020; 529:2125-2137. [PMID: 33247430 PMCID: PMC8009815 DOI: 10.1002/cne.25076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/18/2022]
Abstract
Sensory information is transmitted from peripheral nerves, through the spinal cord, and up to the brain (“bottom up” pathway). Some of this information may be modulated by “top‐down” projections from the brain to the spinal cord. Discovering endogenous mechanisms for reducing pain and itch holds enormous potential for developing new treatments. However, neurons mediating the top‐down inhibition of pain are not well understood, nor has any such pathway been identified for itch sensation. Here, we identify a novel population of GABAergic neurons in the ventral brainstem, distinguished by prodynorphin expression, which we named LJA5. LJA5 neurons provide the only known inhibitory projection specifically to lamina I of the spinal cord, which contains sensory neurons that transmit pain and itch information up to the brain. Chemogenetically activating LJA5 neurons in male mice reduces capsaicin‐induced pain and histamine‐induced itch. Identifying this new pathway opens new treatment opportunities for chronic, refractory pain, and pruritis.
Collapse
Affiliation(s)
- Lindsay J Agostinelli
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Alexander G Bassuk
- Department of Neurology, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA.,Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
35
|
Koba S, Hanai E, Kumada N, Watanabe T. Sympathoexcitatory input from hypothalamic paraventricular nucleus neurons projecting to rostral ventrolateral medulla is enhanced after myocardial infarction. Am J Physiol Heart Circ Physiol 2020; 319:H1197-H1207. [PMID: 32946261 DOI: 10.1152/ajpheart.00273.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elevated sympathetic vasomotor tone seen in heart failure (HF) may involve dysfunction of the hypothalamic paraventricular nucleus neurons that project to the rostral ventrolateral medulla (PVN-RVLM neurons). This study aimed to elucidate the role of PVN-RVLM neurons in the maintenance of resting renal sympathetic nerve activity (RSNA) after myocardial infarction (MI). In male rats, the left coronary artery was chronically ligated to induce MI. The rats received PVN microinjections of an adeno-associated viral (AAV) vector encoding archaerhodopsin T (ArchT) with the reporter yellow fluorescence protein (eYFP). The ArchT rats had abundant distributions of eYFP-labeled, PVN-derived axons in the RVLM. In anesthetized ArchT rats with MI (n = 12), optogenetic inhibition of the PVN-RVLM pathway achieved by 532-nm-wavelength laser illumination to the RVLM significantly decreased RSNA. This effect was not found in sham-operated ArchT rats (n = 6). Other rat groups received RVLM microinjections of a retrograde AAV vector encoding the red light-drivable halorhodopsin Jaws (Jaws) with the reporter green fluorescence protein (GFP) and showed expression of GFP-labeled cell bodies and dendrites in the PVN. Laser illumination of the PVN at a 635 nm wavelength elicited significant renal sympathoinhibition in Jaws rats with MI (n = 9) but not in sham-operated Jaws rats (n = 8). These results indicate that sympathoexcitatory input from PVN-RVLM neurons is enhanced after MI, suggesting that this monosynaptic pathway is part of the central nervous system circuitry that plays a critical role in generating an elevated sympathetic vasomotor tone commonly seen with HF.NEW & NOTEWORTHY Using optogenetics in rats, we report that sympathoexcitatory input from hypothalamic paraventricular nucleus neurons that project to the rostral ventrolateral medulla is enhanced after myocardial infarction. It is suggested that this monosynaptic pathway makes up a key part of central nervous system circuitry underlying sympathetic hyperactivation commonly seen in heart failure.
