1
|
Varga AG, Reid BT, Maletz SN, Dossat AM, Levitt ES. Opposing control of the respiratory brainstem on multiple timescales achieved by transmitter co-release from the locus coeruleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639476. [PMID: 40027822 PMCID: PMC11870594 DOI: 10.1101/2025.02.21.639476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The locus coeruleus (LC) provides widespread noradrenergic (NAergic) modulation throughout the brain to influence a wide range of functions, including breathing. Although both anatomical and physiological evidence supports the involvement of the LC in both the upstream integration and the downstream modulation of breathing, the circuitry behind the latter is unknown. Here, we show that NAergic LC neurons send projections to the Kӧlliker-Fuse nucleus (KF), a critical site in the control of breathing. Long duration activation of NAergic LC neuron terminals in pontine slices induces persistent inhibitory and excitatory NA currents or increases firing rate in postsynaptic KF neurons. Short stimulation on the other hand leads to the VGluT2-dependent release of glutamate that may be co-released with NA in a monosynaptic circuit. Together these results demonstrate that LC neurons can exert flexible, opposing effects on different timescales via glutamatergic and NAergic signaling onto a key respiratory brainstem nucleus.
Collapse
|
2
|
Dereli AS, Apaire A, El Tahry R. Sudden Unexpected Death in Epilepsy: Central Respiratory Chemoreception. Int J Mol Sci 2025; 26:1598. [PMID: 40004062 PMCID: PMC11855741 DOI: 10.3390/ijms26041598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a critical concern for individuals suffering from epilepsy, with respiratory dysfunction playing a significant role in its pathology. Fatal seizures are often characterized by central apnea and hypercapnia (elevated CO2 levels), indicating a failure in ventilatory control. Research has shown that both human epilepsy patients and animal models exhibit a reduced hypercapnic ventilatory response in the interictal (non-seizure) period, suggesting an impaired ability to regulate breathing in response to high CO2 levels. This review examines the role of central chemoreceptors-specifically the retrotrapezoid nucleus, raphe nuclei, nucleus tractus solitarius, locus coeruleus, and hypothalamus in this pathology. These structures are critical for sensing CO2 and maintaining respiratory homeostasis. Emerging evidence also implicates neuropeptidergic pathways within these chemoreceptive regions in SUDEP. Neuropeptides like galanin, pituitary adenylate cyclase-activating peptide (PACAP), orexin, somatostatin, and bombesin-like peptides may modulate chemosensitivity and respiratory function, potentially exacerbating respiratory failure during seizures. Understanding the mechanisms linking central chemoreception, respiratory control, and neuropeptidergic signaling is essential to developing targeted interventions to reduce the risk of SUDEP in epilepsy patients.
Collapse
Affiliation(s)
- Ayse S. Dereli
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
| | - Auriane Apaire
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
| | - Riem El Tahry
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
- Center for Refractory Epilepsy, Department of Neurology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
3
|
Baldo BA. Opioid-induced respiratory depression: clinical aspects and pathophysiology of the respiratory network effects. Am J Physiol Lung Cell Mol Physiol 2025; 328:L267-L289. [PMID: 39726397 DOI: 10.1152/ajplung.00314.2024] [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: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Important insights and consensus remain lacking for risk prediction of opioid-induced respiratory depression (OIRD), reversal of respiratory depression (RD), the pathophysiology of OIRD, and which sites make the most significant contribution to its induction. The ventilatory response to inhaled carbon dioxide is the most sensitive biomarker of OIRD. To accurately predict respiratory depression (RD), a multivariant RD prospective trial using continuous capnography and oximetry examining five independent variables, age ≥60, sex, opioid naivety, sleep disorders, and chronic heart failure (PRODIGY trial), were undertaken. Intermittent oximetry alone substantially underestimates the incidence of RD. Naloxone, with an elimination half-life of ∼33 min (cf. morphine 2-3 h; fentanyl and congeners only 5-15 min), has limitations for the rescue of patients with severe OIRD. Buprenorphine is potentially valuable in patients being treated long term since its high µ-receptor (MOR) affinity makes it difficult for an opioid of lower affinity (e.g., fentanyl) to displace it from the receptor. In the last decade, synthetic opioids, for example, fentanyl, its potent analogs such as carfentanil, and the benzimidazole derivative nitazene "superagonists" have contributed to the exponential growth in opioid deaths due to RD. The MOR, encoded by gene Oprm1, is widely expressed in the central and peripheral nervous systems, including centers that modulate breathing. Opioids bind to the receptors, but consensus is lacking on which site(s) makes the most significant contribution to the induction of OIRD. Both the preBötzinger complex (preBötC), the inspiratory rhythm generator, and the Kölliker-Fuse nucleus (KFN), the respiratory modulator, contribute to RD, but receptor binding is not restricted to a single site. Breathing is composed of three phases, inspiration, postinspiration, and active expiration, each generated by distinct rhythm-generating networks: the preBötC, the postinspiratory complex (PiCo), and the lateral parafacial nucleus (pFL), respectively. Somatostatin-expressing mouse cells involved in breathing regulation are not involved in opioid-induced RD.
Collapse
Affiliation(s)
- Brian A Baldo
- Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, Sydney, New South Wales, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Graça SC, Bustelli IB, Santos ÉVD, Fernandes CG, Lanaro R, Stilhano RS, Linardi A, Caetano AL. Banisteriopsis caapi extract: Implications for neuroinflammatory pathways in Locus coeruleus lesion rodent model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118775. [PMID: 39244172 DOI: 10.1016/j.jep.2024.118775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Ayahuasca is a beverage obtained from the decoctions of Banisteriopsis caapi (Spruce ex Griseb.) Morton and Psychotria viridis Ruiz & Pav., used throughout the Amazon as a medicinal beverage for healing and spiritual exploration. The Banisteriopsis caapi extract consists of harmine, harmaline, and tetrahydroharmine (THH); which inhibit the isoforms of monoamine oxidase A and B. In the central nervous system (CNS), it can increase the norepinephrine (NE) concentration, produced in the Locus coeruleus (LC), reducing inflammation that is associated with some neurological disease, such as Parkinson's disease and Alzheimer's disease. AIM OF THE STUDY evaluate the effects of treatment with B. caapi extract on the neuroinflammatory profile in animals with selective LC lesions. MATERIAL AND METHODS male Wistar rats with LC lesions induced by 6-hydroxydopamine were treated with B. caapi extract. Subsequently, behavioral tests were conducted, including the elevated plus maze, rotarod, and open field. Tyrosine hydroxylase positive (TH+) neurons and IBA-1 positive microglia were quantified from the LC inflammatory markers and free radical products were assessed. RESULTS Both 6-Hydroxydopamine hydrochloride and the Banisteriopsis caapi extract causes reduction of LC neurons, at the concentration and frequency used. The LC depletion and the treatment of B. caapi extract interfere with locomotion. B. caapi extract and the LC lesion increased the number and activation of inflammatory cells, such as microglia. B. caapi extract decreases IL-10 in the hippocampus and BDNF gene expression. CONCLUSION This study suggests that B. caapi extract (at the concentration and frequency used) promotes noradrenergic neuron depletion and creates a proinflammatory environment in the CNS.
