1
|
Pearcey GEP, Afsharipour B, Holobar A, Sandhu MS, Rymer WZ. Acute intermittent hypoxia increases maximal motor unit discharge rates in people with chronic incomplete spinal cord injury. J Physiol 2024; 602:5699-5711. [PMID: 39058666 DOI: 10.1113/jp285049] [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: 05/29/2023] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Acute intermittent hypoxia (AIH) is an emerging technique for enhancing neuroplasticity and motor function in respiratory and limb musculature. Thus far, AIH-induced improvements in strength have been reported for upper and lower limb muscles after chronic incomplete cervical spinal cord injury (iSCI), but the underlying mechanisms have been elusive. We used high-density surface EMG (HDsEMG) to determine if motor unit discharge behaviour is altered after 15 × 60 s exposures to 9% inspired oxygen, interspersed with 21% inspired oxygen (AIH), compared to breathing only 21% air (SHAM). We recorded HDsEMG from the biceps and triceps brachii of seven individuals with iSCI during maximal elbow flexion and extension contractions, and motor unit spike trains were identified using convolutive blind source separation. After AIH, elbow flexion and extension torque increased by 54% and 59% from baseline (P = 0.003), respectively, whereas there was no change after SHAM. Across muscles, motor unit discharge rates increased by ∼4 pulses per second (P = 0.002) during maximal efforts, from before to after AIH. These results suggest that excitability and/or activation of spinal motoneurons is augmented after AIH, providing a mechanism to explain AIH-induced increases in voluntary strength. Pending validation, AIH may be helpful in conjunction with other therapies to enhance rehabilitation outcomes after incomplete spinal cord injury, due to these enhancements in motor unit function and strength. KEY POINTS: Acute intermittent hypoxia (AIH) causes increases in muscular strength and neuroplasticity in people living with chronic incomplete spinal cord injury (SCI), but how it affects motor unit discharge rates is unknown. Motor unit spike times were identified from high-density surface electromyograms during maximal voluntary contractions and tracked from before to after AIH. Motor unit discharge rates were increased following AIH. These findings suggest that AIH can facilitate motoneuron function in people with incomplete SCI.
Collapse
Affiliation(s)
- Gregory E P Pearcey
- School of Human Kinetics and Recreation, and Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
- Department of Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Babak Afsharipour
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Aleš Holobar
- Institute of Computer Science, Faculty of Electrical Engineering and Computer Science, University of Maribor, Maribor, Slovenia
| | - Milap S Sandhu
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Shirley Ryan AbilityLab, Chicago, IL, USA
| | - W Zev Rymer
- Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Shirley Ryan AbilityLab, Chicago, IL, USA
| |
Collapse
|
2
|
Miskowiak KW, Damgaard V, Schandorff JM, Macoveanu J, Knudsen GM, Johansen A, Plaven-Sigray P, Svarer C, Fussing CB, Cramer K, Jørgensen MB, Kessing LV, Ehrenreich H. Effects of cognitive training under hypoxia on cognitive proficiency and neuroplasticity in remitted patients with mood disorders and healthy individuals: ALTIBRAIN study protocol for a randomized controlled trial. Trials 2024; 25:648. [PMID: 39363230 PMCID: PMC11447976 DOI: 10.1186/s13063-024-08463-5] [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: 05/02/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Cognitive impairment is prevalent across neuropsychiatric disorders but there is a lack of treatment strategies with robust, enduring effects. Emerging evidence indicates that altitude-like hypoxia cognition training may induce long-lasting neuroplasticity and improve cognition. We will investigate whether repeated cognition training under normobaric hypoxia can improve cognitive functions in healthy individuals and patients with affective disorders and the neurobiological underpinnings of such effects. METHODS In sub-study 1, 120 healthy participants are randomized to one of four treatment arms in a double-blind manner, allowing for examination of separate and combined effects of three-week repeated moderate hypoxia and cognitive training, respectively. In sub-study 2, 60 remitted patients with major depressive disorder or bipolar disorder are randomized to hypoxia with cognition training or treatment as usual. Assessments of cognition, psychosocial functioning, and quality of life are performed at baseline, end-of-treatment, and at 1-month follow-up. Functional magnetic resonance imaging (fMRI) scans are conducted at baseline and 1-month follow-up, and [11C]UCB-J positron emission tomography (PET) scans are performed at end-of-treatment to quantify the synaptic vesicle glycoprotein 2A (SV2A). The primary outcome is a cognitive composite score of attention, verbal memory, and executive functions. Statistical power of ≥ 80% is reached to detect a clinically relevant between-group difference with minimum n = 26 per treatment arm. Behavioral data are analyzed with an intention-to-treat approach using mixed models. fMRI data is analyzed with the FMRIB Software Library, while PET data is quantified using the simplified reference tissue model (SRTM) with centrum semiovale as reference region. DISCUSSION The results will provide novel insights into whether repeated hypoxia cognition training increases cognition and brain plasticity, which can aid future treatment development strategies. TRIAL REGISTRATION ClinicalTrials.gov, NCT06121206 . Registered on 31 October 2023.