Collapse
Affiliation(s)
- Satoshi Koba
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Eri Hanai
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Nao Kumada
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| |
Collapse
|
36
|
Barretto-de-Souza L, Benini R, Reis-Silva LL, Crestani CC. Corticotropin-releasing factor neurotransmission in the lateral hypothalamus modulates the tachycardiac response during acute emotional stress in rats. Brain Res Bull 2020; 166:102-109. [PMID: 33227387 DOI: 10.1016/j.brainresbull.2020.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/25/2020] [Accepted: 11/14/2020] [Indexed: 12/29/2022]
Abstract
The lateral hypothalamus (LH) is implicated in the physiological and behavioral responses during stressful events. However, the local neurochemical mechanisms related to control of stress responses by this hypothalamic area are not completely understood. Therefore, in this study we evaluated the involvement of CRFergic neurotransmission acting through the CRF1 receptor within the LH in cardiovascular responses evoked by an acute session of restraint stress in rats. For this, we investigated the effect of bilateral microinjection of different doses (0.01, 0.1 and 1 nmol/100 nL) of the selective CRF1 receptor antagonist CP376395 into the LH on arterial pressure and heart rate increases and decrease in tail skin temperature evoked by acute restraint stress. We found that all doses of the CRF1 receptor antagonist microinjected into the LH decreased the restraint-evoked tachycardia, but without affecting the arterial pressure and tail skin temperature responses. Additionally, treatment of the LH with CP376395 at the doses of 0.1 and 1 nmol/100 nL increased the basal values of both heart rate and arterial pressure, whereas the dose of 0.1 nmol/100 nL decreased the skin temperature. Taken together, these findings indicate that CRFergic neurotransmission in the LH, acting through activation of local CRF1 receptors, plays a facilitatory role in the tachycardia observed during aversive threats, but without affecting the pressor and tail skin temperature responses. Our results also provide evidence that LH CRFergic neurotransmission in involved in tonic maintenance of cardiovascular function.
Collapse
Affiliation(s)
- Lucas Barretto-de-Souza
- Laboratory of Pharmacology, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Ricardo Benini
- Laboratory of Pharmacology, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Lilian L Reis-Silva
- Laboratory of Pharmacology, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
37
|
Baker J, Kimpinski K. Evidence of Impaired Cerebellar Connectivity at Rest and During Autonomic Maneuvers in Patients with Autonomic Failure. THE CEREBELLUM 2020; 19:30-39. [PMID: 31529276 DOI: 10.1007/s12311-019-01076-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The objective of the current study was to investigate whether patients with neurogenic orthostatic hypotension (NOH) secondary to autonomic failure have impaired functional connectivity between the cerebellum and central autonomic structures during autonomic challenges. Fifteen healthy controls (61 ± 14 years) and 15 NOH patients (67 ± 6 years; p = 0.12) completed the following tasks during a functional brain MRI: (1) 5 min of rest, (2) 5 min of lower-body negative pressure (LBNP) performed at - 35 mmHg, and (3) Three, 15-s Valsalva maneuvers (VM) at 40 mmHg. Functional connectivity (Conn Toolbox V18) between central autonomic structures and discrete cerebellar regions involved in cardiovascular autonomic control, including the vermis and posterior cerebellum, was assessed using a regions-of-interest approach during rest, LBNP and VM. Functional connectivity was contrasted between controls and patients with autonomic failure. At rest, controls had significantly more intra-cerebellar connectivity and more connectivity between cerebellar lobule 9 and key central autonomic structures, including: bilateral anterior insula (TR-value: 4.84; TL-value: 4.51), anterior cingulate cortex (T-value: 3.41) and bilateral thalamus (TR-value: 3.95; TL-value: 4.51). During autonomic maneuvers, controls showed significantly more connectivity between cardiovascular cerebellar regions (lobule 9 and anterior vermis) and important autonomic regulatory sites, including the brainstem, hippocampus and cingulate: vermis-brainstem (T-value: 4.31), lobule 9-brainstem (TR-value, 5.29; TL-value, 4.53), vermis-hippocampus (T-value, 4.63), and vermis-cingulate (T-value, 4.18). Anatomical and functional studies in animals and humans substantiate a significant role for the cerebellum in cardiovascular autonomic control during postural adjustments. In the current study, patients with NOH related to autonomic failure showed evidence of reduced connectivity between cardiovascular cerebellar regions and several important central autonomic structures, including the brainstem. The cerebellum is an established structure in cardiovascular autonomic control; therefore, evidence of impaired cerebellar connectivity to other autonomic structures may further contribute to the inability to properly regulate blood pressure during postural changes in NOH patients.