Collapse
Affiliation(s)
- Santhiago C Graça
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| | - Isabella B Bustelli
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil
| | - Érica V Dos Santos
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| | - Carolina G Fernandes
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| | - Rafael Lanaro
- Faculty of Medical Sciences, State University of Campinas (UNICAMP), 13083-894, Campinas, SP, Brazil.
| | - Roberta S Stilhano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| | - Alessandra Linardi
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| | - Ariadiny L Caetano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences (FCMSCSP), 01221-020, São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Davis MP, DiScala S, Davis A. Respiratory Depression Associated with Opioids: A Narrative Review. Curr Treat Options Oncol 2024; 25:1438-1450. [PMID: 39432171 DOI: 10.1007/s11864-024-01274-5] [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] [Accepted: 10/10/2024] [Indexed: 10/22/2024]
Abstract
OPINION All opioids have a risk of causing respiratory depression and reduced cerebral circulation. Fentanyl has the greatest risk of causing both. This is particularly a concern when combined with illicit opioids such as diamorphine (also known as heroin). Fentanyl should not be used as a frontline potent opioid due its significant risks. Buprenorphine, a schedule III opioid, morphine, or hydromorphone is preferred, followed by oxycodone, which has a significant risk of abuse relative to buprenorphine and morphine. Although all opioids were equally effective in producing analgesia, the relative safety of each opioid is no longer a secondary concern when prescribing. In the face of an international opioid epidemic, clinicians need to choose opioid analgesics safely, wisely, and carefully.
Collapse
Affiliation(s)
| | - Sandra DiScala
- West Palm Beach VA Healthcare System, West Palm Beach, Florida, USA
| | - Amy Davis
- Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
6
|
Mir FA, Jha SK. The Kir channel in the nucleus tractus solitarius integrates the chemosensory system with REM sleep executive machinery for homeostatic balance. Sci Rep 2024; 14:21651. [PMID: 39289431 PMCID: PMC11408532 DOI: 10.1038/s41598-024-71818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
The locus coeruleus (LC), nucleus tractus solitarius (NTS), and retrotrapezoid nucleus (RTN) are critical chemosensory regions in the brainstem. In the LC, acid-sensing ion channels and proton pumps serve as H+ sensors and facilitate the transition from non-rapid eye movement (NREM) to rapid eye movement (REM) sleep. Interestingly, the potassium inward rectifier (KIR) channels in the LC, NTS, and RTN also act as H+-sensors and are a primary target for improving sleep in obstructive sleep apnea and Rett syndrome patients. However, the role of Kir channels in NREM to REM sleep transition for H+ homeostasis is not known. Male Wistar rats were surgically prepared for chronic sleep-wake recording and drug delivery into the LC, NTS, and RTN. In different animal cohorts, microinjections of the Kir channel inhibitor, barium chloride (BaCl2), at concentrations of 1 mM (low dose) and 2 mM (high dose) in the LC and RTN significantly increased wakefulness and decreased NREM sleep. However, BaCl2 microinjection into the LC notably reduced REM sleep, whereas it didn't change in the RTN-injected group. Interestingly, BaCl2 microinjections into the NTS significantly decreased wakefulness and increased the percent amount of NREM and REM sleep. Additionally, with the infusion of BaCl2 into the NTS, the mean REM sleep episode numbers significantly increased, but the length of the REM sleep episode didn't change. These findings suggest that the Kir channels in the NTS, but not in the LC and RTN, modulate state transition from NREM to REM sleep.
Collapse
Affiliation(s)
- Fayaz A Mir
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
- Department of Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Sushil K Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Plini ERG, Melnychuk MC, Andrews R, Boyle R, Whelan R, Spence JS, Chapman SB, Robertson IH, Dockree PM. Greater physical fitness ( VO 2 max ) in healthy older adults associated with increased integrity of the locus coeruleus-noradrenergic system. Acta Physiol (Oxf) 2024; 240:e14191. [PMID: 38895950 PMCID: PMC11250687 DOI: 10.1111/apha.14191] [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: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
AIM Physical activity (PA) is a key component for brain health and Reserve, and it is among the main dementia protective factors. However, the neurobiological mechanisms underpinning Reserve are not fully understood. In this regard, a noradrenergic (NA) theory of cognitive reserve (Robertson, 2013) has proposed that the upregulation of NA system might be a key factor for building reserve and resilience to neurodegeneration because of the neuroprotective role of NA across the brain. PA elicits an enhanced catecholamine response, in particular for NA. By increasing physical commitment, a greater amount of NA is synthetised in response to higher oxygen demand. More physically trained individuals show greater capabilities to carry oxygen resulting in greaterVo 2 max - a measure of oxygen uptake and physical fitness (PF). METHODS We hypothesized that greaterVo 2 max would be related to greater Locus Coeruleus (LC) MRI signal intensity. In a sample of 41 healthy subjects, we performed Voxel-Based Morphometry analyses, then repeated for the other neuromodulators as a control procedure (Serotonin, Dopamine and Acetylcholine). RESULTS As hypothesized, greaterVo 2 max related to greater LC signal intensity, and weaker associations emerged for the other neuromodulators. CONCLUSION This newly established link betweenVo 2 max and LC-NA system offers further understanding of the neurobiology underpinning Reserve in relationship to PA. While this study supports Robertson's theory proposing the upregulation of the NA system as a possible key factor building Reserve, it also provides ground for increasing LC-NA system resilience to neurodegeneration viaVo 2 max enhancement.
Collapse
Affiliation(s)
- Emanuele R G Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Michael C Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Ralph Andrews
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Robert Whelan
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jeffrey S Spence
- Center for BrainHealth, The University of Texas at Dallas, Dallas, Texas, USA
| | - Sandra B Chapman
- Center for BrainHealth, The University of Texas at Dallas, Dallas, Texas, USA
| | - Ian H Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Paul M Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Souza GMPR, Abbott SBG. Loss-of-function of chemoreceptor neurons in the retrotrapezoid nucleus: What have we learned from it? Respir Physiol Neurobiol 2024; 322:104217. [PMID: 38237884 PMCID: PMC10922619 DOI: 10.1016/j.resp.2024.104217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
Central respiratory chemoreceptors are cells in the brain that regulate breathing in relation to arterial pH and PCO2. Neurons located at the retrotrapezoid nucleus (RTN) have been hypothesized to be central chemoreceptors and/or to be part of the neural network that drives the central respiratory chemoreflex. The inhibition or ablation of RTN chemoreceptor neurons has offered important insights into the role of these cells on central respiratory chemoreception and the neural control of breathing over almost 60 years since the original identification of acid-sensitive properties of this ventral medullary site. Here, we discuss the current definition of chemoreceptor neurons in the RTN and describe how this definition has evolved over time. We then summarize the results of studies that use loss-of-function approaches to evaluate the effects of disrupting the function of RTN neurons on respiration. These studies offer evidence that RTN neurons are indispensable for the central respiratory chemoreflex in mammals and exert a tonic drive to breathe at rest. Moreover, RTN has an interdependent relationship with oxygen sensing mechanisms for the maintenance of the neural drive to breathe and blood gas homeostasis. Collectively, RTN neurons are a genetically-defined group of putative central respiratory chemoreceptors that generate CO2-dependent drive that supports eupneic breathing and stimulates the hypercapnic ventilatory reflex.