Collapse
Affiliation(s)
- Kamilla Woznica Miskowiak
- NEAD Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Psychiatric Centre Copenhagen, Mental Health Services, Hovedvejen 17, Frederiksberg, Capital Region of Denmark, DK-2000, Denmark.
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, Copenhagen, DK-1353, Denmark.
| | - Viktoria Damgaard
- NEAD Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Psychiatric Centre Copenhagen, Mental Health Services, Hovedvejen 17, Frederiksberg, Capital Region of Denmark, DK-2000, Denmark
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, Copenhagen, DK-1353, Denmark
| | - Johanna Mariegaard Schandorff
- NEAD Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Psychiatric Centre Copenhagen, Mental Health Services, Hovedvejen 17, Frederiksberg, Capital Region of Denmark, DK-2000, Denmark
- Department of Psychology, University of Copenhagen, Øster Farimagsgade 2A, Copenhagen, DK-1353, Denmark
| | - Julian Macoveanu
- NEAD Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Psychiatric Centre Copenhagen, Mental Health Services, Hovedvejen 17, Frederiksberg, Capital Region of Denmark, DK-2000, Denmark
| | - Gitte Moos Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Annette Johansen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pontus Plaven-Sigray
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Claus Svarer
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Caroline Bruun Fussing
- Copenhagen Affective Disorder Research Centre (CADIC), Psychiatric Centre Copenhagen, Psychiatric Centre Copenhagen, Mental Health Services, Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Cramer
- NEAD Centre, Psychiatric Centre Copenhagen, Frederiksberg Hospital, Psychiatric Centre Copenhagen, Mental Health Services, Hovedvejen 17, Frederiksberg, Capital Region of Denmark, DK-2000, Denmark
| | - Martin Balslev Jørgensen
- Copenhagen Affective Disorder Research Centre (CADIC), Psychiatric Centre Copenhagen, Psychiatric Centre Copenhagen, Mental Health Services, Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lars Vedel Kessing
- Copenhagen Affective Disorder Research Centre (CADIC), Psychiatric Centre Copenhagen, Psychiatric Centre Copenhagen, Mental Health Services, Frederiksberg, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max-Planck-Institute of Experimental Medicine, City Campus, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
3
|
Witkin JM, Radin DP, Rana S, Fuller DD, Fusco AF, Demers JC, Pradeep Thakre P, Smith JL, Lippa A, Cerne R. AMPA receptors play an important role in the biological consequences of spinal cord injury: Implications for AMPA receptor modulators for therapeutic benefit. Biochem Pharmacol 2024; 228:116302. [PMID: 38763261 DOI: 10.1016/j.bcp.2024.116302] [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: 03/04/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Spinal cord injury (SCI) afflicts millions of individuals globally. There are few therapies available to patients. Ascending and descending excitatory glutamatergic neural circuits in the central nervous system are disrupted by SCI, making α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) a potential therapeutic drug target. Emerging research in preclinical models highlights the involvement of AMPARs in vital processes following SCI including breathing, pain, inflammation, bladder control, and motor function. However, there are no clinical trial data reported in this patient population to date. No work on the role of AMPA receptors in sexual dysfunction after SCI has been disclosed. Compounds with selective antagonist and potentiating effects on AMPA receptors have benefit in animal models of SCI, with antagonists generally showing protective effects early after injury and potentiators (ampakines) producing improved breathing and bladder function. The role of AMPARs in pathophysiology and recovery after SCI depends upon the time post injury, and the timing of AMPAR augmentation or antagonism. The roles of inflammation, synaptic plasticity, sensitization, neurotrophic factors, and neuroprotection are considered in this context. The data summarized and discussed in this paper document proof of principle and strongly encourage additional studies on AMPARs as novel gateways to therapeutic benefit for patients suffering from SCI. The availability of both AMPAR antagonists such as perampanel and AMPAR allosteric modulators (i.e., ampakines) such as CX1739, that have been safely administered to humans, provides an expedited means of clinical inquiry for possible therapeutic advances.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | | | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Anna F Fusco