Collapse
Affiliation(s)
- Jacquie Baker
- School of Kinesiology, Western University, London, Ontario, Canada. .,Department of Clinical Neurological Sciences, University Hospital, London Health Sciences Centre, Rm. B7-140, 339 Windermere Road, London, Ontario, N6A 5A5, Canada.
| | - Kurt Kimpinski
- School of Kinesiology, Western University, London, Ontario, Canada.,Department of Clinical Neurological Sciences, University Hospital, London Health Sciences Centre, Rm. B7-140, 339 Windermere Road, London, Ontario, N6A 5A5, Canada.,Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
38
|
Nowacki A, Schober M, Nader L, Saryyeva A, Nguyen TK, Green AL, Pollo C, Krauss JK, Fontaine D, Aziz TZ. Deep Brain Stimulation for Chronic Cluster Headache: Meta‐Analysis of Individual Patient Data. Ann Neurol 2020; 88:956-969. [DOI: 10.1002/ana.25887] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Andreas Nowacki
- Department of Neurosurgery, Inselspital Bern University Hospital, University Bern Bern Switzerland
| | - Martin Schober
- Department of Neurosurgery, Inselspital Bern University Hospital, University Bern Bern Switzerland
| | - Lydia Nader
- Thuy Hospital Universitario Central de Asturias Oviedo Spain
| | - Assel Saryyeva
- Department of Neurosurgery Medical School Hannover Hannover Germany
| | - Thuy‐Anh Khoa Nguyen
- Department of Neurosurgery, Inselspital Bern University Hospital, University Bern Bern Switzerland
- ARTORG Center for Biomedical Engineering Research University of Bern Bern Switzerland
| | - Alexander L. Green
- Nuffield Department of Clinical Neuroscience University of Oxford Oxford UK
| | - Claudio Pollo
- Department of Neurosurgery, Inselspital Bern University Hospital, University Bern Bern Switzerland
| | | | - Denys Fontaine
- Department of Neurosurgery, Centre Hospitalier Universitaire de Nice, FHU INOVPAIN University Cote d'Azur Nice France
| | - Tipu Z. Aziz
- Nuffield Department of Clinical Neuroscience University of Oxford Oxford UK
| |
Collapse
|
39
|
Markovich-Molochnikov I, Cohen D. Bilateral responses of rat ventral striatum tonically active neurons to unilateral medial forebrain bundle stimulation. Eur J Neurosci 2020; 52:4499-4516. [PMID: 32810912 DOI: 10.1111/ejn.14939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 11/29/2022]
Abstract
Unilateral medial forebrain bundle (MFB) stimulation is an extremely effective promoter of reinforcement learning irrespective of the conditioned cue's laterality. The effectiveness of unilateral MFB stimulation, which activates the mesolimbic pathway connecting the ventral tegmental area to the ventral striatum (vStr), is surprising considering that these fibers rarely cross to the contralateral hemisphere. Specifically, this type of biased fiber distribution entails the activation of brain structures that are primarily ipsilateral to the stimulated MFB, along with weak to negligible activation of the contralateral structures, thus impeding the formation of a cue-outcome association. To better understand the spread of activation of MFB stimulation across hemispheres, we studied whether unilateral MFB stimulation primarily activates the ipsilateral vStr or the vStr of both hemispheres. We simultaneously recorded neuronal activity in the vStr of both hemispheres in response to several sets of unilateral MFB stimulation in anesthetized and freely moving rats. Unilateral MFB stimulation evoked strong stimulus-dependent activation of vStr tonically active neurons (TANs), presumably the cholinergic interneurons, in both hemispheres. However, the TANs' activation patterns and responsiveness depended on whether the stimulus was delivered ipsilaterally or contralaterally to the recorded neuron. These findings indicate that unilateral MFB stimulation effectively activates the vStr in both hemispheres in a stimulus-dependent manner which may serve as neuronal substrate for the formation of cue-outcome associations during reinforcement learning.
Collapse
Affiliation(s)
| | - Dana Cohen
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
40
|
Gasparini S, Howland JM, Thatcher AJ, Geerling JC. Central afferents to the nucleus of the solitary tract in rats and mice. J Comp Neurol 2020; 528:2708-2728. [PMID: 32307700 DOI: 10.1002/cne.24927] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022]
Abstract
The nucleus of the solitary tract (NTS) regulates life-sustaining functions ranging from appetite and digestion to heart rate and breathing. It is also the brain's primary sensory nucleus for visceral sensations relevant to symptoms in medical and psychiatric disorders. To better understand which neurons may exert top-down control over the NTS, here we provide a brain-wide map of all neurons that project axons directly to the caudal, viscerosensory NTS, focusing on a medial subregion with aldosterone-sensitive HSD2 neurons. Injecting an axonal tracer (cholera toxin b) into the NTS produces a similar pattern of retrograde labeling in rats and mice. The paraventricular hypothalamic nucleus (PVH), lateral hypothalamic area, and central nucleus of the amygdala (CeA) contain the densest concentrations of NTS-projecting neurons. PVH afferents are glutamatergic (express Slc17a6/Vglut2) and are distinct from neuroendocrine PVH neurons. CeA afferents are GABAergic (express Slc32a1/Vgat) and are distributed largely in the medial CeA subdivision. Other retrogradely labeled neurons are located in a variety of brain regions, including the cerebral cortex (insular and infralimbic areas), bed nucleus of the stria terminalis, periaqueductal gray, Barrington's nucleus, Kölliker-Fuse nucleus, hindbrain reticular formation, and rostral NTS. Similar patterns of retrograde labeling result from tracer injections into different NTS subdivisions, with dual retrograde tracing revealing that many afferent neurons project axon collaterals to both the lateral and medial NTS subdivisions. This information provides a roadmap for studying descending axonal projections that may influence visceromotor systems and visceral "mind-body" symptoms.
