1
|
Tsai LP, Luo DZ, Chan H, Hung WC, Lai WS, Min MY, Wong SB. Implication of locus coeruleus dysfunction in Prader-Willi syndrome: Insights from a mouse model. Exp Neurol 2024; 381:114927. [PMID: 39159912 DOI: 10.1016/j.expneurol.2024.114927] [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: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Prader-Willi syndrome (PWS) is a multisystemic disorder. Notably, many characteristic symptoms of PWS are correlated with locus coeruleus norepinephrine system (LC-NE) dysfunction, including impairment in arousal, learning, pain modulation, and stress-induced negative affective states. Although electrophysiological experiments in necdin-deficient mice, an established PWS animal model, have revealed decreased spontaneous neuronal firing activity in the LC and impaired excitability, the behavioral phenotypes related to LC-NE dysfunction remain unexplored. In this study, heterozygous necdin-deficient mice (B6.Cg-Ndntm1ky) were bred from wild-type (WT) females to generate WT (+m/+p) and heterozygous (+m/-p) animals. Compared to WT mice, Ndn + m/-p mice demonstrated impaired visual-spatial memory in the Y-maze test, reduced social interaction, impaired sexual recognition, and shorter falling latency on the Rotarod. Using the open field test (OFT) and elevated plus maze (EPM), we observed similar locomotion activity of Ndn + m/-p and WT mice, but Ndn + m/-p mice were less anxious. After acute restraint, Ndn + m/-p mice exhibited significant impairment in stress-induced anxiety. Additionally, the plasma norepinephrine surge following exposure to acute restraint stress was also impaired. Pretreatment with atomoxetine, a norepinephrine reuptake inhibitor aimed to enhance LC function, restored Ndn + m/-p mice to exhibit a normal response to acute restraint stress. Furthermore, by employing chemogenetic approaches to facilitate LC neuronal firing, post-stress anxious responses were also partially rescued in Ndn + m/-p mice. These data strongly suggest that LC dysfunction is implicated in the pathogenesis of stress-related neuropsychiatric symptoms in PWS. Manipulation of LC activity may hold therapeutic potential for patients with PWS.
Collapse
Affiliation(s)
- Li-Ping Tsai
- Department of Pediatrics, Taipei Tzu Chi Hospital, Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; School of Medicine, Tzu Chi University, Hualien 97071, Taiwan; Department of Pediatrics, Heping Fuyou Branch, Taipei City Hospital, Taipei 100027, Taiwan
| | - Da-Zhong Luo
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan
| | - Hao Chan
- Department of Pediatrics, Taipei Tzu Chi Hospital, Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Wei-Chen Hung
- Department of Pediatrics, Taipei Tzu Chi Hospital, Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, Taipei 10617, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Shi-Bing Wong
- Department of Pediatrics, Taipei Tzu Chi Hospital, Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; School of Medicine, Tzu Chi University, Hualien 97071, Taiwan.
| |
Collapse
|
2
|
Rosatti MR, Gargaglioni LH, Dias MB. Lateral hypothalamic astrocytes contribute to the hypercapnic chemoreflex in a light-dark cycle-dependent manner in unanesthetized rats. Respir Physiol Neurobiol 2024; 331:104352. [PMID: 39299615 DOI: 10.1016/j.resp.2024.104352] [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: 07/15/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Brainstem astrocytes are important for CO2/H+ chemoreception. Lateral Hypothalamus/Perifornicial Area (LH/PFA) neurons have an excitatory effect on the ventilatory response to CO2, however the role of the astrocytes is unknown. We hypothesized that LH/PFA astrocytes play an excitatory role in the hypercapnic ventilatory response in a sleep-wake and light-dark cycles-dependent manner. We manipulated the activity of astrocytes in the LH/PFA of male Wistar rats through microinjection of Fluorocitrate (Fct), which selectively affects astrocytes, inducing the exocytosis of gliotransmitters. We investigated the effects of intra-LH/PFA Fct microinjection on resting breathing and ventilatory responses to hypercapnia and hypoxia during wakefulness and NREM sleep, in the light and dark phases. Fct increased ventilation during hypercapnia but not during room air or hypoxia. The hypercapnic chemoreflex was increased exclusively during the dark-active phase during both, wakefulness and NREM sleep, indicating that LH/PFA astrocytes play an excitatory role in hypercapnic ventilatory response in a light-dark cycle-dependent manner.
Collapse
Affiliation(s)
- Marcelo Rafanelli Rosatti
- Department of Structural and Functional Biology, Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil. Botucatu, SP, Brazil.
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil.
| | - Mirela B Dias
- Department of Structural and Functional Biology, Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil. Botucatu, SP, Brazil.
| |
Collapse
|
3
|
Tao Y, Li X, Dong Q, Kong L, Petersen AJ, Yan Y, Xu K, Zima S, Li Y, Schmidt DK, Ayala M, Mathivanan S, Sousa AMM, Chang Q, Zhang SC. Generation of locus coeruleus norepinephrine neurons from human pluripotent stem cells. Nat Biotechnol 2024; 42:1404-1416. [PMID: 37974010 PMCID: PMC11392812 DOI: 10.1038/s41587-023-01977-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/30/2023] [Indexed: 11/19/2023]
Abstract
Central norepinephrine (NE) neurons, located mainly in the locus coeruleus (LC), are implicated in diverse psychiatric and neurodegenerative diseases and are an emerging target for drug discovery. To facilitate their study, we developed a method to generate 40-60% human LC-NE neurons from human pluripotent stem cells. The approach depends on our identification of ACTIVIN A in regulating LC-NE transcription factors in dorsal rhombomere 1 (r1) progenitors. In vitro generated human LC-NE neurons display extensive axonal arborization; release and uptake NE; and exhibit pacemaker activity, calcium oscillation and chemoreceptor activity in response to CO2. Single-nucleus RNA sequencing (snRNA-seq) analysis at multiple timepoints confirmed NE cell identity and revealed the differentiation trajectory from hindbrain progenitors to NE neurons via an ASCL1-expressing precursor stage. LC-NE neurons engineered with an NE sensor reliably reported extracellular levels of NE. The availability of functional human LC-NE neurons enables investigation of their roles in psychiatric and neurodegenerative diseases and provides a tool for therapeutics development.
Collapse
Affiliation(s)
- Yunlong Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| | - Xueyan Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiping Dong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Linghai Kong
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Yuanwei Yan
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ke Xu
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Seth Zima
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Yanru Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Melvin Ayala
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Andre M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Neuroscience, Department of Neurology, University of Wisconsin-Madison, Madison, WI, USA.
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
4
|
Ripamonte GC, Fonseca EM, Frias AT, Patrone LGA, Vilela-Costa HH, Silva KSC, Szawka RE, Bícego KC, Zangrossi H, Plummer NW, Jensen P, Gargaglioni LH. Locus coeruleus noradrenaline depletion and its differential impact on CO 2-induced panic and hyperventilation in male and female mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111063. [PMID: 38908504 PMCID: PMC11323958 DOI: 10.1016/j.pnpbp.2024.111063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
CO2 exposure has been used to investigate the panicogenic response in patients with panic disorder. These patients are more sensitive to CO2, and more likely to experience the "false suffocation alarm" which triggers panic attacks. Imbalances in locus coeruleus noradrenergic (LC-NA) neurotransmission are responsible for psychiatric disorders, including panic disorder. These neurons are sensitive to changes in CO2/pH. Therefore, we investigated if LC-NA neurons are differentially activated after severe hypercapnia in mice. Further, we evaluated the participation of LC-NA neurons in ventilatory and panic-like escape responses induced by 20% CO2 in male and female wild type mice and two mouse models of altered LC-NA synthesis. Hypercapnia activates the LC-NA neurons, with males presenting a heightened level of activation. Mutant males lacking or with reduced LC-NA synthesis showed hypoventilation, while animals lacking LC noradrenaline present an increased metabolic rate compared to wild type in normocapnia. When exposed to CO2, males lacking LC noradrenaline showed a lower respiratory frequency compared to control animals. On the other hand, females lacking LC noradrenaline presented a higher tidal volume. Nevertheless, no change in ventilation was observed in either sex. CO2 evoked an active escape response. Mice lacking LC noradrenaline had a blunted jumping response and an increased freezing duration compared to the other groups. They also presented fewer racing episodes compared to wild type animals, but not different from mice with reduced LC noradrenaline. These findings suggest that LC-NA has an important role in ventilatory and panic-like escape responses elicited by CO2 exposure in mice.
Collapse
Affiliation(s)
- Gabriel C Ripamonte
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, Jaboticabal, São Paulo, 14884-900, Brazil
| | - Elisa M Fonseca
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alana T Frias
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, Jaboticabal, São Paulo, 14884-900, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, Jaboticabal, São Paulo, 14884-900, Brazil
| | - Heloísa H Vilela-Costa
- Department of Biochemistry, Pharmacology and Physiology, Institute of Biological and Natural Sciences, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Kaoma S C Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais - UFMG, Belo Horizonte, MG, Brazil
| | - Raphael E Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais - UFMG, Belo Horizonte, MG, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, Jaboticabal, São Paulo, 14884-900, Brazil
| | - Hélio Zangrossi
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto CEP:14049-900, Brazil
| | - Nicholas W Plummer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, United States Department of Health and Human Services, Durham, NC, USA
| | - Patricia Jensen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, United States Department of Health and Human Services, Durham, NC, USA
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, Jaboticabal, São Paulo, 14884-900, Brazil.
| |
Collapse
|
5
|
Furdui A, da Silveira Scarpellini C, Montandon G. Anatomical distribution of µ-opioid receptors, neurokinin-1 receptors, and vesicular glutamate transporter 2 in the mouse brainstem respiratory network. J Neurophysiol 2024; 132:108-129. [PMID: 38748514 DOI: 10.1152/jn.00478.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 07/03/2024] Open
Abstract
µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.
Collapse
Affiliation(s)
- Andreea Furdui
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Gaspard Montandon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Ahlbrand R, Wilson A, Woller P, Sachdeva Y, Lai J, Davis N, Wiggins J, Sah R. Sex-specific threat responding and neuronal engagement in carbon dioxide associated fear and extinction: Noradrenergic involvement in female mice. Neurobiol Stress 2024; 30:100617. [PMID: 38433995 PMCID: PMC10907837 DOI: 10.1016/j.ynstr.2024.100617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Difficulty in appropriately responding to threats is a key feature of psychiatric disorders, especially fear-related conditions such as panic disorder (PD) and posttraumatic stress disorder (PTSD). Most prior work on threat and fear regulation involves exposure to external threatful cues. However, fear can also be triggered by aversive, within-the-body, sensations. This interoceptive signaling of fear is highly relevant to PD and PTSD but is not well understood, especially in the context of sex. Using female and male mice, the current study investigated fear-associated spontaneous and conditioned behaviors to carbon dioxide (CO2) inhalation, a potent interoceptive threat that induces fear and panic. We also investigated whether behavioral sensitivity to CO2 is associated with delayed PTSD-relevant behaviors. CO2 evoked heterogenous freezing behaviors in both male and female animals. However, active, rearing behavior was significantly reduced in CO2-exposed male but not female mice. Interestingly, behavioral sensitivity to CO2 was associated with compromised fear extinction, independent of sex. However, in comparison to CO2-exposed males, females elicited less freezing and higher rearing during extinction suggesting an engagement of active versus passive defensive coping. Persistent neuronal activation marker ΔFosB immuno-mapping revealed attenuated engagement of infralimbic-prefrontal areas in both sexes but higher activation of brain stem locus coeruleus (LC) area in females. Inter-regional co-activation mapping revealed sex-independent disruptions in the infralimbic-amygdala associations but altered LC associations only in CO2-exposed female mice. Lastly, dopamine β hydroxylase positive (DβH + ve) noradrenergic neuronal cell counts in the LC correlated with freezing and rearing behaviors during CO2 inhalation and extinction only in female but not male mice. Collectively, these data provide evidence for higher active defensive responding to interoceptive threat CO2-associated fear in females that may stem from increased recruitment of the brainstem noradrenergic system. Our findings reveal distinct contributory mechanisms that may promote sex differences in fear and panic associated pathologies.