Collapse
|
13
|
Bustelli IB, Oliveira LM, Correa-Netto NF, Stilhano RS, Caetano AL. Behavioral effects of 6-hydroxydopamine-induced damage to nigro-striatal pathway and Locus coeruleus as a rodent model of Parkinson's disease. Behav Brain Res 2024; 462:114873. [PMID: 38266776 DOI: 10.1016/j.bbr.2024.114873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the Substantia nigra pars compacta (SNpc), which leads to motor and non-motor symptoms (NMS). NMS can appear many years before the classical motor symptoms and are associated with the neurodegeneration of several nuclei; in this work, we highlight the neurodegeneration of Locus coeruleus (LC) in PD. The aim was to investigate the effects of depleting SNpc and LC catecholaminergic neurons on behavioral and neurobiological endpoints. Here we used 6-hydroxydopamine (6-OHDA) in order to induced neurotoxic damage in three independent experimental groups: SNpc lesion group, which 6-OHDA was injected into CPu (CPu-6-OHDA), LC lesion group, which 6-OHDA was injected directly on LC to selectively caused a damage on this nucleus (LC-6-OHDA), and the combined SNpc and LC lesion group (CL-6-OHDA). Next, the behavioral studies were performed using the Morris water maze (MWM), open field (OF), and elevated plus maze (EPM). After stereotaxic surgeries, the animals showed a loss of 67% and 77% of Tyrosine hydroxylase (TH) reactive neurons in the SNpc and LC, respectively. The behavioral analysis showed the anxiety-like behavior in CL-6-OHDA group in the EPM test; in the MWM test, the combined lesions (CL-6-OHDA) showed an impairment in memory acquisition and spatial memory; and no changes were observed in locomotor activity in all the tests. Furthermore, our investigation demonstrating the effects of depleting SN and LC catecholaminergic neurons on behavioral and neurobiological parameters. All these data together lead us to believe that a bilateral PD model including a LC bilateral degeneration is potentially a more accurate model to evaluate the NMS in the pathological development of the disease in rodents.
Collapse
Affiliation(s)
- Isabella B Bustelli
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP 01221-020, Brazil
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Nelson F Correa-Netto
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP 01221-020, Brazil
| | - Roberta S Stilhano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP 01221-020, Brazil
| | - Ariadiny L Caetano
- Department of Physiological Sciences, Santa Casa de São Paulo School of Medical Sciences, São Paulo, SP 01221-020, Brazil.
| |
Collapse
|
14
|
Wrzesień A, Andrzejewski K, Jampolska M, Kaczyńska K. Respiratory Dysfunction in Alzheimer's Disease-Consequence or Underlying Cause? Applying Animal Models to the Study of Respiratory Malfunctions. Int J Mol Sci 2024; 25:2327. [PMID: 38397004 PMCID: PMC10888758 DOI: 10.3390/ijms25042327] [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: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disease that is the most common cause of dementia among the elderly. In addition to dementia, which is the loss of cognitive function, including thinking, remembering, and reasoning, and behavioral abilities, AD patients also experience respiratory disturbances. The most common respiratory problems observed in AD patients are pneumonia, shortness of breath, respiratory muscle weakness, and obstructive sleep apnea (OSA). The latter is considered an outcome of Alzheimer's disease and is suggested to be a causative factor. While this narrative review addresses the bidirectional relationship between obstructive sleep apnea and Alzheimer's disease and reports on existing studies describing the most common respiratory disorders found in patients with Alzheimer's disease, its main purpose is to review all currently available studies using animal models of Alzheimer's disease to study respiratory impairments. These studies on animal models of AD are few in number but are crucial for establishing mechanisms, causation, implementing potential therapies for respiratory disorders, and ultimately applying these findings to clinical practice. This review summarizes what is already known in the context of research on respiratory disorders in animal models, while pointing out directions for future research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (K.A.); (M.J.)
| |
Collapse
|
15
|
Fu M, Xu R, Chen G, Zheng X, Shu B, Huang H, Duan G, Chen Y. Postoperative esketamine improves ventilation after video-assisted thoracoscopic lung resection: A double-blinded randomized controlled trial. Heliyon 2024; 10:e25100. [PMID: 38322862 PMCID: PMC10844121 DOI: 10.1016/j.heliyon.2024.e25100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/08/2024] Open
Abstract
Background Pain management after lung resection plays a crucial role in reducing postoperative pulmonary complications (PPCs). This study aimed to examine the effect of postoperative esketamine infusion as an adjunct to opioid analgesia on ventilation and pulmonary complications in patients underwent lung resection. Methods Patients undergoing video-assisted thoracoscopic lung resection were randomly assigned to either the esketamine group or the control group. The esketamine group received a 24-h infusion of 1.5 mcg/ml sufentanil combined with 0.75 mcg/ml esketamine after surgery, while the control group received 1.5 mcg/ml sufentanil alone. The primary outcome measure was low minute ventilation, and the secondary outcome measures were hypoxemia, PaO2/FiO2 levels, postoperative pulmonary complications, hospital stay duration, ambulation time, Visual Analogue Scale (VAS) score, depression and anxiety levels, sleep quality, and analgesia satisfaction. Results 80 patients were randomly divided into two groups: the esketamine group (n = 40) and the control group (n = 40). The esketamine group exhibited notably reduced incidence of low minute ventilation (P = 0.014), lower occurrence of postoperative pulmonary complications (PPCs) compared to the control group (P = 0.039), and decreased incidence of hypoxemia (P = 0.003). Furthermore, the esketamine group showed improved outcomes with lower VAS scores on the second postoperative day and enhanced sleep quality (P < 0.001) after the surgery. Conclusions Postoperative esketamine infusion with opioids improved ventilation and reduced PPCs after lung resection, warranting further clinical studies. Trial registration This study was registered on ClinicalTrials.gov (Trial ID: NCT05458453, https://clinicaltrials.gov/ct2/show/NCT05458453).
Collapse
Affiliation(s)
| | | | - Guizhen Chen
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xuemei Zheng
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Bin Shu
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - He Huang
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guangyou Duan
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuanjing Chen
- Department of Anaesthesiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Plini ERG, Melnychuk MC, Andrews R, Boyle R, Whelan R, Spence JS, Chapman SB, Robertson IH, Dockree PM. Greater physical fitness (Vo2Max) in healthy older adults associated with increased integrity of the Locus Coeruleus-Noradrenergic system. RESEARCH SQUARE 2023:rs.3.rs-2556690. [PMID: 36798156 PMCID: PMC9934752 DOI: 10.21203/rs.3.rs-2556690/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Physical activity (PA) is a key component for brain health and Reserve, and it is among the main dementia protective factors. However, the neurobiological mechanisms underpinning Reserve are not fully understood. In this regard, a noradrenergic (NA) theory of cognitive reserve (Robertson, 2013) has proposed that the upregulation of NA system might be a key factor for building reserve and resilience to neurodegeneration because of the neuroprotective role of NA across the brain. PA elicits an enhanced catecholamine response, in particular for NA. By increasing physical commitment, a greater amount of NA is synthetised in response to higher oxygen demand. More physically trained individuals show greater capabilities to carry oxygen resulting in greater Vo2max - a measure of oxygen uptake and physical fitness (PF). In the current study, we hypothesised that greater Vo2 max would be related to greater Locus Coeruleus (LC) MRI signal intensity. As hypothesised, greater Vo2max related to greater LC signal intensity across 41 healthy adults (age range 60-72). As a control procedure, in which these analyses were repeated for the other neuromodulators' seeds (for Serotonin, Dopamine and Acetylcholine), weaker associations emerged. This newly established link between Vo2max and LC-NA system offers further understanding of the neurobiology underpinning Reserve in relationship to PA. While this study supports Robertson's theory proposing the upregulation of the noradrenergic system as a possible key factor building Reserve, it also provide grounds for increasing LC-NA system resilience to neurodegeneration via Vo2max enhancement.