Collapse
Affiliation(s)
- Silvia Gasparini
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Jacob M Howland
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Andrew J Thatcher
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| | - Joel C Geerling
- Department of Neurology, Iowa Neuroscience Institute, University of Iowa, Iowa city, Iowa, USA
| |
Collapse
|
41
|
Cowen N, Bhatnagar A. The Potential Role of Activating the ATP-Sensitive Potassium Channel in the Treatment of Hyperphagic Obesity. Genes (Basel) 2020; 11:genes11040450. [PMID: 32326226 PMCID: PMC7230375 DOI: 10.3390/genes11040450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
To evaluate the potential role of ATP-sensitive potassium (KATP) channel activation in the treatment of hyperphagic obesity, a PubMed search was conducted focused on the expression of genes encoding the KATP channel, the response to activating the KATP channel in tissues regulating appetite and the establishment and maintenance of obesity, the evaluation of KATP activators in obese hyperphagic animal models, and clinical studies on syndromic obesity. KATP channel activation is mechanistically involved in the regulation of appetite in the arcuate nucleus; the regulation of hyperinsulinemia, glycemic control, appetite and satiety in the dorsal motor nucleus of vagus; insulin secretion by β-cells; and the synthesis and β-oxidation of fatty acids in adipocytes. KATP channel activators have been evaluated in hyperphagic obese animal models and were shown to reduce hyperphagia, induce fat loss and weight loss in older animals, reduce the accumulation of excess body fat in growing animals, reduce circulating and hepatic lipids, and improve glycemic control. Recent experience with a KATP channel activator in Prader-Willi syndrome is consistent with the therapeutic responses observed in animal models. KATP channel activation, given the breadth of impact and animal model and clinical results, is a viable target in hyperphagic obesity.
Collapse
|
42
|
Schaeuble D, Packard AEB, McKlveen JM, Morano R, Fourman S, Smith BL, Scheimann JR, Packard BA, Wilson SP, James J, Hui DY, Ulrich‐Lai YM, Herman JP, Myers B. Prefrontal Cortex Regulates Chronic Stress-Induced Cardiovascular Susceptibility. J Am Heart Assoc 2019; 8:e014451. [PMID: 31838941 PMCID: PMC6951062 DOI: 10.1161/jaha.119.014451] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Background The medial prefrontal cortex is necessary for appropriate appraisal of stressful information, as well as coordinating visceral and behavioral processes. However, prolonged stress impairs medial prefrontal cortex function and prefrontal-dependent behaviors. Additionally, chronic stress induces sympathetic predominance, contributing to health detriments associated with autonomic imbalance. Previous studies identified a subregion of rodent prefrontal cortex, infralimbic cortex (IL), as a key regulator of neuroendocrine-autonomic integration after chronic stress, suggesting that IL output may prevent chronic stress-induced autonomic imbalance. In the current study, we tested the hypothesis that the IL regulates hemodynamic, vascular, and cardiac responses to chronic stress. Methods and Results A viral-packaged small interfering RNA construct was used to knockdown vesicular glutamate transporter 1 (vGluT1) and reduce glutamate packaging and release from IL projection neurons. Male rats were injected with a vGluT1 small interfering RNA-expressing construct or GFP (green fluorescent protein) control into the IL and then remained as unstressed controls or were exposed to chronic variable stress. IL vGluT1 knockdown increased heart rate and mean arterial pressure reactivity, while chronic variable stress increased chronic mean arterial pressure only in small interfering RNA-treated rats. In another cohort, chronic variable stress and vGluT1 knockdown interacted to impair both endothelial-dependent and endothelial-independent vasoreactivity ex vivo. Furthermore, vGluT1 knockdown and chronic variable stress increased histological markers of fibrosis and hypertrophy. Conclusions Knockdown of glutamate release from IL projection neurons indicates that these cells are necessary to prevent the enhanced physiological responses to stress that promote susceptibility to cardiovascular pathophysiology. Ultimately, these findings provide evidence for a neurobiological mechanism mediating the relationship between stress and poor cardiovascular health outcomes.