Collapse
Affiliation(s)
- Rebecca Ahlbrand
- Department of Pharmacology and Systems Physiology, University of Cincinnati, USA
- Veterans Affairs Medical Center, Cincinnati, OH, USA
| | - Allison Wilson
- Neuroscience Undergraduate Program, University of Cincinnati, USA
| | - Patrick Woller
- Neuroscience Graduate Program, University of Cincinnati, USA
| | - Yuv Sachdeva
- Department of Pharmacology and Systems Physiology, University of Cincinnati, USA
| | - Jayden Lai
- Department of Pharmacology and Systems Physiology, University of Cincinnati, USA
| | - Nikki Davis
- Neuroscience Undergraduate Program, University of Cincinnati, USA
| | - James Wiggins
- Neuroscience Undergraduate Program, University of Cincinnati, USA
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati, USA
- Neuroscience Graduate Program, University of Cincinnati, USA
- Veterans Affairs Medical Center, Cincinnati, OH, USA
| |
Collapse
|
7
|
Janes TA, Cardani S, Saini JK, Pagliardini S. Etonogestrel promotes respiratory recovery in an in vivo rat model of central chemoreflex impairment. Acta Physiol (Oxf) 2024; 240:e14093. [PMID: 38258900 DOI: 10.1111/apha.14093] [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: 08/08/2023] [Revised: 10/11/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024]
Abstract
AIM The central CO2 chemoreflex is a vital component of respiratory control networks, providing excitatory drive during resting conditions and challenges to blood gas homeostasis. The retrotrapezoid nucleus is a crucial hub for CO2 chemosensitivity; its ablation or inhibition attenuates CO2 chemoreflexes and diminishes restful breathing. Similar phenotypes characterize certain hypoventilation syndromes, suggesting underlying retrotrapezoid nucleus impairment in these disorders. Progesterone stimulates restful breathing and CO2 chemoreflexes. However, its mechanisms and sites of actions remain unknown and the experimental use of synthetic progestins in patients and animal models have been met with mixed respiratory outcomes. METHODS We investigated whether acute or chronic administration of the progestinic drug, etonogestrel, could rescue respiratory chemoreflexes following selective lesion of the retrotrapezoid nucleus with saporin toxin. Adult female Sprague Dawley rats were grouped based on lesion size determined by the number of surviving chemosensitive neurons, and ventilatory responses were measured by whole body plethysmography. RESULTS Ventilatory responses to hypercapnia (but not hypoxia) were compromised in a lesion-dependent manner. Chronic etonogestrel treatment improved CO2 chemosensitivity selectively in rats with moderate lesion, suggesting that a residual number of chemosensitive neurons are required for etonogestrel-induced CO2 chemoreflex recovery. CONCLUSION This study provides new evidence for the use of progestins as respiratory stimulants under conditions of central hypoventilation and provides a new testable model for assessing the mechanism of action of progestins in the respiratory network.
Collapse
Affiliation(s)
- Tara A Janes
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Silvia Cardani
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jasmeen K Saini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Silvia Pagliardini
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Eugenín J, Beltrán-Castillo S, Irribarra E, Pulgar-Sepúlveda R, Abarca N, von Bernhardi R. Microglial reactivity in brainstem chemosensory nuclei in response to hypercapnia. Front Physiol 2024; 15:1332355. [PMID: 38476146 PMCID: PMC10927973 DOI: 10.3389/fphys.2024.1332355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Microglia, the resident immune cells of the CNS, surveil, detect, and respond to various extracellular signals. Depending on the nature of these signals, an integrative microglial response can be triggered, resulting in a phenotypic transformation. Here, we evaluate whether hypercapnia modifies microglia phenotype in brainstem respiratory-related nuclei. Adult C57BL/6 inbred mice were exposed to 10% CO2 enriched air (hypercapnia), or pure air (control), for 10 or 30 min and immediately processed for immunohistochemistry to detect the ubiquitous microglia marker, ionized calcium binding adaptor molecule 1 (Iba1). Hypercapnia for thirty, but not 10 min reduced the Iba1 labeling percent coverage in the ventral respiratory column (VRC), raphe nucleus (RN), and nucleus tractus solitarius (NTS) and the number of primary branches in VRC. The morphological changes persisted, at least, for 60 min breathing air after the hypercapnic challenge. No significant changes were observed in Iba1+ cells in the spinal trigeminal nucleus (Sp5) and the hippocampus. In CF-1 outbred mice, 10% CO2 followed by 60 min of breathing air, resulted in the reduction of Iba1 labeling percent coverage and the number and length of primary branches in VRC, RN, and NTS. No morphological change was observed in Iba1+ cells in Sp5 and hippocampus. Double immunofluorescence revealed that prolonged hypercapnia increased the expression of CD86, an inflammatory marker for reactive state microglia, in Iba1+ cells in VRC, RN, and NTS, but not in Sp5 and hippocampus in CF-1 mice. By contrast, the expression of CD206, a marker of regulatory state microglia, persisted unmodified. In brainstem, but not in hippocampal microglia cultures, hypercapnia increased the level of IL1β, but not that of TGFβ measured by ELISA. Our results show that microglia from respiratory-related chemosensory nuclei, are reactive to prolonged hypercapnia acquiring an inflammatory-like phenotype.
Collapse
Affiliation(s)
- Jaime Eugenín
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Sebastián Beltrán-Castillo
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Estefanía Irribarra
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | | | - Nicolás Abarca
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Rommy von Bernhardi
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
9
|
Novello M, Bosman LWJ, De Zeeuw CI. A Systematic Review of Direct Outputs from the Cerebellum to the Brainstem and Diencephalon in Mammals. CEREBELLUM (LONDON, ENGLAND) 2024; 23:210-239. [PMID: 36575348 PMCID: PMC10864519 DOI: 10.1007/s12311-022-01499-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
The cerebellum is involved in many motor, autonomic and cognitive functions, and new tasks that have a cerebellar contribution are discovered on a regular basis. Simultaneously, our insight into the functional compartmentalization of the cerebellum has markedly improved. Additionally, studies on cerebellar output pathways have seen a renaissance due to the development of viral tracing techniques. To create an overview of the current state of our understanding of cerebellar efferents, we undertook a systematic review of all studies on monosynaptic projections from the cerebellum to the brainstem and the diencephalon in mammals. This revealed that important projections from the cerebellum, to the motor nuclei, cerebral cortex, and basal ganglia, are predominantly di- or polysynaptic, rather than monosynaptic. Strikingly, most target areas receive cerebellar input from all three cerebellar nuclei, showing a convergence of cerebellar information at the output level. Overall, there appeared to be a large level of agreement between studies on different species as well as on the use of different types of neural tracers, making the emerging picture of the cerebellar output areas a solid one. Finally, we discuss how this cerebellar output network is affected by a range of diseases and syndromes, with also non-cerebellar diseases having impact on cerebellar output areas.
Collapse
Affiliation(s)
- Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands.
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Rossano F, Caiazza C, Zotti N, Viacava L, Irano A, Solini N, Pistone L, Pezone R, Cilmi F, Ricci C, De Prisco M, Iasevoli F, Kishi T, Solmi M, de Bartolomeis A, Fornaro M. The efficacy, safety, and adverse events of azapirones in anxiety disorders: A systematic review and meta-analysis of randomized controlled trials. Eur Neuropsychopharmacol 2023; 76:23-51. [PMID: 37544075 DOI: 10.1016/j.euroneuro.2023.07.008] [Citation(s) in RCA: 1] [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: 04/25/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 08/08/2023]
Abstract
Azapirones have been proposed as anxiety and mood modulators. We assessed azapirones' viability in anxiety disorders via systematic review and random-effects meta-analysis, inquiring PubMed/MEDLINE/CENTRAL/WHO-ICTRP/WebOfScience/VIP up-to 05/01/2023. We conducted sensitivity, and subgroup analyses assessing heterogeneity, publication bias, risk of bias, and confidence in the evidence within the GRADE framework. Symptom reduction (mean difference/MD), study-defined response (risk ratios/RRs), and acceptability were co-primary outcomes. Adverse events and withdrawal were secondary. Seventy studies were included. In generalized anxiety disorder (GAD), azapirones largely outperformed placebo (MD=-4.91, 95%C.I.[-5.91, -3.90], Hedges'g -1.37 [-1.02, -0.73]), k = 22, n = 2,567; RR=1.64, 95%C.I.[1.45, 1.86], k = 9, n = 1,346). While azapirones overlapped benzodiazepines in symptom reduction (MD=-0.12, 95%C.I.[-0.70, 0.45], k = 34, n = 3,160), they were slightly outperformed in response rate (RR=0.94, 95%C.I.[0.90, 0.99], k = 18, n = 2,423). Azapirones overlapped SRIs (MD=0.09, 95%C.I.[-0.49, 0.67], k = 8, n = 747; RR=0.97, 95%C.I.[0.89, 1.07], k = 7, n = 737). Confidence in estimates was high/moderate vs. placebo, moderate/low vs. benzodiazepine, very-low vs. SRIs. Azapirones failed to outperform the placebo in panic and social anxiety disorders. Azapirones overlapped placebo and SRIs in drop-out rates, while they showed higher treatment discontinuation rates than benzodiazepines (RR=1.33, 95%C.I.[1.16, 1.53], k = 23, n = 2,768). Azapirones caused less sedation/fatigue/drowsiness/weakness/cognitive issues than benzodiazepines, resembling placebo. They caused more nausea and dizziness than placebo, more headache and nausea than benzodiazepines, and less nausea and xerostomia than SRIs. Azapirones proved effective and relatively well-tolerated for GAD. They should be preferred over benzodiazepines, especially in the long-term, considering their lower sedation and addiction potential, representing a potential SRI alternative. Further research is warranted to prove efficacy in panic and social anxiety.
Collapse
Affiliation(s)
- Flavia Rossano
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Claudio Caiazza
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Nicolas Zotti
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Luca Viacava
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Antonella Irano
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Niccolò Solini
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Luca Pistone
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Rosanna Pezone
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Flavia Cilmi
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Claudio Ricci
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy
| | - Michele De Prisco
- Bipolar and Depressive Disorders Unit, Hospìtal Clinic de Barcelona, C. Villarroel, 170, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), c. Villarroel, 170, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Felice Iasevoli
- Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy; Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy
| | - Taro Kishi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ontario, Canada; On Track: The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada; Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| | - Andrea de Bartolomeis
- Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, University School of Medicine of Naples Federico II, Naples, Italy; Laboratory of Molecular and Translational Psychiatry, University School of Medicine of Naples Federico II, Naples, Italy
| | - Michele Fornaro
- Clinical Section of Psychiatry and Psychology - Department of Neuroscience, Reproductive Sciences, and Odontostomatology, University School of Medicine Federico II, Naples, Italy.
| |
Collapse
|
11
|
Kelly L, Brown C, Gibbard AG, Jackson T, Swinny JD. Subunit-specific expression and function of AMPA receptors in the mouse locus coeruleus. J Anat 2023; 243:813-825. [PMID: 37391270 PMCID: PMC10557397 DOI: 10.1111/joa.13922] [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: 02/02/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/02/2023] Open
Abstract
The locus coeruleus (LC) provides the principal supply of noradrenaline (NA) to the brain, thereby modulating an array of brain functions. The release of NA, and therefore its impact on the brain, is governed by LC neuronal excitability. Glutamatergic axons, from various brain regions, topographically innervate different LC sub-domains and directly alter LC excitability. However, it is currently unclear whether glutamate receptor sub-classes, such as α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, are divergently expressed throughout the LC. Immunohistochemistry and confocal microscopy were used to identify and localise individual GluA subunits in the mouse LC. Whole-cell patch clamp electrophysiology and subunit-preferring ligands were used to assess their impact on LC spontaneous firing rate (FR). GluA1 immunoreactive clusters were associated with puncta immunoreactive for VGLUT2 on somata, and VGLUT1 on distal dendrites. GluA4 was associated with these synaptic markers only in the distal dendrites. No specific signal was detected for the GluA2-3 subunits. The GluA1/2 receptor agonist (S)-CPW 399 increased LC FR, whilst the GluA1/3 receptor antagonist philanthotoxin-74 decreased it. 4-[2-(phenylsulfonylamino)ethylthio]-2,6-difluoro-phenoxyacetamide (PEPA), a positive allosteric modulator of GluA3/4 receptors, had no significant effect on spontaneous FR. The data suggest distinct AMPA receptor subunits are targeted to different LC afferent inputs and have contrasting effects on spontaneous neuronal excitability. This precise expression profile could be a mechanism for LC neurons to integrate diverse information contained in various glutamate afferents.