Collapse
Affiliation(s)
- Emanuele RG Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Michael C Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Ralph Andrews
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown MA, USA
| | - Robert Whelan
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Jeffrey S. Spence
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Sandra B. Chapman
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Ian H Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown MA, USA
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Paul M Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| |
Collapse
|
17
|
Amaral-Silva L, Santin JM. Molecular profiling of CO 2/pH-sensitive neurons in the locus coeruleus of bullfrogs reveals overlapping noradrenergic and glutamatergic cell identity. Comp Biochem Physiol A Mol Integr Physiol 2023; 283:111453. [PMID: 37230318 PMCID: PMC10492231 DOI: 10.1016/j.cbpa.2023.111453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/03/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Locus coeruleus (LC) neurons regulate breathing by sensing CO2/pH. Neurons within the vertebrate LC are the main source of norepinephrine within the brain. However, they also use glutamate and GABA for fast neurotransmission. Although the amphibian LC is recognized as a site involved in central chemoreception for the control of breathing, the neurotransmitter phenotype of these neurons is unknown. To address this question, we combined electrophysiology and single-cell quantitative PCR to detect mRNA transcripts that define norepinephrinergic, glutamatergic, and GABAergic phenotypes in LC neurons activated by hypercapnic acidosis (HA) in American bullfrogs. Most LC neurons activated by HA had overlapping expression of noradrenergic and glutamatergic markers but did not show strong support for GABAergic transmission. Genes that encode the pH-sensitive K+ channel, TASK2, and acid-sensing cation channel, ASIC2, were most abundant, while Kir5.1 was present in 1/3 of LC neurons. The abundance of transcripts related to norepinephrine biosynthesis linearly correlated with those involved in pH sensing. These results suggest that noradrenergic neurons in the amphibian LC also use glutamate as a neurotransmitter and that CO2/pH sensitivity may be linkedto the noradrenergic cell identity.
Collapse
Affiliation(s)
- Lara Amaral-Silva
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA. https://twitter.com/amaralsilva_l
| | - Joseph M Santin
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
18
|
Jampolska M, Andrzejewski K, Boguszewski PM, Kaczyńska K. L-DOPA Improves Ventilation but Not the Ventilatory Response to Hypercapnia in a Reserpine Model of Parkinson's Disease. Brain Sci 2023; 13:brainsci13050775. [PMID: 37239247 DOI: 10.3390/brainsci13050775] [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: 04/04/2023] [Revised: 04/27/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by progressive degeneration of the substantia nigra that affects mainly movement control. However, pathological changes associated with the development of PD may also alter respiration and can lead to chronic episodes of hypoxia and hypercapnia. The mechanism behind impaired ventilation in PD is unclear. Therefore, in this study, we explore the hypercapnic ventilatory response in a reproducible reserpine-induced (RES) model of PD and parkinsonism. We also investigated how dopamine supplementation with L-DOPA, a classic drug used to treat PD, would affect the breathing and respiratory response to hypercapnia. Reserpine treatment resulted in decreased normocapnic ventilation and behavioral changes manifested as low physical activity and exploratory behavior. The respiratory rate and the minute ventilation response to hypercapnia were significantly higher in sham rats compared to the RES group, while the tidal volume response was lower. All of this appears to be due to reduced baseline ventilation values produced by reserpine. L-DOPA reversed reduced ventilation, indicating a stimulatory effect of DA on breathing, and showed the potency of DA supplementation in restoring normal respiratory activity.
Collapse
Affiliation(s)
- Monika Jampolska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Kryspin Andrzejewski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Paweł M Boguszewski
- Laboratory of Animal Models, Neurobiology Centre, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Ludwika Pasteura 3 St., 02-093 Warsaw, Poland
| | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
19
|
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: 54] [Impact Index Per Article: 27.0] [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
|
20
|
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
|
21
|
Cardoso F, Fávero MT, Veríssimo NV, Furtado Menezes M, Menani JV, de Paula PM. Participation of the angiotensinergic and vasopressinergic mechanisms in the maintenance of cardiorespiratory parameters in sodium depleted rats. Heliyon 2022; 8:e12221. [PMID: 36582730 PMCID: PMC9793166 DOI: 10.1016/j.heliyon.2022.e12221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/26/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Changes in blood volume can be caused by different conditions, such as vomiting, diarrhea, alteration of sodium intake, trauma, or the use of diuretics, which can lead to severe health deterioration. Understanding the mechanisms involved in the maintenance of physiological parameters and the hydroelectrolytic balance of the human body during hypovolemia, can help with preventing and handling these high-risk situations. Hence, this study investigated cardiorespiratory [mean arterial pressure (MAP), heart rate (HR), pulmonary ventilation (VE)] and blood parameters, of sodium depleted rats with furosemide and the roles of the central and peripheral renin-angiotensin and the peripheral vasopressinergic systems in controlling blood pressure in these animals. Different groups under the same conditions received subcutaneous (s.c.) injections of furosemide (diuretic/saliuretic) or vehicle, intracerebroventricular (i.c.v.) or intravenous (i.v.) injections of losartan [angiotensin II (ANG II) AT1 receptor antagonist] or saline, and i.v. injections of Manning compound (AVPX, vasopressin V1 receptor antagonist). Sodium depletion increased the VE (708 ± 71, vs. normovolemic: 478 ± 40 mL/min/kg body wt) and did not modify baseline mean arterial pressure (104 ± 4, vs. normovolemic: 105 ± 4 mmHg) and heart rate (334 ± 20, vs. normovolemic: 379 ± 13 bpm). The i.v. losartan (10 mg/kg of body wt) treatment significantly reduced MAP in all groups and elevated HR, with a greater impact in sodium depleted rats before repletion. On the other hand, the i.c.v. losartan (3.3 μg/kg of body wt) and i.v. AVPX (10 μg/kg of body wt) treatments did not alter the MAP and HR in control, sodium depleted, and sodium repleted rats. These results indicate that sodium depletion affects cardiorespiratory control increasing baseline ventilation and peripheral angiotensinergic mechanisms are relevant for maintaining cardiovascular parameters in sodium depleted rats. Besides, this study suggests vasopressin V1 receptors do not participate in the maintenance of MAP and HR in sodium depleted animals with furosemide.
Collapse
|
22
|
CO 2 exposure enhances Fos expression in hypothalamic neurons in rats during the light and dark phases of the diurnal cycle. Brain Struct Funct 2022; 227:2667-2679. [PMID: 36109371 DOI: 10.1007/s00429-022-02562-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 08/29/2022] [Indexed: 12/30/2022]
Abstract
Orexinergic (OX) neurons in the lateral hypothalamus (LH), perifornical area (PFA) and dorsomedial hypothalamus (DMH) play a role in the hypercapnic ventilatory response, presumably through direct inputs to central pattern generator sites and/or through interactions with other chemosensitive regions. OX neurons can produce and release orexins, excitatory neuropeptides involved in many functions, including physiological responses to changes in CO2/pH. Thus, in the present study, we tested the hypothesis that different nuclei (LH, PFA and DMH) where the orexinergic neurons are located, show distinct activation by CO2 during the light-dark cycle phases. For this purpose, we evaluated the Fos and OXA expression by immunohistochemistry to identify neurons that co-localize Fos + OXA in the LH, LPeF, MPeF and DMH in the light-inactive and dark-active phase in Wistar rats subjected to 3 h of normocapnia or hypercapnia (7% CO2). Quantitative analyses of immunoreactive neurons show that hypercapnia caused an increase in the number of neurons expressing Fos in the LH, LPeF, MPeF and DMH in the light and dark phases. In addition, the number of Fos + OXA neurons increased in the LPeF and DMH independently of the phases of the diurnal cycle; whereas in the MPeF, this increase was observed exclusively in the light phase. Thus, we suggest that OX neurons are selectively activated by hypercapnia throughout the diurnal cycle, reinforcing the differential role of nuclei in the hypothalamus during central chemosensitivity.