Collapse
Affiliation(s)
| | | | - Jessica M. McKlveen
- National Institutes of HealthNational Center for Complimentary and Integrative HealthBethesdaMD
| | - Rachel Morano
- Pharmacology and Systems PhysiologyUniversity of CincinnatiOH
| | - Sarah Fourman
- Pathology and Laboratory MedicineUniversity of CincinnatiOH
| | | | | | | | - Steven P. Wilson
- Pharmacology, Physiology, and NeuroscienceUniversity of South CarolinaColumbiaSC
| | - Jeanne James
- Division of CardiologyDepartment of PediatricsMedical College of WisconsinMilwaukeeWI
| | - David Y. Hui
- Pathology and Laboratory MedicineUniversity of CincinnatiOH
| | | | - James P. Herman
- Pharmacology and Systems PhysiologyUniversity of CincinnatiOH
| | - Brent Myers
- Biomedical SciencesColorado State UniversityFort CollinsCO
| |
Collapse
|
43
|
Effects of mu opioid receptors in paraventricular nucleus on ejaculation through mediating sympathetic nerve system activity. Neuropharmacology 2019; 158:107709. [DOI: 10.1016/j.neuropharm.2019.107709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/07/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
|
44
|
Crevier-Sorbo G, Atkinson J, Di Genova T, Puligandla P, Dudley RWR. Hydrocephalus-induced neurogenic stunned myocardium and cardiac arrest in a child: completely reversed with CSF diversion. J Neurosurg Pediatr 2019; 24:35-40. [PMID: 31003226 DOI: 10.3171/2019.2.peds18711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 02/28/2019] [Indexed: 01/11/2023]
Abstract
Neurogenic stunned myocardium (NSM) is a potentially fatal cause of sudden cardiogenic dysfunction due to an acute neurological event, most commonly aneurysmal subarachnoid hemorrhage in adults. Only two pediatric cases of hydrocephalus-induced NSM have been reported. Here the authors report a third case in a 14-year-old boy who presented with severe headache, decreased level of consciousness, and shock in the context of acute hydrocephalus secondary to fourth ventricular outlet obstruction 3 years after standard-risk medulloblastoma treatment. He was initially stabilized with the insertion of an external ventricular drain and vasopressor treatment. He had a profoundly reduced cardiac contractility and became asystolic for 1 minute, requiring cardiopulmonary resuscitation when vasopressors were inadvertently discontinued. Over 1 week, his ventricles decreased in size and his cardiac function returned to normal. All other causes of heart failure were ruled out, and his impressive response to CSF diversion clarified the diagnosis of NSM secondary to hydrocephalus. He was unable to be weaned from his drain during his time in the hospital, so he underwent an endoscopic third ventriculostomy and has remained well with normal cardiac function at more than 6 months' follow-up. This case highlights the importance of prompt CSF diversion and cardiac support for acute hydrocephalus presenting with heart failure in the pediatric population.
Collapse
|
45
|
Gomes-de-Souza L, Benini R, Costa-Ferreira W, Crestani CC. GABA A but not GABA B receptors in the lateral hypothalamus modulate the tachycardic response to emotional stress in rats. Eur Neuropsychopharmacol 2019; 29:672-680. [PMID: 30878320 DOI: 10.1016/j.euroneuro.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/05/2019] [Accepted: 03/02/2019] [Indexed: 01/26/2023]
Abstract
The lateral hypothalamus (LH) has been described as one of the hypothalamic areas involved in the behavioral and physiological responses triggered by aversive stimuli. Previous studies indicated involvement of the LH in cardiovascular responses to stress. Despite this evidence, the local neurochemical mechanisms involved in LH control of stress responses is still poorly understood. Therefore, in the present study, we investigated the role of GABAergic neurotransmission within the LH in cardiovascular responses induced by an acute session of restraint stress in rats. For this, we evaluated the effect of bilateral microinjection of selective antagonists of either GABAA or GABAB receptors into the LH on arterial pressure increase, heart rate (HR) increase and reduction in tail skin temperature induced by restraint stress. We found that microinjection of the selective GABAA receptor antagonist SR95531 into the LH decreased the increase in HR caused by restraint stress, but without affecting the increase in arterial pressure increase or the reduction in tail skin temperature. Conversely, LH treatment with the selective GABAB receptor antagonist CGP35348 did not affect the restraint-evoked cardiovascular changes. These findings indicate that GABAergic neurotransmission in the LH, acting through activation of local GABAA receptors, plays a facilitatory role in the tachycardic response observed during aversive threats.