Collapse
Affiliation(s)
- Louise Kelly
- School of Pharmacy & Biomedical SciencesUniversity of PortsmouthPortsmouthUK
| | - Christopher Brown
- School of Pharmacy & Biomedical SciencesUniversity of PortsmouthPortsmouthUK
| | - Adina G. Gibbard
- School of Pharmacy & Biomedical SciencesUniversity of PortsmouthPortsmouthUK
| | - Torquil Jackson
- School of Pharmacy & Biomedical SciencesUniversity of PortsmouthPortsmouthUK
| | - Jerome D. Swinny
- School of Pharmacy & Biomedical SciencesUniversity of PortsmouthPortsmouthUK
| |
Collapse
|
12
|
Sun L, Zhu M, Wang M, Hao Y, Hao Y, Jing X, Yu H, Shi Y, Zhang X, Wang S, Yuan F, Yuan XS. Whole-brain monosynaptic inputs and outputs of leptin receptor b neurons of the nucleus tractus solitarii in mice. Brain Res Bull 2023; 201:110693. [PMID: 37348822 DOI: 10.1016/j.brainresbull.2023.110693] [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: 04/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
The nucleus tractus solitarii (NTS) is the primary central station that integrates visceral afferent information and regulates respiratory, gastrointestinal, cardiovascular, and other physiological functions. Leptin receptor b (LepRb)-expressing neurons of the NTS (NTSLepRb neurons) are implicated in central respiration regulation, respiratory facilitation, and respiratory drive enhancement. Furthermore, LepRb dysfunction is involved in obesity, insulin resistance, and sleep-disordered breathing. However, the monosynaptic inputs and outputs of NTSLepRb neurons in whole-brain mapping remain to be elucidated. Therefore, the exploration of its whole-brain connection system may provide strong support for comprehensively understanding the physiological and pathological functions of NTSLepRb neurons. In the present study, we used a cell type-specific, modified rabies virus and adeno-associated virus with the Cre-loxp system to map monosynaptic inputs and outputs of NTSLepRb neurons in LepRb-Cre mice. The results showed that NTSLepRb neurons received inputs from 48 nuclei in the whole brain from five brain regions, including especially the medulla. We found that NTSLepRb neurons received inputs from nuclei associated with respiration, such as the pre-Bötzinger complex, ambiguus nucleus, and parabrachial nucleus. Interestingly, some brain areas related to cardiovascular regulation-i.e., the ventrolateral periaqueductal gray and locus coeruleus-also sent a small number of inputs to NTSLepRb neurons. In addition, anterograde tracing results demonstrated that NTSLepRb neurons sent efferent projections to 15 nuclei, including the dorsomedial hypothalamic nucleus and arcuate hypothalamic nucleus, which are involved in regulation of energy metabolism and feeding behaviors. Quantitative statistical analysis revealed that the inputs of the whole brain to NTSLepRb neurons were significantly greater than the outputs. Our study comprehensively revealed neuronal connections of NTSLepRb neurons in the whole brain and provided a neuroanatomical basis for further research on physiological and pathological functions of NTSLepRb neurons.
Collapse
Affiliation(s)
- Lu Sun
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mengchu Zhu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yinchao Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yaxin Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Xinyi Jing
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China
| | - Yishuo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Xiang Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China.
| | - Xiang Shan Yuan
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
13
|
Caddye E, Pineau J, Reyniers J, Ronen I, Colasanti A. Lactate: A Theranostic Biomarker for Metabolic Psychiatry? Antioxidants (Basel) 2023; 12:1656. [PMID: 37759960 PMCID: PMC10526106 DOI: 10.3390/antiox12091656] [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: 07/05/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
Alterations in neurometabolism and mitochondria are implicated in the pathophysiology of psychiatric conditions such as mood disorders and schizophrenia. Thus, developing objective biomarkers related to brain mitochondrial function is crucial for the development of interventions, such as central nervous system penetrating agents that target brain health. Lactate, a major circulatory fuel source that can be produced and utilized by the brain and body, is presented as a theranostic biomarker for neurometabolic dysfunction in psychiatric conditions. This concept is based on three key properties of lactate that make it an intriguing metabolic intermediate with implications for this field: Firstly, the lactate response to various stimuli, including physiological or psychological stress, represents a quantifiable and dynamic marker that reflects metabolic and mitochondrial health. Second, lactate concentration in the brain is tightly regulated according to the sleep-wake cycle, the dysregulation of which is implicated in both metabolic and mood disorders. Third, lactate universally integrates arousal behaviours, pH, cellular metabolism, redox states, oxidative stress, and inflammation, and can signal and encode this information via intra- and extracellular pathways in the brain. In this review, we expand on the above properties of lactate and discuss the methodological developments and rationale for the use of functional magnetic resonance spectroscopy for in vivo monitoring of brain lactate. We conclude that accurate and dynamic assessment of brain lactate responses might contribute to the development of novel and personalized therapies that improve mitochondrial health in psychiatric disorders and other conditions associated with neurometabolic dysfunction.
Collapse
Affiliation(s)
- Edward Caddye
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| | - Julien Pineau
- Independent Researcher, Florianópolis 88062-300, Brazil
| | - Joshua Reyniers
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- School of Life Sciences, University of Sussex, Falmer BN1 9RR, UK
| | - Itamar Ronen
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| | - Alessandro Colasanti
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| |
Collapse
|
14
|
Krohn F, Novello M, van der Giessen RS, De Zeeuw CI, Pel JJM, Bosman LWJ. The integrated brain network that controls respiration. eLife 2023; 12:83654. [PMID: 36884287 PMCID: PMC9995121 DOI: 10.7554/elife.83654] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/29/2023] [Indexed: 03/09/2023] Open
Abstract
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
Collapse
Affiliation(s)
- Friedrich Krohn
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
15
|
da Silva Junior CA, Marques DA, Patrone LGA, Biancardi V, Bícego KC, Gargaglioni LH. Intra-uterine diazepam exposure decreases the number of catecholaminergic and serotoninergic neurons of neonate rats. Neurosci Lett 2023; 795:137014. [PMID: 36521643 DOI: 10.1016/j.neulet.2022.137014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Benzodiazepines, such as diazepam (DZP), are used to treat anxiety disorders, and are prescribed to pregnant woman for therapeutic purposes. Concerns regarding their consequences on postnatal development rise as they cross the placenta and interact with the embryo. Occurrence of malformation and behavioral syndromes have been reported for different ages, but little is known about their effects on the brain after exposure during intrauterine life. Thus, we sought to evaluate the effects of intrauterine exposure to DZP on the number of brainstem's catecholaminergic and serotonergic neurons, implicated in respiratory control, in male and female rats on postnatal (P) day 12-13, using immunofluorescence labeling for tyrosine-hydroxylase (TH) and serotonin (5-HT). We observed a reduction in the number of catecholaminergic neurons for males and females. Special attention is given to the reduction in the density of neurons in the A6 region, involved in ventilatory responses to CO2. Interestingly, only males showed a reduction in the number of serotonergic neurons, while females were not affected. These findings suggest that in utero exposure to DZP results in deleterious neuroanatomical effects on P12-13 rats and raises a note of concern for women clinicians to make more informed choices about the use of anxiolytic treatments during gestation.
Collapse
Affiliation(s)
- Carlos Aparecido da Silva Junior
- Department of Applied Science, William & Mary, Williamsburg, VA, United States; Department of Animal Morphology and Physiology, FCAV - UNESP - São Paulo State University, Jaboticabal, SP, Brazil
| | - Danuzia A Marques
- Department of Pediatrics, Québec Heart and Lung Institute, Université Laval, Québec, QC, Canada
| | - Luís Gustavo A Patrone
- Department of Animal Morphology and Physiology, FCAV - UNESP - São Paulo State University, Jaboticabal, SP, Brazil
| | - Vivian Biancardi
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, FCAV - UNESP - São Paulo State University, Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, FCAV - UNESP - São Paulo State University, Jaboticabal, SP, Brazil.
| |
Collapse
|
16
|
Patrone LGA, Ferrari GD, da Silva RM, Alberici LC, Lopes NP, Stabile AM, Klein W, Bícego KC, Gargaglioni LH. Sex- and age-specific respiratory alterations induced by prenatal exposure to the cannabinoid receptor agonist WIN 55,212-2 in rats. Br J Pharmacol 2023. [PMID: 36710256 DOI: 10.1111/bph.16044] [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: 01/27/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabis legalization has risen in many countries, and its use during pregnancy has increased. The endocannabinoid system is present in the CNS at early stages of embryonic development, and regulates functional brain maturation including areas responsible for respiratory control, data on the influence of external cannabinoids on the development of the respiratory system and possible consequences during postnatal life are limited. EXPERIMENTAL APPROACH We evaluated the effects of prenatal exposure to synthetic cannabinoid (WIN 55,212-2 [WIN], 0.5 mg·kg-1 ·day-1 ) on the respiratory control system in neonatal (P0, P6-7 and P12-13) and juvenile (P27-28) male and female rats. KEY RESULTS WIN administration to pregnant rats interfered sex-specifically with breathing regulation of offspring, promoting a greater sensitivity to CO2 at all ages in males (except P6-7) and in juvenile females. An altered hypoxic chemoreflex was observed in P0 (hyperventilation) and P6-7 (hypoventilation) males, which was absent in females. Along with breathing alterations, brainstem analysis showed an increase in the number of catecholaminergic neurons and cannabinoid receptor type 1 (CB1 ) and changes in tissue respiration in the early males. A reduction in pulmonary compliance was observed in juvenile male rats. Preexposure to WIN enhanced spontaneous apnoea and reduced the number of serotoninergic (5-HT) neurons in the raphe magnus nucleus of P0 females. CONCLUSIONS AND IMPLICATIONS These data demonstrate that excess stimulation of the endocannabinoid system during gestation has prolonged and sex-specific consequences for the respiratory control system.
Collapse
Affiliation(s)
- Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, São Paulo State University - UNESP/FCAV, Jaboticabal, São Paulo, Brazil
| | - Gustavo D Ferrari
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Moreira da Silva
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luciane C Alberici
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Norberto Peporine Lopes
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Angelita M Stabile
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Wilfried Klein
- Department of Biology, School of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, São Paulo State University - UNESP/FCAV, Jaboticabal, São Paulo, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, São Paulo State University - UNESP/FCAV, Jaboticabal, São Paulo, Brazil
| |
Collapse
|
17
|
Rodrigues LTC, Patrone LGA, Gargaglioni LH, Dias MB. Melanin-concentrating hormone regulates the hypercapnic chemoreflex by acting in the lateral hypothalamic area. Exp Physiol 2022; 107:1298-1311. [PMID: 35930596 DOI: 10.1113/ep090318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/22/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? MCH suppresses the hypercapnic chemoreflex but the mechanism by which this effect is produced has not been previously explored. What is the main finding and its importance? MCH acting in the lateral hypothalamic area but not in the locus coeruleus in rats, in the light period, attenuates the hypercapnic chemoreflex. Our data provide new insight regarding the role of MCH in the modulation of the hypercapnic ventilatory response. ABSTRACT Melanin-concentrating hormone (MCH) is a hypothalamic neuropeptide involved in a broad range of homeostatic functions including regulation of the hypercapnic chemoreflex. We evaluated whether MCH modulates the hypercapnic ventilatory response by acting in the lateral hypothalamic area (LHA) and/or in the locus coeruleus (LC). Here, we measured pulmonary ventilation (VE ), body temperature, electroencephalogram (EEG) and electromyogram (EMG) of unanesthetized adult male Wistar rats before and after microinjection of MCH [0.4 mM] or MCH1-R antagonist (SNAP-94847 [63 mM]) into the LHA and LC, in room air and 7% CO2 conditions during wakefulness and sleep, in the dark and light periods. MCH intra-LHA caused a decreased CO2 ventilatory response during wakefulness and sleep in the light period, while SNAP-94847 intra-LHA increased this response, during wakefulness in the light period. In the LC, MCH or the MCH1-R antagonist caused no change in the hypercapnic ventilatory response. Our results suggest that MCH, in the LHA, exerts an inhibitory modulation of the hypercapnic ventilatory response during the light-inactive period in rats. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laísa T C Rodrigues
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Mirela B Dias
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| |
Collapse
|
18
|
da Silva Junior CA, Patrone LGA, Biancardi V, Vilela-Costa HH, Marques DA, Cristina-Silva C, da Costa Silva KS, Bícego KC, Szawka RE, Gargaglioni LH. Sexually dimorphic effects of prenatal diazepam exposure on respiratory control and the monoaminergic system of neonate and young rats. Pflugers Arch 2022; 474:1185-1200. [DOI: 10.1007/s00424-022-02730-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/05/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022]
|
19
|
Noga BR, Whelan PJ. The Mesencephalic Locomotor Region: Beyond Locomotor Control. Front Neural Circuits 2022; 16:884785. [PMID: 35615623 PMCID: PMC9124768 DOI: 10.3389/fncir.2022.884785] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/14/2022] [Indexed: 11/25/2022] Open
Abstract
The mesencephalic locomotor region (MLR) was discovered several decades ago in the cat. It was functionally defined based on the ability of low threshold electrical stimuli within a region comprising the cuneiform and pedunculopontine nucleus to evoke locomotion. Since then, similar regions have been found in diverse vertebrate species, including the lamprey, skate, rodent, pig, monkey, and human. The MLR, while often viewed under the lens of locomotion, is involved in diverse processes involving the autonomic nervous system, respiratory system, and the state-dependent activation of motor systems. This review will discuss the pedunculopontine nucleus and cuneiform nucleus that comprises the MLR and examine their respective connectomes from both an anatomical and functional angle. From a functional perspective, the MLR primes the cardiovascular and respiratory systems before the locomotor activity occurs. Inputs from a variety of higher structures, and direct outputs to the monoaminergic nuclei, allow the MLR to be able to respond appropriately to state-dependent locomotion. These state-dependent effects are roughly divided into escape and exploratory behavior, and the MLR also can reinforce the selection of these locomotor behaviors through projections to adjacent structures such as the periaqueductal gray or to limbic and cortical regions. Findings from the rat, mouse, pig, and cat will be discussed to highlight similarities and differences among diverse species.