Collapse
|
23
|
Maletz SN, Reid BT, Varga AG, Levitt ES. Nucleus Tractus Solitarius Neurons Activated by Hypercapnia and Hypoxia Lack Mu Opioid Receptor Expression. Front Mol Neurosci 2022; 15:932189. [PMID: 35898697 PMCID: PMC9309891 DOI: 10.3389/fnmol.2022.932189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Impaired chemoreflex responses are a central feature of opioid-induced respiratory depression, however, the mechanism through which mu opioid receptor agonists lead to diminished chemoreflexes is not fully understood. One brainstem structure involved in opioid-induced impairment of chemoreflexes is the nucleus of the solitary tract (NTS), which contains a population of neurons that express mu opioid receptors. Here, we tested whether caudal NTS neurons activated during the chemoreflex challenge express mu opioid receptors and overlap with neurons activated by opioids. Using genetic labeling of mu opioid receptor-expressing neurons and cFos immunohistochemistry as a proxy for neuronal activation, we examined the distribution of activated NTS neurons following hypercapnia, hypoxia, and morphine administration. The main finding was that hypoxia and hypercapnia primarily activated NTS neurons that did not express mu opioid receptors. Furthermore, concurrent administration of morphine with hypercapnia induced cFos expression in non-overlapping populations of neurons. Together these results suggest an indirect effect of opioids within the NTS, which could be mediated through mu opioid receptors on afferents and/or inhibitory interneurons.
Collapse
Affiliation(s)
- Sebastian N. Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Brandon T. Reid
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Adrienn G. Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
| | - Erica S. Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
- *Correspondence: Erica S. Levitt ; orcid.org/0000-0002-3634-6594
| |
Collapse
|
24
|
Biancardi V, Patrone LGA, Vicente MC, Marques DA, Bicego KC, Funk GD, Gargaglioni LH. Prenatal fluoxetine has long lasting, differential effects on respiratory control in male and female rats. J Appl Physiol (1985) 2022; 133:371-389. [PMID: 35708704 DOI: 10.1152/japplphysiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Serotonin (5-HT) is an important modulator of brain networks that control breathing. The selective serotonin reuptake inhibitor fluoxetine (FLX) is the first-line antidepressant drug prescribed during pregnancy. We investigated the effects of prenatal FLX on baseline breathing, ventilatory and metabolic responses to hypercapnia and hypoxia as well as number of brainstem 5-HT and tyrosine hydroxylase (TH) neurons of rats during postnatal development (P0-82). Prenatal FLX exposure of males showed a lower baseline that appeared in juveniles and remained in adulthood, with no sleep-wake state dependency. Prenatal FLX exposure of females did not affect baseline breathing. Juvenile male FLX rats showed increased CO2 and hypoxic ventilatory responses, normalizing by adulthood. Alterations in juvenile-FLX treated males were associated with greater number of 5-HT neurons in the ROB and RMAG. Adult FLX-exposed males showed greater number of 5-HT neurons in the RPA and TH neurons in the A5, while reduced number of TH neurons in A7. Prenatal FLX exposure of female rats was associated with greater hyperventilation induced by hypercapnia at P0-2 and juveniles whereas P12-14 and adult FLX (NREM sleep) rats showed an attenuation of the hypercapnic hyperventilation.FLX-exposed females had fewer 5-HT neurons in the RPA and reduced TH A6 density at P0-2; and greater number of TH neurons in the A7 at P12-14. These data indicate that prenatal FLX exposure affects the number of neurons of some monoaminergic regions in the brain and results in long lasting, sex specific changes in baseline breathing pattern and ventilatory responses to respiratory challenges.
Collapse
Affiliation(s)
- Vivian Biancardi
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Kênia C Bicego
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Gregory D Funk
- Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| |
Collapse
|
25
|
Madirazza K, Pecotic R, Pavlinac Dodig I, Valic M, Dogas Z. Blockade of alpha2-adrenergic receptors in the caudal raphe region enhances the renal sympathetic nerve activity response to acute intermittent hypercapnia in rats. Physiol Res 2022; 71:159-169. [PMID: 35043650 DOI: 10.33549/physiolres.934717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The study investigated the role of alpha2-adrenergic receptors of the caudal raphe region in the sympathetic and cardiovascular responses to the acute intermittent hypercapnia (AIHc). Urethane-anesthetized, vagotomized, mechanically ventilated Sprague-Dawley rats (n=38) were exposed to the AIHc protocol (5×3 min, 15 % CO2+50 % O2) in hyperoxic background (50 % O2). alpha2-adrenergic receptor antagonist-yohimbine was applied intravenously (1 mg/kg, n=9) or microinjected into the caudal raphe region (2 mM, n=12) prior to exposure to AIHc. Control groups of animals received saline intravenously (n=7) or into the caudal raphe region (n=10) prior to exposure to AIHc. Renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP) and heart rate (HR) were monitored before exposure to the AIHc protocol (T0), during five hypercapnic episodes (THc1-5) and at 15 min following the end of the last hypercapnic episode (T15). Following intravenous administration of yohimbine, RSNA was significantly greater during THc1-5 and at T15 than in the control group (P<0.05). When yohimbine was microinjected into the caudal raphe region, AIHc elicited greater increases in RSNA during THc1-5 when compared to the controls (THc1: 138.0+/-4.0 % vs. 123.7+/-4.8 %, P=0.032; THc2: 137.1+/-5.0 % vs. 124.1+/-4.5 %, P=0.071; THc3: 143.1+/-6.4 % vs. 122.0±4.8 %, P=0.020; THc4: 146.1+/-6.2 % vs. 120.7+/-5.7 %, P=0.007 and THc5: 143.2+/-7.7 % vs. 119.2+/-7.2 %, P=0.038). During THc1-5, significant decreases in HR from T0 were observed in all groups, while changes in MAP were observed in the group that received yohimbine intravenously. These findings suggest that blockade of the alpha2-adrenegic receptors in the caudal raphe region might have an important role in sympathetic responses to AIHc.
Collapse
Affiliation(s)
- K Madirazza
- Department of Neuroscience, University of Split School of Medicine, Split, Croatia.
| | | | | | | | | |
Collapse
|
26
|
Kaczyńska K, Orłowska ME, Andrzejewski K. Respiratory Abnormalities in Parkinson's Disease: What Do We Know from Studies in Humans and Animal Models? Int J Mol Sci 2022; 23:ijms23073499. [PMID: 35408858 PMCID: PMC8998219 DOI: 10.3390/ijms23073499] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common progressive neurodegenerative disease characterized by movement disorders due to the progressive loss of dopaminergic neurons in the ventrolateral region of the substantia nigra pars compacta (SNpc). Apart from the cardinal motor symptoms such as rigidity and bradykinesia, non-motor symptoms including those associated with respiratory dysfunction are of increasing interest. Not only can they impair the patients’ quality of life but they also can cause aspiration pneumonia, which is the leading cause of death among PD patients. This narrative review attempts to summarize the existing literature on respiratory impairments reported in human studies, as well as what is newly known from studies in animal models of the disease. Discussed are not only respiratory muscle dysfunction, apnea, and dyspnea, but also altered central respiratory control, responses to hypercapnia and hypoxia, and how they are affected by the pharmacological treatment of PD.