Collapse
Affiliation(s)
- Lucas Gomes-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Ricardo Benini
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Willian Costa-Ferreira
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jau Km 01 (Campus Universitário), Campus Ville, 14800-903 Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
46
|
The location and characteristics of the thermal sudomotor pathways in the human brainstem: A reappraisal. Auton Neurosci 2019; 217:80-90. [PMID: 30744906 DOI: 10.1016/j.autneu.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 11/21/2018] [Accepted: 01/27/2019] [Indexed: 11/21/2022]
Abstract
To elucidate location and characteristics of the central thermoregulatory sudomotor pathway in the human brainstem, thermoregulatory sweating (TS) in 91 patients with focal brainstem lesions was studied. TS was symmetric or minimally asymmetric in 40 subjects (Group S), and was apparently asymmetric in 51 patients (Group AS). In Group AS, the main abnormality was ipsilateral segmental hypohidrosis with a varying extent, involving predominantly the upper half of the body. Lesion locations, correlations between thermoregulatory sweat test results, and other autonomic and somatic functions were compared between the groups. The results suggested following: (1) The hypothalamospinal pathway related to TS may pass through the posterior hypothalamus and descend in the dorsolateral part of the brainstem, near the spinal trigeminal and spinothalamic tracts; (2) the pathway may descend together with those related to oculosympathetic and vasoconstrictor systems, but each of these may form distinct fiber groups; (3) the majority of the central TS fibers may reach ipsilateral sudomotor sympathetic neurons of the spinal cord, even though some fibers may cross at various levels; (4) in this descending pathway, somatotopic arrangements corresponding to each of the spinal sympathetic segments must be present; (5) There may be another fiber group passing through the central to dorsal paramedian portions of the brainstem, and lesions of these fibers also result in asymmetric TS, but seldom in oculosympathetic dysfunction. This second pathway probably exerts contralateral inhibitory influence on TS, but its origin, intracerebral course and exact physiological function require further clinical investigations.
Collapse
|
47
|
Impaired Hypothalamic Regulation of Sympathetic Outflow in Primary Hypertension. Neurosci Bull 2018; 35:124-132. [PMID: 30506315 DOI: 10.1007/s12264-018-0316-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/01/2018] [Indexed: 01/01/2023] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is a crucial region involved in maintaining homeostasis through the regulation of cardiovascular, neuroendocrine, and other functions. The PVN provides a dominant source of excitatory drive to the sympathetic outflow through innervation of the brainstem and spinal cord in hypertension. We discuss current findings on the role of the PVN in the regulation of sympathetic output in both normotensive and hypertensive conditions. The PVN seems to play a major role in generating the elevated sympathetic vasomotor activity that is characteristic of multiple forms of hypertension, including primary hypertension in humans. Recent studies in the spontaneously hypertensive rat model have revealed an imbalance of inhibitory and excitatory synaptic inputs to PVN pre-sympathetic neurons as indicated by impaired inhibitory and enhanced excitatory synaptic inputs in hypertension. This imbalance of inhibitory and excitatory synaptic inputs in the PVN forms the basis for elevated sympathetic outflow in hypertension. In this review, we discuss the disruption of balance between glutamatergic and GABAergic inputs and the associated cellular and molecular alterations as mechanisms underlying the hyperactivity of PVN pre-sympathetic neurons in hypertension.