Collapse
Affiliation(s)
- Brian R. Noga
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- *Correspondence: Brian R. Noga Patrick J. Whelan
| | - Patrick J. Whelan
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Brian R. Noga Patrick J. Whelan
| |
Collapse
|
20
|
Lusk SJ, McKinney A, Hunt PJ, Fahey PG, Patel J, Chang A, Sun JJ, Martinez VK, Zhu PJ, Egbert JR, Allen G, Jiang X, Arenkiel BR, Tolias AS, Costa-Mattioli M, Ray RS. A CRISPR toolbox for generating intersectional genetic mouse models for functional, molecular, and anatomical circuit mapping. BMC Biol 2022; 20:28. [PMID: 35086530 PMCID: PMC8796356 DOI: 10.1186/s12915-022-01227-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 01/06/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The functional understanding of genetic interaction networks and cellular mechanisms governing health and disease requires the dissection, and multifaceted study, of discrete cell subtypes in developing and adult animal models. Recombinase-driven expression of transgenic effector alleles represents a significant and powerful approach to delineate cell populations for functional, molecular, and anatomical studies. In addition to single recombinase systems, the expression of two recombinases in distinct, but partially overlapping, populations allows for more defined target expression. Although the application of this method is becoming increasingly popular, its experimental implementation has been broadly restricted to manipulations of a limited set of common alleles that are often commercially produced at great expense, with costs and technical challenges associated with production of intersectional mouse lines hindering customized approaches to many researchers. Here, we present a simplified CRISPR toolkit for rapid, inexpensive, and facile intersectional allele production. RESULTS Briefly, we produced 7 intersectional mouse lines using a dual recombinase system, one mouse line with a single recombinase system, and three embryonic stem (ES) cell lines that are designed to study the way functional, molecular, and anatomical features relate to each other in building circuits that underlie physiology and behavior. As a proof-of-principle, we applied three of these lines to different neuronal populations for anatomical mapping and functional in vivo investigation of respiratory control. We also generated a mouse line with a single recombinase-responsive allele that controls the expression of the calcium sensor Twitch-2B. This mouse line was applied globally to study the effects of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) on calcium release in the ovarian follicle. CONCLUSIONS The lines presented here are representative examples of outcomes possible with the successful application of our genetic toolkit for the facile development of diverse, modifiable animal models. This toolkit will allow labs to create single or dual recombinase effector lines easily for any cell population or subpopulation of interest when paired with the appropriate Cre and FLP recombinase mouse lines or viral vectors. We have made our tools and derivative intersectional mouse and ES cell lines openly available for non-commercial use through publicly curated repositories for plasmid DNA, ES cells, and transgenic mouse lines.
Collapse
Affiliation(s)
- Savannah J Lusk
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Andrew McKinney
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Patrick J Hunt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Paul G Fahey
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Jay Patel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Andersen Chang
- Department of Statistics, Rice University, Houston, TX, USA
| | - Jenny J Sun
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Vena K Martinez
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Ping Jun Zhu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Jeremy R Egbert
- Department of Cell Biology, University of Connecticut, Farmington, CT, USA
| | - Genevera Allen
- Department of Statistics, Computer Science, and Electrical and Computer Engineering, Rice University, Houston, TX, USA
- Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Xiaolong Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Houston, TX, USA
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | | | - Russell S Ray
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Houston, TX, USA.
| |
Collapse
|
21
|
Locus Coeruleus in Non-Mammalian Vertebrates. Brain Sci 2022; 12:brainsci12020134. [PMID: 35203898 PMCID: PMC8870555 DOI: 10.3390/brainsci12020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/08/2022] [Accepted: 01/15/2022] [Indexed: 11/30/2022] Open
Abstract
The locus coeruleus (LC) is a vertebrate-specific nucleus and the primary source of norepinephrine (NE) in the brain. This nucleus has conserved properties across species: highly homogeneous cell types, a small number of cells but extensive axonal projections, and potent influence on brain states. Comparative studies on LC benefit greatly from its homogeneity in cell types and modularity in projection patterns, and thoroughly understanding the LC-NE system could shed new light on the organization principles of other more complex modulatory systems. Although studies on LC are mainly focused on mammals, many of the fundamental properties and functions of LC are readily observable in other vertebrate models and could inform mammalian studies. Here, we summarize anatomical and functional studies of LC in non-mammalian vertebrate classes, fish, amphibians, reptiles, and birds, on topics including axonal projections, gene expressions, homeostatic control, and modulation of sensorimotor transformation. Thus, this review complements mammalian studies on the role of LC in the brain.
Collapse
|
22
|
Kubin L. Breathing during sleep. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:179-199. [PMID: 35965026 DOI: 10.1016/b978-0-323-91534-2.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The depth, rate, and regularity of breathing change following transition from wakefulness to sleep. Interactions between sleep and breathing involve direct effects of the central mechanisms that generate sleep states exerted at multiple respiratory regulatory sites, such as the central respiratory pattern generator, respiratory premotor pathways, and motoneurons that innervate the respiratory pump and upper airway muscles, as well as effects secondary to sleep-related changes in metabolism. This chapter discusses respiratory effects of sleep as they occur under physiologic conditions. Breathing and central respiratory neuronal activities during nonrapid eye movement (NREM) sleep and REM sleep are characterized in relation to activity of central wake-active and sleep-active neurons. Consideration is given to the obstructive sleep apnea syndrome because in this common disorder, state-dependent control of upper airway patency by upper airway muscles attains high significance and recurrent arousals from sleep are triggered by hypercapnic and hypoxic episodes. Selected clinical trials are discussed in which pharmacological interventions targeted transmission in noradrenergic, serotonergic, cholinergic, and other state-dependent pathways identified as mediators of ventilatory changes during sleep. Central pathways for arousals elicited by chemical stimulation of breathing are given special attention for their important role in sleep loss and fragmentation in sleep-related respiratory disorders.
Collapse
Affiliation(s)
- Leszek Kubin
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Abstract
Brain PCO2 is sensed primarily via changes in [H+]. Small pH changes are detected in the medulla oblongata and trigger breathing adjustments that help maintain arterial PCO2 constant. Larger perturbations of brain CO2/H+, possibly also sensed elsewhere in the CNS, elicit arousal, dyspnea, and stress, and cause additional breathing modifications. The retrotrapezoid nucleus (RTN), a rostral medullary cluster of glutamatergic neurons identified by coexpression of Phoxb and Nmb transcripts, is the lynchpin of the central respiratory chemoreflex. RTN regulates breathing frequency, inspiratory amplitude, and active expiration. It is exquisitely responsive to acidosis in vivo and maintains breathing autorhythmicity during quiet waking, slow-wave sleep, and anesthesia. The RTN response to [H+] is partly an intrinsic neuronal property mediated by proton sensors TASK-2 and GPR4 and partly a paracrine effect mediated by astrocytes and the vasculature. The RTN also receives myriad excitatory or inhibitory synaptic inputs including from [H+]-responsive neurons (e.g., serotonergic). RTN is silenced by moderate hypoxia. RTN inactivity (periodic or sustained) contributes to periodic breathing and, likely, to central sleep apnea. RTN development relies on transcription factors Egr2, Phox2b, Lbx1, and Atoh1. PHOX2B mutations cause congenital central hypoventilation syndrome; they impair RTN development and consequently the central respiratory chemoreflex.
Collapse
Affiliation(s)
- Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States.
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
24
|
Guyenet PG, Stornetta RL. Rostral ventrolateral medulla, retropontine region and autonomic regulations. Auton Neurosci 2021; 237:102922. [PMID: 34814098 DOI: 10.1016/j.autneu.2021.102922] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022]
Abstract
The rostral half of the ventrolateral medulla (RVLM) and adjacent ventrolateral retropontine region (henceforth RVLMRP) have been divided into various sectors by neuroscientists interested in breathing or autonomic regulations. The RVLMRP regulates respiration, glycemia, vigilance and inflammation, in addition to blood pressure. It contains interoceptors that respond to acidification, hypoxia and intracranial pressure and its rostral end contains the retrotrapezoid nucleus (RTN) which is the main central respiratory chemoreceptor. Acid detection by the RTN is an intrinsic property of the principal neurons that is enhanced by paracrine influences from surrounding astrocytes and CO2-dependent vascular constriction. RTN mediates the hypercapnic ventilatory response via complex projections to the respiratory pattern generator (CPG). The RVLM contributes to autonomic response patterns via differential recruitment of several subtypes of adrenergic (C1) and non-adrenergic neurons that directly innervate sympathetic and parasympathetic preganglionic neurons. The RVLM also innervates many brainstem and hypothalamic nuclei that contribute, albeit less directly, to autonomic responses. All lower brainstem noradrenergic clusters including the locus coeruleus are among these targets. Sympathetic tone to the circulatory system is regulated by subsets of presympathetic RVLM neurons whose activity is continuously restrained by the baroreceptors and modulated by the respiratory CPG. The inhibitory input from baroreceptors and the excitatory input from the respiratory CPG originate from neurons located in or close to the rhythm generating region of the respiratory CPG (preBötzinger complex).
Collapse
Affiliation(s)
- Patrice G Guyenet
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| | - Ruth L Stornetta
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| |
Collapse
|
25
|
A Bridge between the Breath and the Brain: Synchronization of Respiration, a Pupillometric Marker of the Locus Coeruleus, and an EEG Marker of Attentional Control State. Brain Sci 2021; 11:brainsci11101324. [PMID: 34679389 PMCID: PMC8534189 DOI: 10.3390/brainsci11101324] [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: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
Yogic and meditative traditions have long held that the fluctuations of the breath and the mind are intimately related. While respiratory modulation of cortical activity and attentional switching are established, the extent to which electrophysiological markers of attention exhibit synchronization with respiration is unknown. To this end, we examined (1) frontal midline theta-beta ratio (TBR), an indicator of attentional control state known to correlate with mind wandering episodes and functional connectivity of the executive control network; (2) pupil diameter (PD), a known proxy measure of locus coeruleus (LC) noradrenergic activity; and (3) respiration for evidence of phase synchronization and information transfer (multivariate Granger causality) during quiet restful breathing. Our results indicate that both TBR and PD are simultaneously synchronized with the breath, suggesting an underlying oscillation of an attentionally relevant electrophysiological index that is phase-locked to the respiratory cycle which could have the potential to bias the attentional system into switching states. We highlight the LC’s pivotal role as a coupling mechanism between respiration and TBR, and elaborate on its dual functions as both a chemosensitive respiratory nucleus and a pacemaker of the attentional system. We further suggest that an appreciation of the dynamics of this weakly coupled oscillatory system could help deepen our understanding of the traditional claim of a relationship between breathing and attention.