Collapse
|
27
|
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: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
28
|
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
|
29
|
Aquino YC, Cabral LM, Miranda NC, Naccarato MC, Falquetto B, Moreira TS, Takakura AC. Respiratory disorders of Parkinson's disease. J Neurophysiol 2022; 127:1-15. [PMID: 34817281 DOI: 10.1152/jn.00363.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra, mainly affecting people over 60 yr of age. Patients develop both classic symptoms (tremors, muscle rigidity, bradykinesia, and postural instability) and nonclassical symptoms (orthostatic hypotension, neuropsychiatric deficiency, sleep disturbances, and respiratory disorders). Thus, patients with PD can have a significantly impaired quality of life, especially when they do not have multimodality therapeutic follow-up. The respiratory alterations associated with this syndrome are the main cause of mortality in PD. They can be classified as peripheral when caused by disorders of the upper airways or muscles involved in breathing and as central when triggered by functional deficits of important neurons located in the brainstem involved in respiratory control. Currently, there is little research describing these disorders, and therefore, there is no well-established knowledge about the subject, making the treatment of patients with respiratory symptoms difficult. In this review, the history of the pathology and data about the respiratory changes in PD obtained thus far will be addressed.
Collapse
Affiliation(s)
- Yasmin C Aquino
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Laís M Cabral
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Nicole C Miranda
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Monique C Naccarato
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Bárbara Falquetto
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
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
|
31
|
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: 10] [Impact Index Per Article: 2.5] [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
|
32
|
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: 45] [Impact Index Per Article: 11.3] [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
|
33
|
Marques DA, Gargaglioni LH, Joseph V, Bretzner F, Bícego KC, Fournier S, Kinkead R. Impact of ovariectomy and CO 2 inhalation on microglia morphology in select brainstem and hypothalamic areas regulating breathing in female rats. Brain Res 2021; 1756:147276. [PMID: 33422531 DOI: 10.1016/j.brainres.2021.147276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 01/02/2021] [Indexed: 11/17/2022]
Abstract
The neural network that regulates breathing shows a significant sexual dimorphism. Ovarian hormones contribute to this distinction as, in rats, ovariectomy reduces the ventilatory response to CO2. Microglia are neuroimmune cells that are sensitive to neuroendocrine changes in their environment. When reacting to challenging conditions, these cells show changes in their morphology that reflect an augmented capacity for producing pro- and anti-inflammatory cytokines. Based on evidence suggesting that microglia contribute to sex-based differences in reflexive responses to hypercapnia, we hypothesized that ovariectomy and hypercapnia promote microglial reactivity in selected brain areas that regulate breathing. We used ionized calcium-binding-adapter molecule-1 (Iba1) immunolabeling to compare the density and morphology of microglia in the locus coeruleus (LC), the caudal medullary raphe, the caudal part of the nucleus of the tractus solitarius (cNTS), and the paraventricular nucleus of the hypothalamus (PVN). Tissue was obtained from SHAM (metaestrus) female rats or following ovariectomy. Rats were exposed to normocapnia or hypercapnia (5% CO2, 20 min). Ovariectomy and hypercapnia did not affect microglial density in any of the structures studied. Ovariectomy promoted a reactive phenotype in the cNTS and LC, as indicated by a larger morphological index. In these structures, hypercapnia had a relatively modest opposing effect; the medullary raphe or the PVN were not affected. We conclude that ovarian hormones attenuate microglial reactivity in CO2/H+ sensing structures. These data suggest that microglia may contribute to neurological diseases in which anomalies of respiratory control are associated with cyclic fluctuations of ovarian hormones or menopause.
Collapse
Affiliation(s)
- Danuzia A Marques
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada.
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Vincent Joseph
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Frédéric Bretzner
- Département de Psychiatrie et Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Stéphanie Fournier
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Université Laval, Québec, QC, Canada
| |
Collapse
|
34
|
Leibold NK, van den Hove DLA, Weidner MT, Buchanan GF, Steinbusch HWM, Lesch KP, Schruers KRJ. Effect of serotonin transporter genotype on carbon dioxide-induced fear-related behavior in mice. J Psychopharmacol 2020; 34:1408-1417. [PMID: 33103571 PMCID: PMC7708670 DOI: 10.1177/0269881120959611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Inhaling 35% carbon dioxide induces an emotional and symptomatic state in humans closely resembling naturally occurring panic attacks, the core symptom of panic disorder. Previous research has suggested a role of the serotonin system in the individual sensitivity to carbon dioxide. In line with this, we previously showed that a variant in the SLC6A4 gene, encoding the serotonin transporter, moderates the fear response to carbon dioxide in humans. To study the etiological basis of carbon dioxide-reactivity and panic attacks in more detail, we recently established a translational mouse model. AIM The purpose of this study was to investigate whether decreased expression of the serotonin transporter affects the sensitivity to carbon dioxide. METHODS Based on our previous work, wildtype and serotonin transporter deficient (+/-, -/-) mice were monitored while being exposed to carbon dioxide-enriched air. In wildtype and serotonin transporter +/- mice, also cardio-respiration was assessed. RESULTS For most behavioral measures under air exposure, wildtype and serotonin transporter +/- mice did not differ, while serotonin transporter -/- mice showed more fear-related behavior. Carbon dioxide exposure evoked a marked increase in fear-related behaviors, independent of genotype, with the exception of time serotonin transporter -/- mice spent in the center zone of the modified open field test and freezing in the two-chamber test. On the physiological level, when inhaling carbon dioxide, the respiratory system was strongly activated and heart rate decreased independent of genotype. CONCLUSION Carbon dioxide is a robust fear-inducing stimulus. It evokes inhibitory behavioral responses such as decreased exploration and is associated with a clear respiratory profile independent of serotonin transporter genotype.
Collapse
Affiliation(s)
- Nicole K Leibold
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,Department of Neurology, Yale School of Medicine, New Haven, USA,Nicole K Leibold, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616 (Vijverdal), 6200 MD Maastricht, The Netherlands.
| | - Daniel LA van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Magdalena T Weidner
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany,Department of Psychiatry and Psychotherapy, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gordon F Buchanan
- Department of Neurology, Yale School of Medicine, New Haven, USA,Department of Neurology, University of Iowa, Iowa City, USA,University of Iowa Graduate College, Iowa City, USA
| | - Harry WM Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technoglogy (DGIST), Daegu, South Korea
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany,Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Koen RJ Schruers
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Department of Psychology, University of Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Leirão IP, Colombari DSA, da Silva GSF, Zoccal DB. Lesion of Serotonergic Afferents to the Retrotrapezoid Nucleus Impairs the Tachypneic Response to Hypercapnia in Unanesthetized Animals. Neuroscience 2020; 452:63-77. [PMID: 33212216 DOI: 10.1016/j.neuroscience.2020.11.005] [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: 04/28/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Hypercapnia promotes an increase in pulmonary ventilation due to the stimulation of brainstem chemosensory cells that are connected to the respiratory network. Among these cells are the raphe serotonergic neurons which widely send projections to distinct central respiratory compartments. Nevertheless, the physiological role of specific raphe serotonergic projections to other chemosensitive sites on the emergence of hypercapnia ventilatory response in vivo still remains to be elucidated. Here we investigated whether the ventilatory response to hypercapnia requires serotonergic inputs to the chemosensitive cells of the retrotrapezoid nucleus (RTN) in the ventrolateral medulla. To test this, pulmonary ventilation was evaluated under baseline conditions and during hypercapnia (7% CO2) in unanesthetized juvenile Holtzman rats (60-90 g) that received bilateral microinjections of either vehicle (control) or anti-SERT-SAP (0.1 mM, 10 pmol/100 nl) toxin in the RTN to retrogradely destroy serotonergic afferents to this region. Fifteen days after microinjections, baseline ventilation was not different between anti-SERT-SAP (n = 8) and control animals (n = 9). In contrast, the ablation of RTN-projecting serotonergic neurons markedly attenuated the hypercapnia-induced increase in respiratory frequency which was correlated with reduced numbers of serotonergic neurons in the raphe obscurus and magnus, but not in the raphe pallidus. The increase in tidal volume during hypercapnia was not significantly affected by anti-SERT-SAP microinjections in the RTN. Our data indicate that serotoninergic neurons that send projections to the RTN region are required for the processing of ventilatory reflex response during exposure to high CO2 in unanesthetized conditions.