Collapse
|
48
|
Dampney RA, Michelini LC, Li DP, Pan HL. Regulation of sympathetic vasomotor activity by the hypothalamic paraventricular nucleus in normotensive and hypertensive states. Am J Physiol Heart Circ Physiol 2018; 315:H1200-H1214. [PMID: 30095973 PMCID: PMC6297824 DOI: 10.1152/ajpheart.00216.2018] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 12/22/2022]
Abstract
The hypothalamic paraventricular nucleus (PVN) is a unique and important brain region involved in the control of cardiovascular, neuroendocrine, and other physiological functions pertinent to homeostasis. The PVN is a major source of excitatory drive to the spinal sympathetic outflow via both direct and indirect projections. In this review, we discuss the role of the PVN in the regulation of sympathetic output in normal physiological conditions and in hypertension. In normal healthy animals, the PVN presympathetic neurons do not appear to have a major role in sustaining resting sympathetic vasomotor activity or in regulating sympathetic responses to short-term homeostatic challenges such as acute hypotension or hypoxia. Their role is, however, much more significant during longer-term challenges, such as sustained water deprivation, chronic intermittent hypoxia, and pregnancy. The PVN also appears to have a major role in generating the increased sympathetic vasomotor activity that is characteristic of multiple forms of hypertension. Recent studies in the spontaneously hypertensive rat model have shown that impaired inhibitory and enhanced excitatory synaptic inputs to PVN presympathetic neurons are the basis for the heightened sympathetic outflow in hypertension. We discuss the molecular mechanisms underlying the presynaptic and postsynaptic alterations in GABAergic and glutamatergic inputs to PVN presympathetic neurons in hypertension. In addition, we discuss the ability of exercise training to correct sympathetic hyperactivity by restoring blood-brain barrier integrity, reducing angiotensin II availability, and decreasing oxidative stress and inflammation in the PVN.
Collapse
Affiliation(s)
- Roger A Dampney
- Department of Physiology, University of Sydney , Sydney, New South Wales , Australia
| | - Lisete C Michelini
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - De-Pei Li
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center , Houston, Texas
| | - Hui-Lin Pan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center , Houston, Texas
| |
Collapse
|
49
|
Sturm VE, Brown JA, Hua AY, Lwi SJ, Zhou J, Kurth F, Eickhoff SB, Rosen HJ, Kramer JH, Miller BL, Levenson RW, Seeley WW. Network Architecture Underlying Basal Autonomic Outflow: Evidence from Frontotemporal Dementia. J Neurosci 2018; 38:8943-8955. [PMID: 30181137 PMCID: PMC6191520 DOI: 10.1523/jneurosci.0347-18.2018] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/23/2018] [Accepted: 08/27/2018] [Indexed: 12/22/2022] Open
Abstract
The salience network is a distributed neural system that maintains homeostasis by regulating autonomic nervous system activity and social-emotional function. Here we examined how within-network connectivity relates to individual differences in human (including males and females) baseline parasympathetic and sympathetic nervous activity. We measured resting autonomic nervous system physiology in 24 healthy controls and 23 patients with behavioral variant frontotemporal dementia (bvFTD), a neurodegenerative disease characterized by baseline autonomic deficits. Participants also underwent structural and task-free fMRI. First, we used voxel-based morphometry to determine whether salience network atrophy was associated with lower baseline respiratory sinus arrhythmia (a parasympathetic measure) and skin conductance level (a sympathetic measure) in bvFTD. Next, we examined whether functional connectivity deficits in 21 autonomic-relevant, salience network node-pairs related to baseline autonomic dysfunction. Lower baseline respiratory sinus arrhythmia was associated with smaller volume in left ventral anterior insula (vAI), weaker connectivity between bilateral vAI and bilateral anterior cingulate cortex (ACC), and stronger connectivity between bilateral ACC and bilateral hypothalamus/amygdala. Lower baseline skin conductance level, in contrast, was associated with smaller volume in inferior temporal gyrus, dorsal mid-insula, and hypothalamus; weaker connectivity between bilateral ACC and right hypothalamus/amygdala; and stronger connectivity between bilateral dorsal anterior insula and periaqueductal gray. Our results suggest that baseline parasympathetic and sympathetic tone depends on the integrity of lateralized salience network hubs (left vAI for parasympathetic and right hypothalamus/amygdala for sympathetic) and highly calibrated ipsilateral and contralateral network connections. In bvFTD, deficits in this system may underlie resting parasympathetic and sympathetic disruption.SIGNIFICANCE STATEMENT The salience network maintains homeostasis and regulates autonomic nervous system activity. Whether within-network connectivity patterns underlie individual differences in resting parasympathetic and sympathetic nervous system activity, however, is not well understood. We measured baseline autonomic nervous system activity in healthy controls and patients with behavioral variant frontotemporal dementia, a neurodegenerative disease characterized by resting autonomic deficits, and probed how salience network dysfunction relates to diminished parasympathetic and sympathetic outflow. Our results indicate that baseline parasympathetic and sympathetic tone are the product of complex, opposing intranetwork nodal interactions and depend on the integrity of highly tuned, lateralized salience network hubs (i.e., left ventral anterior insula for parasympathetic activity and right hypothalamus/amygdala for sympathetic activity).