Collapse
|
26
|
Sainju RK, Dragon DN, Winnike HB, Ten Eyck P, Granner MA, Gehlbach BK, Richerson GB. Hypercapnic ventilatory response in epilepsy patients treated with VNS: A case-control study. Epilepsia 2021; 62:e140-e146. [PMID: 34265074 PMCID: PMC9113047 DOI: 10.1111/epi.16997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Central CO2 chemoreception (CCR), a major chemical drive for breathing, can be quantified with a CO2 re-breathing test to measure the hypercapnic ventilatory response (HCVR). An attenuated HCVR correlates with the severity of respiratory dysfunction after generalized convulsive seizures and is a potential biomarker for sudden unexpected death in epilepsy (SUDEP) risk. Vagus nerve stimulation (VNS) may reduce SUDEP risk, but for unclear reasons the risk remains higher during the first 2 years after implantation. The vagus nerve has widespread connections in the brainstem, including key areas related to CCR. Here we examined whether chronic electrical stimulation of the vagus nerve induces changes in CCR. METHODS We compared the HCVR in epilepsy patients with or without an active VNS in a sex- and age-matched case-control study. Eligible subjects were selected from a cohort of patients who previously underwent HCVR testing. The HCVR slope, change in minute ventilation (VE) with respect to change in end tidal (ET) CO2 (∆ VE/ ∆ ETCO2) during the test was calculated for each subject. Key variables were compared between the two groups. Univariate and multivariate analyses were carried out for HCVR slope as dependent variable. RESULTS A total of 86 subjects were in the study. HCVR slope was significantly lower in the cases compared to the controls. Cases had longer duration of epilepsy and higher number of anti-epileptic drugs (AEDs) tried during lifetime. Having active VNS and ETCO2 were associated with a low HCVR slope while high BMI was associated with high HCVR slope in both univariate and multivariate analyses. DISCUSSION We found having an active VNS was associated with relatively attenuated HCVR slope. Although duration of epilepsy and number of AEDs tried during lifetime was significantly different between the groups, they were not predictors of HCVR slope in subsequent analysis. CONCLUSION Chronic electrical stimulation of the vagus nerve by VNS may be associated with an attenuated CCR [Correction added on 24 November 2021, after first online publication: The preceding sentence has been revised from “Chronic electrical stimulation of VNS nerve by VNS…”]. A larger prospective study may help to establish the time course of this effect in relation to the time of VNS implantation, whether there is a causal relationship, and determine how it affects SUDEP risk.
Collapse
Affiliation(s)
- Rup K Sainju
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deidre N Dragon
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Harold B Winnike
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Patrick Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, USA
| | - Mark A Granner
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Brian K Gehlbach
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - George B Richerson
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Veterans Affairs Medical Center, Iowa City, IA, USA
| |
Collapse
|
27
|
Astrocytic contribution to glutamate-related central respiratory chemoreception in vertebrates. Respir Physiol Neurobiol 2021; 294:103744. [PMID: 34302992 DOI: 10.1016/j.resp.2021.103744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/01/2021] [Accepted: 07/18/2021] [Indexed: 12/24/2022]
Abstract
Central respiratory chemoreceptors play a key role in the respiratory homeostasis by sensing CO2 and H+ in brain and activating the respiratory neural network. This ability of specific brain regions to respond to acidosis and hypercapnia is based on neuronal and glial mechanisms. Several decades ago, glutamatergic transmission was proposed to be involved as a main mechanism in central chemoreception. However, a complete identification of mechanism has been elusive. At the rostral medulla, chemosensitive neurons of the retrotrapezoid nucleus (RTN) are glutamatergic and they are stimulated by ATP released by RTN astrocytes in response to hypercapnia. In addition, recent findings show that caudal medullary astrocytes in brainstem can also contribute as CO2 and H+ sensors that release D-serine and glutamate, both gliotransmitters able to activate the respiratory neural network. In this review, we describe the mammalian astrocytic glutamatergic contribution to the central respiratory chemoreception trying to trace in vertebrates the emergence of several components involved in this process.
Collapse
|
28
|
Lim R, Messineo L, Grunstein RR, Carberry JC, Eckert DJ. The noradrenergic agent reboxetine plus the antimuscarinic hyoscine butylbromide reduces sleep apnoea severity: a double-blind, placebo-controlled, randomised crossover trial. J Physiol 2021; 599:4183-4195. [PMID: 34174090 DOI: 10.1113/jp281912] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/23/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Recent animal and human physiology studies indicate that noradrenergic and muscarinic processes are key mechanisms that mediate pharyngeal muscle control during sleep. The noradrenergic agent reboxetine combined with the anti-muscarinic hyoscine butylbromide has recently been shown to improve upper airway function during sleep in healthy individuals. However, whether these findings translate to the clinically relevant patient population of people with obstructive sleep apnoea (OSA), and the effects of the agents on OSA severity, are unknown. We found that reboxetine plus hyoscine butylbromide reduced OSA severity, including overnight hypoxaemia, via increases in pharyngeal muscle responsiveness, improvements in respiratory control and airway collapsibility without changing the respiratory arousal threshold. These findings provide mechanistic insight into the role of noradrenergic and anti-muscarinic agents on upper airway stability and breathing during sleep and are important for pharmacotherapy development for OSA. ABSTRACT The noradrenergic agent reboxetine combined with the anti-muscarinic hyoscine butylbromide has recently been shown to improve upper airway function during sleep in healthy individuals. However, the effects of this drug combination on obstructive sleep apnoea (OSA) severity are unknown. Accordingly, this study aimed to determine if reboxetine plus hyoscine butylbromide reduces OSA severity. Secondary aims were to investigate the effects on key upper airway physiology and endotypic traits. Twelve people with OSA aged 52 ± 13 years, BMI = 30 ± 5 kg/m2 , completed a double-blind, randomised, placebo-controlled, crossover trial (ACTRN12617001326381). Two in-laboratory sleep studies with nasal mask, pneumotachograph, epiglottic pressure sensor and bipolar fine-wire electrodes into genioglossus and tensor palatini muscles were performed separated by approximately 1 week. Each participant received either reboxetine (4 mg) plus hyoscine butylbromide (20 mg), or placebo immediately prior to sleep. Polysomnography, upper airway physiology and endotypic estimates of OSA were compared between conditions. Reboxetine plus hyoscine butylbromide reduced the apnoea/hypopnoea index by (mean ± SD) 17 ± 17 events/h from 51 ± 30 to 33 ± 22 events/h (P = 0.005) and nadir oxygen saturation increased by 6 ± 5% from 82 ± 5 to 88 ± 2% (P = 0.002). The drug combination increased tonic genioglossus muscle responsiveness during non-REM sleep (median [25th, 75th centiles]: -0.007 [-0.0004, -0.07] vs. -0.12 [-0.02, -0.40] %maxEMG/cmH2 O, P = 0.02), lowered loop gain (0.43 ± 0.06 vs. 0.39 ± 0.07, P = 0.01), and improved airway collapsibility (90 [69, 95] vs. 93 [88, 96] %eupnoea, P = 0.02), without changing the arousal threshold (P = 0.39). These findings highlight the important role that noradrenergic and muscarinic processes have on upper airway function during sleep and the potential for pharmacotherapy to target these mechanisms to treat OSA.
Collapse
Affiliation(s)
- Richard Lim
- Neuroscience Research Australia (NeuRA), New South Wales, Sydney, Australia.,School of Medical Sciences, University of New South Wales, New South Wales, Sydney, Australia
| | - Ludovico Messineo
- Flinders Health and Medical Research Institute and Adelaide Institute for Sleep Health, Flinders University, Bedford Park, South Australia, Australia
| | - Ronald R Grunstein
- Woolcock Institute of Medical Research, Sydney Medical School, the University of Sydney, New South Wales, Glebe, Australia.,Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, New South Wales, Camperdown, Australia
| | - Jayne C Carberry
- Neuroscience Research Australia (NeuRA), New South Wales, Sydney, Australia.,Flinders Health and Medical Research Institute and Adelaide Institute for Sleep Health, Flinders University, Bedford Park, South Australia, Australia.,UCD School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Danny J Eckert
- Neuroscience Research Australia (NeuRA), New South Wales, Sydney, Australia.,School of Medical Sciences, University of New South Wales, New South Wales, Sydney, Australia.,Flinders Health and Medical Research Institute and Adelaide Institute for Sleep Health, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
29
|
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: 2.0] [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
|
30
|
Locus Coeruleus Acid-Sensing Ion Channels Modulate Sleep-Wakefulness and State Transition from NREM to REM Sleep in the Rat. Neurosci Bull 2021; 37:684-700. [PMID: 33638800 DOI: 10.1007/s12264-020-00625-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
The locus coeruleus (LC) is one of the essential chemoregulatory and sleep-wake (S-W) modulating centers in the brain. LC neurons remain highly active during wakefulness, and some implicitly become silent during rapid eye movement (REM) sleep. LC neurons are also involved in CO2-dependent modulation of the respiratory drive. Acid-sensing ion channels (ASICs) are highly expressed in some brainstem chemosensory breathing regulatory areas, but their localization and functions in the LC remain unknown. Mild hypercapnia increases the amount of non-REM (NREM) sleep and the number of REM sleep episodes, but whether ASICs in the LC modulate S-W is unclear. Here, we investigated the presence of ASICs in the LC and their role in S-W modulation and the state transition from NREM to REM sleep. Male Wistar rats were surgically prepared for chronic polysomnographic recordings and drug microinjections into the LC. The presence of ASIC-2 and ASIC-3 in the LC was immunohistochemically characterized. Microinjections of amiloride (an ASIC blocker) and APETx2 (a blocker of ASIC-2 and -3) into the LC significantly decreased wakefulness and REM sleep, but significantly increased NREM sleep. Mild hypercapnia increased the amount of NREM and the number of REM episodes. However, APETx2 microinjection inhibited this increase in REM frequency. These results suggest that the ASICs of LC neurons modulate S-W, indicating that ASICs could play an important role in vigilance-state transition. A mild increase in CO2 level during NREM sleep sensed by ASICs could be one of the determinants of state transition from NREM to REM sleep.
Collapse
|
31
|
Di Lascio S, Benfante R, Cardani S, Fornasari D. Research Advances on Therapeutic Approaches to Congenital Central Hypoventilation Syndrome (CCHS). Front Neurosci 2021; 14:615666. [PMID: 33510615 PMCID: PMC7835644 DOI: 10.3389/fnins.2020.615666] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Congenital central hypoventilation syndrome (CCHS) is a genetic disorder of neurodevelopment, with an autosomal dominant transmission, caused by heterozygous mutations in the PHOX2B gene. CCHS is a rare disorder characterized by hypoventilation due to the failure of autonomic control of breathing. Until now no curative treatment has been found. PHOX2B is a transcription factor that plays a crucial role in the development (and maintenance) of the autonomic nervous system, and in particular the neuronal structures involved in respiratory reflexes. The underlying pathogenetic mechanism is still unclear, although studies in vivo and in CCHS patients indicate that some neuronal structures may be damaged. Moreover, in vitro experimental data suggest that transcriptional dysregulation and protein misfolding may be key pathogenic mechanisms. This review summarizes latest researches that improved the comprehension of the molecular pathogenetic mechanisms responsible for CCHS and discusses the search for therapeutic intervention in light of the current knowledge about PHOX2B function.
Collapse
Affiliation(s)
- Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy.,CNR-Institute of Neuroscience, Milan, Italy.,NeuroMi-Milan Center for Neuroscience, University of Milano Bicocca, Milan, Italy
| | - Silvia Cardani
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy.,CNR-Institute of Neuroscience, Milan, Italy
| |
Collapse
|
32
|
Wu RN, Hung WC, Chen CT, Tsai LP, Lai WS, Min MY, Wong SB. Firing activity of locus coeruleus noradrenergic neurons decreases in necdin-deficient mice, an animal model of Prader-Willi syndrome. J Neurodev Disord 2020; 12:21. [PMID: 32727346 PMCID: PMC7389383 DOI: 10.1186/s11689-020-09323-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/17/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by multiple respiratory, cognitive, endocrine, and behavioral symptoms, such as central apnea, intellectual disabilities, exaggerated stress responses, and temper tantrums. The locus coeruleus noradrenergic system (LC-NE) modulates a diverse range of behaviors, including arousal, learning, pain modulation, and stress-induced negative affective states, which are possibly correlated with the pathogenesis of PWS phenotypes. Therefore, we evaluated the LC-NE neuronal activity of necdin-deficient mice, an animal model of PWS. METHODS Heterozygous necdin-deficient mice (B6.Cg-Ndntm1ky) were bred from wild-type (WT) females to generate WT (+m/+p) and heterozygotes (+m/-p) animals, which were examined of LC-NE neuronal activity, developmental reflexes, and plethysmography. RESULTS On slice electrophysiology, LC-NE neurons of Ndntm1ky mice with necdin deficiency showed significantly decreased spontaneous activities and impaired excitability, which was mediated by enhanced A-type voltage-dependent potassium currents. Ndntm1ky mice also exhibited the neonatal phenotypes of PWS, such as hypotonia and blunt respiratory responses to hypercapnia. CONCLUSIONS LC-NE neuronal firing activity decreased in necdin-deficient mice, suggesting that LC, the primary source of norepinephrine in the central nervous system, is possibly involved in PWS pathogenesis.