Collapse
Affiliation(s)
- Isabela P Leirão
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Débora S A Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Glauber S F da Silva
- Department of Physiology and Biophysics. Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, MG, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil.
| |
Collapse
|
36
|
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: 1.4] [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
|
37
|
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: 1.6] [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
|
38
|
Li SB, de Lecea L. The hypocretin (orexin) system: from a neural circuitry perspective. Neuropharmacology 2020; 167:107993. [PMID: 32135427 DOI: 10.1016/j.neuropharm.2020.107993] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/23/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Shi-Bin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305, USA.
| |
Collapse
|
39
|
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: 1.2] [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]
|
40
|
Depletion of hypothalamic hypocretin/orexin neurons correlates with impaired memory in a Parkinson's disease animal model. Exp Neurol 2020; 323:113110. [DOI: 10.1016/j.expneurol.2019.113110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/18/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022]
|
41
|
Furuya WI, Bassi M, Menani JV, Colombari E, Zoccal DB, Colombari DSA. Modulation of hypercapnic respiratory response by cholinergic transmission in the commissural nucleus of the solitary tract. Pflugers Arch 2019; 472:49-60. [PMID: 31884528 DOI: 10.1007/s00424-019-02341-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/28/2019] [Accepted: 12/11/2019] [Indexed: 01/16/2023]
Abstract
The nucleus of the solitary tract (NTS) is an important area of the brainstem that receives and integrates afferent cardiorespiratory sensorial information, including those from arterial chemoreceptors and baroreceptors. It was described that acetylcholine (ACh) in the commissural subnucleus of the NTS (cNTS) promotes an increase in the phrenic nerve activity (PNA) and antagonism of nicotinic receptors in the same region reduces the magnitude of tachypneic response to peripheral chemoreceptor stimulation, suggesting a functional role of cholinergic transmission within the cNTS in the chemosensory control of respiratory activity. In the present study, we investigated whether cholinergic receptor antagonism in the cNTS modifies the sympathetic and respiratory reflex responses to hypercapnia. Using an arterially perfused in situ preparation of juvenile male Holtzman rats, we found that the nicotinic antagonist (mecamylamine, 5 mM), but not the muscarinic antagonist (atropine, 5 mM), into the cNTS attenuated the hypercapnia-induced increase of hypoglossal activity. Furthermore, mecamylamine in the cNTS potentiated the generation of late-expiratory (late-E) activity in abdominal nerve induced by hypercapnia. None of the cholinergic antagonists microinjected in the cNTS changed either the sympathetic or the phrenic nerve responses to hypercapnia. Our data provide evidence for the role of cholinergic transmission in the cNTS, acting on nicotinic receptors, modulating the hypoglossal and abdominal responses to hypercapnia.
Collapse
Affiliation(s)
- Werner I Furuya
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - José V Menani
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Débora S A Colombari
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil.
| |
Collapse
|
42
|
Dereli AS, Yaseen Z, Carrive P, Kumar NN. Adaptation of Respiratory-Related Brain Regions to Long-Term Hypercapnia: Focus on Neuropeptides in the RTN. Front Neurosci 2019; 13:1343. [PMID: 31920508 PMCID: PMC6923677 DOI: 10.3389/fnins.2019.01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/28/2019] [Indexed: 12/21/2022] Open
Abstract
Long-term hypercapnia is associated with respiratory conditions including obstructive sleep apnea, chronic obstructive pulmonary disease and obesity hypoventilation syndrome. Animal studies have demonstrated an initial (within hours) increase in ventilatory drive followed by a decrease in this response over the long-term (days–weeks) in response hypercapnia. Little is known about whether changes in the central respiratory chemoreflex are involved. Here we investigated whether central respiratory chemoreceptor neurons of the retrotrapezoid nucleus (RTN), which project to the respiratory pattern generator within the ventral respiratory column (VRC) have a role in the mechanism of neuroplasticity associated with long-term hypercapnia. Adult male C57BL/6 mice (n = 5/group) were used. Our aims were (1) to determine if galanin, neuromedin B and gastrin-releasing peptide gene expression is altered in the RTN after long-term hypercapnia. This was achieved using qPCR to measure mRNA expression changes of neuropeptides in the RTN after short-term hypercapnia (6 or 8 h, 5 or 8% CO2) or long-term hypercapnia exposure (10 day, 5 or 8% CO2), (2) in the mouse brainstem, to determine the distribution of preprogalanin in chemoreceptors, and the co-occurrence of the galanin receptor 1 (GalR1:Gi-coupled receptor) with inhibitory GlyT2 ventral respiratory column neurons using in situ hybridization (ISH) to better characterize galaninergic RTN-VRC circuitry, (3) to investigate whether long-term hypercapnia causes changes to recruitment (detected by cFos immunohistochemistry) of respiratory related neural populations including the RTN neurons and their galaninergic subset, in vivo. Collectively, we found that hypercapnia decreases neuropeptide expression in the RTN in the short-term and has the opposite effect over the long-term. Following long term hypercapnia, the number of RTN galanin neurons remains unchanged, and their responsiveness to acute chemoreflex is sustained; in contrast, we identified multiple respiratory related sites that exhibit blunted chemoreflex activation. GalR1 was distributed in 11% of preBötC and 30% of BötC glycinergic neurons. Our working hypothesis is that during long-term hypercapnia, galanin co-release from RTN neurons may counterbalance glutamatergic inputs to respiratory centers to downscale energetically wasteful hyperventilation, thereby having a role in neuroplasticity by contributing to a decrease in ventilation, through the inhibitory effects of galanin.
Collapse
Affiliation(s)
- Ayse Sumeyra Dereli
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Zarwa Yaseen
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Pascal Carrive
- Department of Anatomy, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Natasha N Kumar
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
43
|
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: 0.3] [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
|
44
|
Guyenet PG, Stornetta RL, Souza GMPR, Abbott SBG, Shi Y, Bayliss DA. The Retrotrapezoid Nucleus: Central Chemoreceptor and Regulator of Breathing Automaticity. Trends Neurosci 2019; 42:807-824. [PMID: 31635852 DOI: 10.1016/j.tins.2019.09.002] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022]
Abstract
The ventral surface of the rostral medulla oblongata has been suspected since the 1960s to harbor central respiratory chemoreceptors [i.e., acid-activated neurons that regulate breathing to maintain a constant arterial PCO2 (PaCO2)]. The key neurons, a.k.a. the retrotrapezoid nucleus (RTN), have now been identified. In this review we describe their transcriptome, developmental lineage, and anatomical projections. We also review their contribution to CO2 homeostasis and to the regulation of breathing automaticity during sleep and wake. Finally, we discuss several mechanisms that contribute to the activation of RTN neurons by CO2in vivo: cell-autonomous effects of protons; paracrine effects of pH mediated by surrounding astrocytes and blood vessels; and excitatory inputs from other CO2-responsive CNS neurons.