Collapse
Affiliation(s)
- Virginia E Sturm
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158
| | - Jesse A Brown
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158
| | - Alice Y Hua
- Department of Psychology, University of California, Berkeley, California 94720-1650
| | - Sandy J Lwi
- Department of Psychology, University of California, Berkeley, California 94720-1650
| | - Juan Zhou
- Center for Cognitive Neuroscience, Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857
| | - Florian Kurth
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, University of California-Los Angeles School of Medicine, Los Angeles, California 90095
| | - Simon B Eickhoff
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine Universität, 40225 Düsseldorf, Germany
- Institute of Neuroscience and Medicine, Brain and Behaviour, Research Centre Jülich, Jülich, 52425, Germany, and
| | - Howard J Rosen
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158
| | - Joel H Kramer
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158
| | - Bruce L Miller
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158
| | - Robert W Levenson
- Department of Psychology, University of California, Berkeley, California 94720-1650
| | - William W Seeley
- Department of Neurology, University of California-San Francisco, Sandler Neurosciences Center, San Francisco, California 94158,
- Department of Pathology, University of California, San Francisco, California 94143
| |
Collapse
|
50
|
Koba S, Hanai E, Kumada N, Kataoka N, Nakamura K, Watanabe T. Sympathoexcitation by hypothalamic paraventricular nucleus neurons projecting to the rostral ventrolateral medulla. J Physiol 2018; 596:4581-4595. [PMID: 30019338 DOI: 10.1113/jp276223] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023] Open
Abstract
KEY POINTS Causal relationships between central cardiovascular pathways and sympathetic vasomotor tone have not been evidenced. This study aimed to verify the sympathoexcitatory role of hypothalamic paraventricular nucleus neurons that project to the rostral ventrolateral medulla (PVN-RVLM neurons). By using optogenetic techniques, we demonstrated that stimulation of PVN-RVLM glutamatergic neurons increased renal sympathetic nerve activity and arterial pressure via, at least in part, stimulation of RVLM C1 neurons in rats. This monosynaptic pathway may function in acute sympathetic adjustments to stressors and/or be a component of chronic sympathetic hyperactivity in pathological conditions such as heart failure. ABSTRACT The rostral ventrolateral medulla (RVLM), which is known to play an important role in regulating sympathetic vasomotor tone, receives axonal projections from the hypothalamic paraventricular nucleus (PVN). However, no studies have proved that excitation of the PVN neurons that send axonal projections to the RVLM (PVN-RVLM neurons) causes sympathoexcitation. This study aimed to directly examine the sympathoexcitatory role of PVN-RVLM neurons. Male rats received microinjections into the PVN with an adeno-associated virus (AAV) vector that encoded a hybrid of channelrhodopsin-2/1 with the reporter tdTomato (ChIEF-tdTomato), or into the RVLM with a retrograde AAV vector that encoded a channelrhodopsin with green fluorescent protein (ChR2-GFPretro ). Under anaesthesia with urethane and α-chloralose, photostimulation (473 nm wavelength) of PVN-RVLM neurons, achieved by laser illumination of either RVLM of ChIEF-tdTomato rats (n = 8) or PVN of ChR2-GFPretro rats (n = 4), elicited significant renal sympathoexcitation. Immunofluorescence confocal microscopy showed that RVLM adrenergic C1 neurons of ChIEF-tdTomato rats were closely associated with tdTomato-labelled, PVN-derived axons that contained vesicular glutamate transporter 2. In another subset of anaesthetized ChIEF-tdTomato rats (n = 6), the renal sympathoexcitation elicited by photostimulation of the PVN was suppressed by administering ionotropic glutamate receptor blockers into the RVLM. These results demonstrate that excitation of PVN-RVLM glutamatergic neurons leads to sympathoexcitation via, at least in part, stimulation of RVLM C1 neurons.
Collapse
Affiliation(s)
- Satoshi Koba
- Division of Integrative Physiology, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Eri Hanai
- Division of Integrative Physiology, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Nao Kumada
- Division of Integrative Physiology, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| | - Naoya Kataoka
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.,PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Tatsuo Watanabe
- Division of Integrative Physiology, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori, 683-8503, Japan
| |
Collapse
|