Collapse
Affiliation(s)
- Rui-Ni Wu
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan
| | - Wei-Chen Hung
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Ching-Tsuey Chen
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Li-Ping Tsai
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan
- School of Medicine, Tzu Chi University, No. 701, Sec 3, Jhongyang Rd, Hualien, 97071, Taiwan
| | - Wen-Sung Lai
- Department of Psychology, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, No. 1, Sec 4, Roosevelt Rd, Taipei, 10617, Taiwan
| | - Shi-Bing Wong
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 289, Jiangguo Rd, Xindian Dist, New Taipei City, 23142, Taiwan.
- School of Medicine, Tzu Chi University, No. 701, Sec 3, Jhongyang Rd, Hualien, 97071, Taiwan.
| |
Collapse
|
33
|
Liu N, Fu C, Yu H, Wang Y, Shi L, Hao Y, Yuan F, Zhang X, Wang S. Respiratory Control by Phox2b-expressing Neurons in a Locus Coeruleus-preBötzinger Complex Circuit. Neurosci Bull 2020; 37:31-44. [PMID: 32468398 DOI: 10.1007/s12264-020-00519-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
The locus coeruleus (LC) has been implicated in the control of breathing. Congenital central hypoventilation syndrome results from mutation of the paired-like homeobox 2b (Phox2b) gene that is expressed in LC neurons. The present study was designed to address whether stimulation of Phox2b-expressing LC (Phox2bLC) neurons affects breathing and to reveal the putative circuit mechanism. A Cre-dependent viral vector encoding a Gq-coupled human M3 muscarinic receptor (hM3Dq) was delivered into the LC of Phox2b-Cre mice. The hM3Dq-transduced neurons were pharmacologically activated while respiratory function was measured by plethysmography. We demonstrated that selective stimulation of Phox2bLC neurons significantly increased basal ventilation in conscious mice. Genetic ablation of these neurons markedly impaired hypercapnic ventilatory responses. Moreover, stimulation of Phox2bLC neurons enhanced the activity of preBötzinger complex neurons. Finally, axons of Phox2bLC neurons projected to the preBötzinger complex. Collectively, Phox2bLC neurons contribute to the control of breathing most likely via an LC-preBötzinger complex circuit.
Collapse
Affiliation(s)
- Na Liu
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China.,Department of Physiology, Cangzhou Medical College, Cangzhou, 061000, China
| | - Congrui Fu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050000, China
| | - Hongxiao Yu
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yakun Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Shi
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yinchao Hao
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Fang Yuan
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiangjian Zhang
- Hebei Key laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, China
| | - Sheng Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
34
|
Venner A, Todd WD, Fraigne J, Bowrey H, Eban-Rothschild A, Kaur S, Anaclet C. Newly identified sleep-wake and circadian circuits as potential therapeutic targets. Sleep 2020; 42:5306564. [PMID: 30722061 DOI: 10.1093/sleep/zsz023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
Optogenetics and chemogenetics are powerful tools, allowing the specific activation or inhibition of targeted neuronal subpopulations. Application of these techniques to sleep and circadian research has resulted in the unveiling of several neuronal populations that are involved in sleep-wake control, and allowed a comprehensive interrogation of the circuitry through which these nodes are coordinated to orchestrate the sleep-wake cycle. In this review, we discuss six recently described sleep-wake and circadian circuits that show promise as therapeutic targets for sleep medicine. The parafacial zone (PZ) and the ventral tegmental area (VTA) are potential druggable targets for the treatment of insomnia. The brainstem circuit underlying rapid eye movement sleep behavior disorder (RBD) offers new possibilities for treating RBD and neurodegenerative synucleinopathies, whereas the parabrachial nucleus, as a nexus linking arousal state control and breathing, is a promising target for developing treatments for sleep apnea. Therapies that act upon the hypothalamic circuitry underlying the circadian regulation of aggression or the photic regulation of arousal and mood pathway carry enormous potential for helping to reduce the socioeconomic burden of neuropsychiatric and neurodegenerative disorders on society. Intriguingly, the development of chemogenetics as a therapeutic strategy is now well underway and such an approach has the capacity to lead to more focused and less invasive therapies for treating sleep-wake disorders and related comorbidities.
Collapse
Affiliation(s)
- Anne Venner
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA.,Department of Neurology, Harvard Medical School, Boston, MA
| | - William D Todd
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA.,Department of Neurology, Harvard Medical School, Boston, MA
| | - Jimmy Fraigne
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Hannah Bowrey
- Department of Psychiatry, Rutgers Biomedical Health Sciences, Rutgers University, Newark, NJ.,Save Sight Institute, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Satvinder Kaur
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA.,Department of Neurology, Harvard Medical School, Boston, MA
| | - Christelle Anaclet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, NeuroNexus Institute, Graduate Program in Neuroscience - Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
35
|
Vicente MC, Humphrey CM, Gargaglioni LH, Ostrowski TD. Decreased excitability of locus coeruleus neurons during hypercapnia is exaggerated in the streptozotocin-model of Alzheimer's disease. Exp Neurol 2020; 328:113250. [PMID: 32088169 DOI: 10.1016/j.expneurol.2020.113250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/04/2020] [Accepted: 02/17/2020] [Indexed: 10/25/2022]
Abstract
The locus coeruleus (LC) is a pontine nucleus important for respiratory control and central chemoreception. It is affected in Alzheimer's disease (AD) and alteration of LC cell function may account for respiratory problems observed in AD patients. In the current study, we tested the electrophysiological properties and CO2/pH sensitivity of LC neurons in a model for AD. Sporadic AD was induced in rats by intracerebroventricular injection of 2 mg/kg streptozotocin (STZ), which induces behavioral and molecular impairments found in AD. LC neurons were recorded using the patch clamp technique and tested for responses to CO2 (10% CO2, pH = 7.0). The majority (~60%) of noradrenergic LC neurons in adult rats were inhibited by CO2 exposure as indicated by a significant decrease in action potential (AP) discharge to step depolarizations. The STZ-AD rat model had a greater sensitivity to CO2 than controls. The increased CO2-sensitivity was demonstrated by a significantly stronger inhibition of activity during hypercapnia that was in part due to hyperpolarization of the resting membrane potential. Reduction of AP discharge in both groups was generally accompanied by lower LC network activity, depolarized AP threshold, increased AP repolarization, and increased current through a subpopulation of voltage-gated K+ channels (KV). The latter was indicated by enhanced transient KV currents particularly in the STZ-AD group. Interestingly, steady-state KV currents were reduced under hypercapnia, a change that would favor enhanced AP discharge. However, the collective response of most LC neurons in adult rats, and particularly those in the STZ-AD group, was inhibited by CO2.
Collapse
Affiliation(s)
- Mariane C Vicente
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA; Department of Animal Morphology and Physiology, Sao Paulo State University-UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Chuma M Humphrey
- Department of Biology, Truman State University, Kirksville, MO, USA
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Tim D Ostrowski
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA.
| |
Collapse
|
36
|
Postnatal changes in O2 and CO2 sensitivity in rodents. Respir Physiol Neurobiol 2020; 272:103313. [DOI: 10.1016/j.resp.2019.103313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/31/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023]
|
37
|
Opris I, Dai X, Johnson DMG, Sanchez FJ, Villamil LM, Xie S, Lee-Hauser CR, Chang S, Jordan LM, Noga BR. Activation of Brainstem Neurons During Mesencephalic Locomotor Region-Evoked Locomotion in the Cat. Front Syst Neurosci 2019; 13:69. [PMID: 31798423 PMCID: PMC6868058 DOI: 10.3389/fnsys.2019.00069] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/31/2019] [Indexed: 12/26/2022] Open
Abstract
The distribution of locomotor-activated neurons in the brainstem of the cat was studied by c-Fos immunohistochemistry in combination with antibody-based cellular phenotyping following electrical stimulation of the mesencephalic locomotor region (MLR) – the anatomical constituents of which remain debated today, primarily between the cuneiform (CnF) and the pedunculopontine tegmental nuclei (PPT). Effective MLR sites were co-extensive with the CnF nucleus. Animals subject to the locomotor task showed abundant Fos labeling in the CnF, parabrachial nuclei of the subcuneiform region, periaqueductal gray, locus ceruleus (LC)/subceruleus (SubC), Kölliker–Fuse, magnocellular and lateral tegmental fields, raphe, and the parapyramidal region. Labeled neurons were more abundant on the side of stimulation. In some animals, Fos-labeled cells were also observed in the ventral tegmental area, medial and intermediate vestibular nuclei, dorsal motor nucleus of the vagus, n. tractus solitarii, and retrofacial nucleus in the ventrolateral medulla. Many neurons in the reticular formation were innervated by serotonergic fibers. Numerous locomotor-activated neurons in the parabrachial nuclei and LC/SubC/Kölliker–Fuse were noradrenergic. Few cholinergic neurons within the PPT stained for Fos. In the medulla, serotonergic neurons within the parapyramidal region and the nucleus raphe magnus were positive for Fos. Control animals, not subject to locomotion, showed few Fos-labeled neurons in these areas. The current study provides positive evidence for a role for the CnF in the initiation of locomotion while providing little evidence for the participation of the PPT. The results also show that MLR-evoked locomotion involves the parallel activation of reticular and monoaminergic neurons in the pons/medulla, and provides the anatomical and functional basis for spinal monoamine release during evoked locomotion. Lastly, the results indicate that vestibular, cardiovascular, and respiratory centers are centrally activated during MLR-evoked locomotion. Altogether, the results show a complex pattern of neuromodulatory influences of brainstem neurons by electrical activation of the MLR.
Collapse
Affiliation(s)
- Ioan Opris
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xiaohong Dai
- Department of Physiology, Spinal Cord Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Dawn M G Johnson
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Francisco J Sanchez
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Luz M Villamil
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Songtao Xie
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Cecelia R Lee-Hauser
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Stephano Chang
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Larry M Jordan
- Department of Physiology, Spinal Cord Research Centre, University of Manitoba, Winnipeg, MB, Canada
| | - Brian R Noga
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
38
|
Magalhães KS, de Britto AA, Paton JFR, Moraes DJA. A6 neurons simultaneously modulate active expiration and upper airway resistance in rats. Exp Physiol 2019; 105:53-64. [PMID: 31675759 DOI: 10.1113/ep088164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022]
Abstract
NEW FINDINGS What is the central question of this study? Do A6 neurons modulate active expiratory and airway responses evoked by hypercapnia/acidosis? What is the main finding and its importance? Acute inhibition of A6 neurons reduced active expiratory, inspiratory and the associated oropharyngeal and laryngeal motor responses to hypercapnia/acidosis. A6 neurons provide excitatory synaptic drive contributing to the central generation of inspiratory and expiratory motor activity as well as the control of upper airway resistance. ABSTRACT During rest, inspiration is an active phenomenon, whereas expiration is passive. Under conditions of high chemical drive, such as hypercapnia/acidosis, there is an increase in inspiratory activity, expiration becomes active and upper airway resistance is reduced. The locus coeruleus noradrenergic neurons (A6 neurons) are activated when exposed to elevated CO2 /[H+ ] levels and modulate respiratory brainstem neurons regulating ventilation. However, the role of A6 neurons in the control of upper airway resistance is not fully understood. We tested the hypothesis that A6 neurons contribute to the central generation of active inspiratory and expiratory responses and the associated changes in the motor nerves controlling upper airway resistance during hypercapnia/acidosis in rats. Using a perfused brainstem-spinal cord preparation, we inhibited A6 neurons using pharmacogenetics and evaluated the active expiratory (abdominal nerve), laryngeal (cervical vagus nerve), oropharyngeal (hypoglossal nerve) and inspiratory (phrenic nerve) motor nerve responses to hypercapnia/acidosis. Acute inhibition of A6 neurons did not produce significant changes in the respiratory pattern in normocapnia. However, the hypercapnia/acidosis-induced active expiratory response and the associated changes in the motor nerves responsible for control of oropharyngeal and laryngeal resistance, as well as the inspiratory response were all reduced after inhibition of A6 neurons. Our data demonstrate that A6 neurons exert an important excitatory synaptic drive to the central generation of both active inspiratory and expiratory activities and modulate the control of upper airway resistance during hypercapnia/acidosis.