Collapse
Affiliation(s)
- Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA.
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
45
|
Loiseau C, Casciato A, Barka B, Cayetanot F, Bodineau L. Orexin Neurons Contribute to Central Modulation of Respiratory Drive by Progestins on ex vivo Newborn Rodent Preparations. Front Physiol 2019; 10:1200. [PMID: 31611806 PMCID: PMC6776592 DOI: 10.3389/fphys.2019.01200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/03/2019] [Indexed: 11/13/2022] Open
Abstract
Dysfunction of central respiratory CO2/H+ chemosensitivity is a pivotal factor that elicits deep hypoventilation in patients suffering from central hypoventilation syndromes. No pharmacological treatment is currently available. The progestin desogestrel has been suggested to allow recovery of respiratory response to CO2/H+ in patients suffering from central hypoventilation, but except the fact that supramedullary regions may be involved, mechanisms are still unknown. Here, we tested in neonates whether orexin systems contribute to desogestrel’s central effects on respiratory function. Using isolated ex vivo central nervous system preparations from newborn rats, we show orexin and almorexant, an antagonist of orexin receptors, supressed strengthening of the increase in respiratory frequency induced by prolonged metabolic acidosis under exposure to etonogestrel, the active metabolite of desogestrel. In parallel, almorexant suppressed the increase and enhanced increase in c-fos expression in respiratory-related brainstem structures induced by etonogestrel. These results suggest orexin signalisation is a key component of acidosis reinforcement of respiratory drive by etonogestrel in neonates. Although stage of development used is different as that for progestin clinical observations, presents results provide clues about conditions under which desogestrel or etonogestrel may enhance ventilation in patients suffering from central hypoventilation syndromes.
Collapse
Affiliation(s)
- Camille Loiseau
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Alexis Casciato
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Besma Barka
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Florence Cayetanot
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| | - Laurence Bodineau
- Institut National de la Santé et de la Recherche Médicale, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Sorbonne Université, Paris, France
| |
Collapse
|
46
|
Amygdala rapid kindling impairs breathing in response to chemoreflex activation. Brain Res 2019; 1718:159-168. [DOI: 10.1016/j.brainres.2019.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 03/16/2019] [Accepted: 05/12/2019] [Indexed: 01/10/2023]
|
47
|
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: 6.3] [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
|
48
|
Reed MD, Iceman KE, Harris MB, Taylor BE. Buccal rhythmogenesis and CO 2 sensitivity in Lithobates catesbeianus tadpole brainstems across metamorphosis. Respir Physiol Neurobiol 2019; 268:103251. [PMID: 31279052 DOI: 10.1016/j.resp.2019.103251] [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: 04/07/2019] [Revised: 06/19/2019] [Accepted: 07/02/2019] [Indexed: 11/19/2022]
Abstract
Bullfrog tadpoles ventilate both the buccal cavity and lung. In isolated brainstems, the midbrain/pons influences CO2 responsiveness and timing of lung ventilatory bursting, depending on larval development. However, little is known about midbrain/pons influences on buccal burst patterns. As such, we investigated how removal of this region affects buccal burst shape and CO2 responsiveness across development. We measured facial nerve activity in brainstems isolated from tadpoles during early and late developmental stages, under normal and elevated levels of CO2. Brainstems were either left intact or transected by removing the midbrain/pons. In late stage preparations, buccal burst pattern differed between intact and reduced preparations, and bursts were responsive to elevated CO2 in these reduced preparations. These results suggest the midbrain/pons affects tadpole buccal burst pattern and CO2 responsiveness, perhaps similar to its influences on lung ventilation.
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; Department of Biology, 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; Department of Biology, California State University Long Beach, Long Beach, CA, 90840, United States
| |
Collapse
|
49
|
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: 4.2] [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
|
50
|
Zhuang J, Zang N, Ye C, Xu F. Lethal avian influenza A (H5N1) virus replicates in pontomedullary chemosensitive neurons and depresses hypercapnic ventilatory response in mice. Am J Physiol Lung Cell Mol Physiol 2019; 316:L525-L536. [PMID: 30628490 PMCID: PMC6459289 DOI: 10.1152/ajplung.00324.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/04/2018] [Accepted: 12/29/2018] [Indexed: 01/09/2023] Open
Abstract
The highly pathogenic H5N1 (HK483) viral infection causes a depressed hypercapnic ventilatory response (dHCVR, 20%↓) at 2 days postinfection (dpi) and death at 7 dpi in mice, but the relevant mechanisms are not fully understood. Glomus cells in the carotid body and catecholaminergic neurons in locus coeruleus (LC), neurokinin 1 receptor (NK1R)-expressing neurons in the retrotrapezoid nucleus (RTN), and serotonergic neurons in the raphe are chemosensitive and responsible for HCVR. We asked whether the dHCVR became worse over the infection period with viral replication in these cells/neurons. Mice intranasally inoculated with saline or the HK483 virus were exposed to hypercapnia for 5 min at 0, 2, 4, or 6 dpi, followed by immunohistochemistry to determine the expression of nucleoprotein of H5N1 influenza A (NP) alone and coupled with 1) tyrosine hydroxylase (TH) in the carotid body and LC, 2) NK1R in the RTN, and 3) tryptophan hydroxylase (TPH) in the raphe. HK483 viral infection blunted HCVR by ∼20, 50, and 65% at 2, 4, and 6 dpi. The NP was observed in the pontomedullary respiratory-related nuclei (but not in the carotid body) at 4 and 6 dpi, especially in 20% of RTN NK1R, 35% of LC TH, and ∼10% raphe TPH neurons. The infection significantly reduced the local NK1R or TPH immunoreactivity and population of neurons expressing NK1R or TPH. We conclude that the HK483 virus infects the pontomedullary respiratory nuclei, particularly chemosensitive neurons in the RTN, LC, and raphe, contributing to the severe depression of HCVR and respiratory failure at 6 dpi. NEW & NOTEWORTHY The H5N1 virus infection is lethal due to respiratory failure, but the relevant mechanisms remain unclear. In this study, we demonstrated a gradual diminution of hypercapnic ventilatory response to a degree, leading to respiratory failure over a 6-day infection. Death was associated with viral replication in the pontomedullary respiratory-related nuclei, especially the central chemosensitive neurons. These results not only provide insight into the mechanisms of the lethality of H5N1 viral infection but also offer clues in the development of corresponding treatments to minimize and prevent respiratory failure.
Collapse
Affiliation(s)
- Jianguo Zhuang
- Pathophysiology Program, Lovelace Respiratory Research Institute , Albuquerque, New Mexico
| | - Na Zang
- Pathophysiology Program, Lovelace Respiratory Research Institute , Albuquerque, New Mexico
| | - Chunyan Ye
- Pathophysiology Program, Lovelace Respiratory Research Institute , Albuquerque, New Mexico
| | - Fadi Xu
- Pathophysiology Program, Lovelace Respiratory Research Institute , Albuquerque, New Mexico
| |
Collapse
|