Collapse
Affiliation(s)
- Karolyne S Magalhães
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Alan A de Britto
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Julian F R Paton
- Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Davi J A Moraes
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
39
|
Burgraff NJ, Neumueller SE, Buchholz KJ, LeClaire J, Hodges MR, Pan L, Forster HV. Brainstem serotonergic, catecholaminergic, and inflammatory adaptations during chronic hypercapnia in goats. FASEB J 2019; 33:14491-14505. [PMID: 31670983 DOI: 10.1096/fj.201901288rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite the prevalence of CO2 retention in human disease, little is known about the adaptive neurobiological effects of chronic hypercapnia. We have recently shown 30-d exposure to increased inspired CO2 (InCO2) leads to a steady-state ventilation that exceeds the level predicted by the sustained acidosis and the acute CO2/H+ chemoreflex, suggesting plasticity within respiratory control centers. Based on data showing brainstem changes in aminergic and inflammatory signaling during carotid body denervation-induced hypercapnia, we hypothesized chronic hypercapnia per se will lead to similar changes. We found that: 1) increased InCO2 increased IL-1β in the medullary raphe (MR), ventral respiratory column, and cuneate nucleus after 24 h, but not after 30 d of hypercapnia; 2) the number of serotonergic and total neurons were reduced within the MR and ventrolateral medulla following 30 d of increased InCO2; 3) markers of tryptophan metabolism were altered following 24 h, but not 30 d of InCO2; and 4) there were few changes in brainstem amine levels following 24 h or 30 d of increased InCO2. We conclude that these changes may contribute to initiating or maintaining respiratory neuroplasticity during chronic hypercapnia but alone do not account for ventilatory acclimatization to chronic increased InCO2.-Burgraff, N. J., Neumueller, S. E., Buchholz, K. J., LeClaire, J., Hodges, M. R., Pan, L., Forster, H. V. Brainstem serotonergic, catecholaminergic, and inflammatory adaptations during chronic hypercapnia in goats.
Collapse
Affiliation(s)
- Nicholas J Burgraff
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Suzanne E Neumueller
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Kirstyn J Buchholz
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - John LeClaire
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA.,Neuroscience Research Center Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Lawrence Pan
- Department of Physical Therapy, Marquette University, Milwaukee, Wisconsin, USA
| | - Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA.,Neuroscience Research Center Medical College of Wisconsin, Wauwatosa, Wisconsin, USA.,Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
40
|
Souza GMPR, Kanbar R, Stornetta DS, Abbott SBG, Stornetta RL, Guyenet PG. Breathing regulation and blood gas homeostasis after near complete lesions of the retrotrapezoid nucleus in adult rats. J Physiol 2019; 596:2521-2545. [PMID: 29667182 DOI: 10.1113/jp275866] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS The retrotrapezoid nucleus (RTN) drives breathing proportionally to brain PCO2 but its role during various states of vigilance needs clarification. Under normoxia, RTN lesions increased the arterial PCO2 set-point, lowered the PO2 set-point and reduced alveolar ventilation relative to CO2 production. Tidal volume was reduced and breathing frequency increased to a comparable degree during wake, slow-wave sleep and REM sleep. RTN lesions did not produce apnoeas or disordered breathing during sleep. RTN lesions in rats virtually eliminated the central respiratory chemoreflex (CRC) while preserving the cardiorespiratory responses to hypoxia; the relationship between CRC and number of surviving RTN Nmb neurons was an inverse exponential. The CRC does not function without the RTN. In the quasi-complete absence of the RTN and CRC, alveolar ventilation is reduced despite an increased drive to breathe from the carotid bodies. ABSTRACT The retrotrapezoid nucleus (RTN) is one of several CNS nuclei that contribute, in various capacities (e.g. CO2 detection, neuronal modulation) to the central respiratory chemoreflex (CRC). Here we test how important the RTN is to PCO2 homeostasis and breathing during sleep or wake. RTN Nmb-positive neurons were killed with targeted microinjections of substance P-saporin conjugate in adult rats. Under normoxia, rats with large RTN lesions (92 ± 4% cell loss) had normal blood pressure and arterial pH but were hypoxic (-8 mmHg PaO2 ) and hypercapnic (+10 mmHg ). In resting conditions, minute volume (VE ) was normal but breathing frequency (fR ) was elevated and tidal volume (VT ) reduced. Resting O2 consumption and CO2 production were normal. The hypercapnic ventilatory reflex in 65% FiO2 had an inverse exponential relationship with the number of surviving RTN neurons and was decreased by up to 92%. The hypoxic ventilatory reflex (HVR; FiO2 21-10%) persisted after RTN lesions, hypoxia-induced sighing was normal and hypoxia-induced hypotension was reduced. In rats with RTN lesions, breathing was lowest during slow-wave sleep, especially under hyperoxia, but apnoeas and sleep-disordered breathing were not observed. In conclusion, near complete RTN destruction in rats virtually eliminates the CRC but the HVR persists and sighing and the state dependence of breathing are unchanged. Under normoxia, RTN lesions cause no change in VE but alveolar ventilation is reduced by at least 21%, probably because of increased physiological dead volume. RTN lesions do not cause sleep apnoea during slow-wave sleep, even under hyperoxia.
Collapse
Affiliation(s)
- George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Roy Kanbar
- Department of Pharmaceutical Sciences, Lebanese American University, Beyrouth, Lebanon
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
41
|
Sex differences in breathing. Comp Biochem Physiol A Mol Integr Physiol 2019; 238:110543. [PMID: 31445081 DOI: 10.1016/j.cbpa.2019.110543] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/15/2023]
Abstract
Breathing is a vital behavior that ensures both the adequate supply of oxygen and the elimination of CO2, and it is influenced by many factors. Despite that most of the studies in respiratory physiology rely heavily on male subjects, there is much evidence to suggest that sex is an important factor in the respiratory control system, including the susceptibility for some diseases. These different respiratory responses in males and females may be related to the actions of sex hormones, especially in adulthood. These hormones affect neuromodulatory systems that influence the central medullary rhythm/pontine pattern generator and integrator, sensory inputs to the integrator and motor output to the respiratory muscles. In this article, we will first review the sex dependence on the prevalence of some respiratory-related diseases. Then, we will discuss the role of sex and gonadal hormones in respiratory control under resting conditions and during respiratory challenges, such as hypoxia and hypercapnia, and whether hormonal fluctuations during the estrous/menstrual cycle affect breathing control. We will then discuss the role of the locus coeruleus, a sexually dimorphic CO2/pH-chemosensitive nucleus, on breathing regulation in males and females. Next, we will highlight the studies that exist regarding sex differences in respiratory control during development. Finally, the few existing studies regarding the influence of sex on breathing control in non-mammalian vertebrates will be discussed.
Collapse
|
42
|
Martinez VK, Saldana-Morales F, Sun JJ, Zhu PJ, Costa-Mattioli M, Ray RS. Off-Target Effects of Clozapine-N-Oxide on the Chemosensory Reflex Are Masked by High Stress Levels. Front Physiol 2019; 10:521. [PMID: 31178741 PMCID: PMC6538678 DOI: 10.3389/fphys.2019.00521] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/11/2019] [Indexed: 12/28/2022] Open
Abstract
Respiratory chemosensory circuits are implicated in several physiological and behavioral disorders ranging from sudden infant death syndrome to panic disorder. Thus, a comprehensive map of the chemosensory network would be of significant value. To delineate chemosensory neuronal populations, we have utilized pharmacogenetic Designer Receptors Exclusively Activated by Designer Drugs (DREADD) perturbations for acute neuronal perturbations in respiratory circuit mapping. Recent studies show that the biologically inert DREADD ligand clozapine-N-oxide (CNO) is back-metabolized into the bioactive compound clozapine in rodents, emphasizing the need for CNO-only DREADD-free controls, which have been carried out in several studies. However, we show that high CNO doses used in several chemosensory circuit mapping studies nonetheless affect the chemosensory ventilatory reflexes in control mice, which is unmasked by extensive habituation. Here, unhabituated control animals showed no differences in respiratory parameters after CNO administration, whereas habituated animals receiving the commonly used dose of 10 mg/kg of CNO show a deficit in the hypercapnic (high CO2) chemosensory reflex, which is not present in 1 mg/kg CNO treated or saline control groups. Our findings indicate that even in appropriately controlled studies, additional masked CNO off-target effects may exist and underscore the importance of using minimal doses of activating ligand in combination with high levels of habituation.
Collapse
Affiliation(s)
- Vena K Martinez
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, United States.,Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Fatima Saldana-Morales
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Jenny J Sun
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Ping Jun Zhu
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Mauro Costa-Mattioli
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Russell S Ray
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,McNair Medical Institute, Houston, TX, United States
| |
Collapse
|
43
|
Magalhães KS, Spiller PF, da Silva MP, Kuntze LB, Paton JFR, Machado BH, Moraes DJA. Locus Coeruleus as a vigilance centre for active inspiration and expiration in rats. Sci Rep 2018; 8:15654. [PMID: 30353035 PMCID: PMC6199338 DOI: 10.1038/s41598-018-34047-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/08/2018] [Indexed: 01/05/2023] Open
Abstract
At rest, inspiration is an active process while expiration is passive. However, high chemical drive (hypercapnia or hypoxia) activates central and peripheral chemoreceptors triggering reflex increases in inspiration and active expiration. The Locus Coeruleus contains noradrenergic neurons (A6 neurons) that increase their firing frequency when exposed to hypercapnia and hypoxia. Using recently developed neuronal hyperpolarising technology in conscious rats, we tested the hypothesis that A6 neurons are a part of a vigilance centre for controlling breathing under high chemical drive and that this includes recruitment of active inspiration and expiration in readiness for flight or fight. Pharmacogenetic inhibition of A6 neurons was without effect on resting and on peripheral chemoreceptors-evoked inspiratory, expiratory and ventilatory responses. On the other hand, the number of sighs evoked by systemic hypoxia was reduced. In the absence of peripheral chemoreceptors, inhibition of A6 neurons during hypercapnia did not affect sighing, but reduced both the magnitude and incidence of active expiration, and the frequency and amplitude of inspiration. These changes reduced pulmonary ventilation. Our data indicated that A6 neurons exert a CO2-dependent modulation of expiratory drive. The data also demonstrate that A6 neurons contribute to the CO2-evoked increases in the inspiratory motor output and hypoxia-evoked sighing.
Collapse
Affiliation(s)
- Karolyne S Magalhães
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Pedro F Spiller
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Melina P da Silva
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luciana B Kuntze
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Julian F R Paton
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK.,Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Benedito H Machado
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Davi J A Moraes
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
44
|
Vicente MC, Almeida MC, Bícego KC, Carrettiero DC, Gargaglioni LH. Hypercapnic and Hypoxic Respiratory Response During Wakefulness and Sleep in a Streptozotocin Model of Alzheimer's Disease in Rats. J Alzheimers Dis 2018; 65:1159-1174. [PMID: 30124447 DOI: 10.3233/jad-180397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Besides the typical cognitive decline, patients with Alzheimer's disease (AD) develop disorders of the respiratory system, such as sleep apnea, shortness of breath, and arrhythmias. These symptoms are aggravated with the progression of the disease. However, the cause and nature of these disturbances are not well understood. Here, we treated animals with intracerebroventricular streptozotocin (STZ, 2 mg/kg), a drug that has been described to cause Alzheimer-like behavioral and histopathological impairments. We measured ventilation (V̇E), electroencephalography, and electromyography during normocapnia, hypercapnia, and hypoxia in Wistar rats. In addition, we performed western blot analyses for phosphorylated tau, total tau, and amyloid-β (Aβ) peptide in the locus coeruleus (LC), retrotrapezoid nucleus, medullary raphe, pre-Bötzinger/Bötzinger complex, and hippocampus, and evaluated memory and learning acquisition using the Barnes maze. STZ treatment promoted memory and learning deficits and increased the percentage of total wakefulness during normocapnia and hypercapnia due to a reduction in the length of episodes of wakefulness. CO2-drive to breathe during wakefulness was increased by 26% in STZ-treated rats due to an enhanced tidal volume, but no changes in V̇E were observed in room air or hypoxic conditions. The STZ group also showed a 70% increase of Aβ in the LC and no change in tau protein phosphorylation. In addition, no alteration in body temperature was observed. Our findings suggest that AD animals present an increased sensitivity to CO2 during wakefulness, enhanced Aβ in the LC, and sleep disruption.
Collapse
Affiliation(s)
- Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University-UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Maria C Almeida
- Center for Natural and Human Sciences; Universidade Federal do ABC (UFABC); São Bernardo do Campo, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University-UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Daniel C Carrettiero
- Center for Natural and Human Sciences; Universidade Federal do ABC (UFABC); São Bernardo do Campo, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-UNESP/FCAV at Jaboticabal, SP, Brazil
| |
Collapse
|
45
|
Orexinergic neurons are involved in the chemosensory control of breathing during the dark phase in a Parkinson's disease model. Exp Neurol 2018; 309:107-118. [PMID: 30110606 DOI: 10.1016/j.expneurol.2018.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/02/2018] [Accepted: 08/11/2018] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by loss of dopaminergic neurons in the substantia nigra compacta (SNpc) and the only risk factor is aging. We showed that in 6-hydroxydopamine (6-OHDA)-model of PD there is a reduction in the neuronal profile within the brainstem ventral respiratory column with a decrease in the hypercapnic ventilatory response. Here we tested the involvement of orexin cells from the lateral hypothalamus/perifornical area (LH/PeF) on breathing in a 6-OHDA PD model. In this model of PD, there is a reduction in the total number of orexinergic neurons and in the number of orexinergic neurons that project to the RTN, without changing the number of CO2-activated orexinergic neurons during the dark phase. The ventilation at rest and in response to hypercapnia (7% CO2) was assessed in animals that received 6-OHDA or vehicle injections into the striatum and saporin anti-Orexin-B or IgG saporin into the LH/PeF during the sleep and awake states. The experiments showed a reduction of respiratory frequency (fR) at rest during the light phase in PD animals only during sleep. During the dark phase, there was an impaired fR response to hypercapnia in PD animals with depletion of orexinergic neurons in awake and sleeping rats. In conclusion, the degeneration of orexinergic neurons in this model of PD can be related to impaired chemoreceptor function in the dark phase.
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This article outlines the fundamental brain mechanisms that control sleep-wake patterns and reviews how pathologic changes in these control mechanisms contribute to common sleep disorders. RECENT FINDINGS Discrete but interconnected clusters of cells located within the brainstem and hypothalamus comprise the circuits that generate wakefulness, non-rapid eye movement (non-REM) sleep, and REM sleep. These clusters of cells use specific neurotransmitters, or collections of neurotransmitters, to inhibit or excite their respective sleep- and wake-promoting target sites. These excitatory and inhibitory connections modulate not only the presence of wakefulness or sleep, but also the levels of arousal within those states, including the depth of sleep, degree of vigilance, and motor activity. Dysfunction or degeneration of wake- and sleep-promoting circuits is associated with narcolepsy, REM sleep behavior disorder, and age-related sleep disturbances. SUMMARY Research has made significant headway in identifying the brain circuits that control wakefulness, non-REM, and REM sleep and has led to a deeper understanding of common sleep disorders and disturbances.
Collapse
|
47
|
Reed MD, Iceman KE, Harris MB, Taylor BE. The rostral medulla of bullfrog tadpoles contains critical lung rhythmogenic and chemosensitive regions across metamorphosis. Comp Biochem Physiol A Mol Integr Physiol 2018; 225:7-15. [PMID: 29890210 DOI: 10.1016/j.cbpa.2018.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/14/2018] [Accepted: 05/30/2018] [Indexed: 11/18/2022]
Abstract
The development of amphibian breathing provides insight into vertebrate respiratory control mechanisms. Neural oscillators in the rostral and caudal medulla drive ventilation in amphibians, and previous reports describe ventilatory oscillators and CO2 sensitive regions arise during different stages of amphibian metamorphosis. However, inconsistent findings have been enigmatic, and make comparisons to potential mammalian counterparts challenging. In the current study we assessed amphibian central CO2 responsiveness and respiratory rhythm generation during two different developmental stages. Whole-nerve recordings of respiratory burst activity in cranial and spinal nerves were made from intact or transected brainstems isolated from tadpoles during early or late stages of metamorphosis. Brainstems were transected at the level of the trigeminal nerve, removing rostral structures including the nucleus isthmi, midbrain, and locus coeruleus, or transected at the level of the glossopharyngeal nerve, removing the putative buccal oscillator and caudal medulla. Removal of caudal structures stimulated the frequency of lung ventilatory bursts and revealed a hypercapnic response in normally unresponsive preparations derived from early stage tadpoles. In preparations derived from late stage tadpoles, removal of rostral or caudal structures reduced lung burst frequency, while CO2 responsiveness was retained. Our results illustrate that structures within the rostral medulla are capable of sensing CO2 throughout metamorphic development. Similarly, the region controlling lung ventilation appears to be contained in the rostral medulla throughout metamorphosis. This work offers insight into the consistency of rhythmic respiratory and chemosensitive capacities during metamorphosis.
Collapse
Affiliation(s)
- Mitchell D Reed
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK 99775, United States.
| | - Kimberly E Iceman
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK 99775, United States; Department of Biology, Valparaiso University, Valparaiso, IN 46383, United States
| | - Michael B Harris
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK 99775, United States; Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775, United States; Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States
| | - Barbara E Taylor
- Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK 99775, United States; Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775, United States; Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, United States
| |
Collapse
|
48
|
The Role of Ca 2+ and BK Channels of Locus Coeruleus (LC) Neurons as a Brake to the CO 2 Chemosensitivity Response of Rats. Neuroscience 2018; 381:59-78. [PMID: 29698749 DOI: 10.1016/j.neuroscience.2018.03.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
Abstract
The cellular mechanisms by which LC neurons respond to hypercapnia are usually attributed to an "accelerator" whereby hypercapnic acidosis causes an inhibition of K+ channels or activation of Na+ and Ca+2 channels to depolarize CO2-sensitive neurons. Nevertheless, it is still unknown if this "accelerator" mechanism could be controlled by a brake phenomenon. Whole-cell patch clamping, fluorescence imaging microscopy and plethysmography were used to study the chemosensitive response of the LC neurons. Hypercapnic acidosis activates L-type Ca2+ channels and large conductance Ca-activated K+ (BK) channels, which function as a "brake" on the chemosensitive response of LC neurons. Our findings indicate that both Ca2+ and BK currents develop over the first 2 weeks of postnatal life in rat LC slices and that this brake pathway may cause the developmental decrease in the chemosensitive firing rate response of LC neurons to hypercapnic acidosis. Inhibition of this brake by paxilline (BK channel inhibitor) returns the magnitude of the chemosensitive firing rate response from LC neurons in rats older than P10 to high values similar to those in LC neurons from younger rats. Inhibition of BK channels in LC neurons by bilateral injections of paxilline into the LC results in a significant increase in the hypercapnic ventilatory response of adult rats. Our findings indicate that a BK channel-based braking system helps to determine the chemosensitive respiratory drive of LC neurons and contributes to the hypercapnic ventilatory response. Perhaps, abnormalities of this braking system could result in hypercapnia-induced respiratory disorders and panic responses.
Collapse
|
49
|
Melnychuk MC, Dockree PM, O'Connell RG, Murphy PR, Balsters JH, Robertson IH. Coupling of respiration and attention via the locus coeruleus: Effects of meditation and pranayama. Psychophysiology 2018; 55:e13091. [PMID: 29682753 DOI: 10.1111/psyp.13091] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 03/06/2018] [Accepted: 03/13/2018] [Indexed: 01/26/2023]
Abstract
The locus coeruleus (LC) has established functions in both attention and respiration. Good attentional performance requires optimal levels of tonic LC activity, and must be matched to task consistently. LC neurons are chemosensitive, causing respiratory phrenic nerve firing to increase frequency with higher CO2 levels, and as CO2 level varies with the phase of respiration, tonic LC activity should exhibit fluctuations at respiratory frequency. Top-down modulation of tonic LC activity from brain areas involved in attentional regulation, intended to optimize LC firing to suit task requirements, may have respiratory consequences as well, as increases in LC activity influence phrenic nerve firing. We hypothesize that, due to the physiological and functional overlaps of attentional and respiratory functions of the LC, this small neuromodulatory nucleus is ideally situated to act as a mechanism of synchronization between respiratory and attentional systems, giving rise to a low-amplitude oscillation that enables attentional flexibility, but may also contribute to unintended destabilization of attention. Meditative and pranayama practices result in attentional, emotional, and physiological enhancements that may be partially due to the LC's pivotal role as the nexus in this coupled system. We present original findings of synchronization between respiration and LC activity (via fMRI and pupil dilation) and provide evidence of a relationship between respiratory phase modulation and attentional performance. We also present a mathematical dynamical systems model of respiratory-LC-attentional coupling, review candidate neurophysiological mechanisms of changes in coupling dynamics, and discuss implications for attentional theory, meditation, and pranayama, and possible therapeutic applications.
Collapse
Affiliation(s)
| | - Paul M Dockree
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | - Peter R Murphy
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joshua H Balsters
- Department of Psychology, Royal Holloway University of London, Egham, United Kingdom
| | - Ian H Robertson
- Institute of Neuroscience and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
50
|
Patrone LGA, Biancardi V, Marques DA, Bícego KC, Gargaglioni LH. Brainstem catecholaminergic neurones and breathing control during postnatal development in male and female rats. J Physiol 2018; 596:3299-3325. [PMID: 29479699 DOI: 10.1113/jp275731] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/21/2018] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS The brainstem catecholaminergic (CA) modulation on ventilation changes with development. We determined the role of the brainstem CA system in ventilatory control under normocapnic and hypercapnic conditions during different phases of development [postnatal day (P)7-8, P14-15 and P20-21] in male and female Wistar rats. Brainstem CA neurones produce a tonic inhibitory drive that affects breathing frequency in P7-8 rats and provide an inhibitory drive during hypercapnic conditions in both males and females at P7-8 and P14-15. In pre-pubertal rats, brainstem CA neurones become excitatory for the CO2 ventilatory response in males but remain inhibitory in females. Diseases such as sudden infant death syndrome, congenital central hypoventilation syndrome and Rett syndrome have been associated with abnormalities in the functioning of CA neurones; therefore, the results of the present study contribute to a better understanding of this system. ABSTRACT The respiratory network undergoes significant development during the postnatal phase, including the maturation of the catecholaminergic (CA) system. However, postnatal development of this network and its effect on the control of pulmonary ventilation ( V̇E ) is not fully understood. We investigated the involvement of brainstem CA neurones in respiratory control during postnatal development [postnatal day (P)7-8, P14-15 and P20-21], in male and female rats, through chemical injury with conjugated saporin anti-dopamine β-hydroxylase (DβH-SAP). Thus, DβH-SAP (420 ng μL-1 ), saporin (SAP) or phosphate buffered solution (PBS) was injected into the fourth ventricle of neonatal Wistar rats of both sexes. V̇E and oxygen consumption were recorded 1 week after the injections in unanaesthetized neonatal and juvenile rats during room air and hypercapnia. The resting ventilation was higher in both male and female P7-8 lesioned rats by 33%, with a decrease in respiratory variability being observed in males. The hypercapnic ventilatory response (HCVR) was altered in male and female lesioned rats at all postnatal ages. At P7-8, the HCVR for males and females was increased by 37% and 30%, respectively. For both sexes at P14-15 rats, the increase in V̇E during hypercapnia was 37% higher for lesioned rats. A sex-specific difference in HCRV was observed at P20-21, with lesioned males showing a 33% decrease, and lesioned females showing an increase of 33%. We conclude that brainstem CA neurones exert a tonic inhibitory effect on V̇E in the early postnatal days of the life of a rat, increase variability in P7-8 males and modulate HCRV during the postnatal phase.
Collapse
Affiliation(s)
- Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Vivian Biancardi
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| |
Collapse
|