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Julie C Demers
- Indiana University/Purdue University, Indianapolis, IN, USA
| | - Prajwal Pradeep Thakre
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, IN, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA; Faculty of Medicine, University of Ljubljana, Zaloška Cesta 4, Ljubljana, Slovenia
| |
Collapse
|
4
|
Smith CM, Salmon OF. Safety and effectiveness of acute intermittent hypoxia during a single treatment at different hypoxic severities. Respir Physiol Neurobiol 2024; 331:104358. [PMID: 39349270 DOI: 10.1016/j.resp.2024.104358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
PURPOSE Examine the cardiovascular, muscular function, cognitive, and neural plastic responses to determine the safety and effectiveness of acute Intermittent hypoxia (AIH) at a low, high, and control fractional inspired oxygen (FiO2) dosage METHODS: Thirteen human participants performed 30-min of AIH in 60-s intervals at FiO2's of 0.21 (AIH21), 0.15 (AIH15), and 0.09 (AIH9). Heart rate variability (root mean squared of successive differences; RMSSD), heart rate, oxygen saturation (SpO2), blood pressure, muscular strength, neuromuscular activation, cerebral hemodynamic responses, cognition, symptomology, and brain-derived neurotrophic factor (BDNF) responses were measured before (Pre-AIH), after (post-AIH), and at 20-min of recovery (Recovery-AIH) RESULTS: There were no differences between AIH protocols for heart rate, RMSSD, blood pressure, or SpO2. Muscular strength improved Post-AIH for AIH15 (10 %) and AIH9 (14 %) and remained elevated (6 %) at Recovery-AIH. Neuromuscular activation increased Pre-AIH to Post-AIH for AIH15 (10 %) and AIH9 (11 %). Cerebral hemodynamic responses were not impacted between conditions. Both AIH15 and AIH9 increased BDNF Post-AIH (62 %) and Recovery-AIH (63 %) CONCLUSION: Acute intermittent hypoxia is generally safe and effective at producing neural plastic responses, but further examination of co-occurring cardiovascular diseases is needed. This study provides safety focused findings which will widen the adoption and refinement of AIH protocols.
Collapse
Affiliation(s)
- Cory M Smith
- Robbins College of Health and Human Sciences, Human & Environmental Physiology Laboratory, Baylor University, Waco, TX, USA.
| | - Owen F Salmon
- Robbins College of Health and Human Sciences, Human & Environmental Physiology Laboratory, Baylor University, Waco, TX, USA
| |
Collapse
|
5
|
Arboit F, Pereira GC, Fialho MFP, Becker G, Brum EDS, Pillat MM, Bochi GV, Portela LOC, Zanchet EM. Dual Approach to Depression: The Combined Efficacy of Intermittent Hypoxia and Fluoxetine in Modulating Behavioral and Inflammatory Responses. Biomedicines 2024; 12:2116. [PMID: 39335629 PMCID: PMC11430548 DOI: 10.3390/biomedicines12092116] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Mental disorders pose a significant public health challenge, affecting millions worldwide. Given the limitations of current therapies, many patients experience inadequate responses and adverse effects. Intermittent hypoxia (IH) has demonstrated anxiolytic, antidepressant, and neuroprotective properties in various protocols. This study investigated the effects of acute IH (13% O2, 1 h), fluoxetine (FLX) and their combination on depression-like behavior, serum corticosterone, and inflammatory cytokine levels induced by acute restraint stress in C57BL/6 female mice. Methods: Behavioral assessments included the tail suspension test, forced swim test, and open field test. Results: The combined IH + FLX treatment exhibited a synergistic effect, reducing immobility time and increasing latency time, respectively, in the tail suspension test (46%, p = 0.0014; 73%, p = 0.0033) and forced swim test (56%, p = 0.0082; 48%, p = 0.0322) compared to the ARS group. Biochemical analysis revealed that individual and combined treatments significantly reduced most inflammatory interleukins by up to 96%. Corticosterone levels were reduced by 30% only in the IH group. Conclusions: These findings highlight the potential of a one-hour IH session, particularly when combined with fluoxetine, to alleviate depressive-like behaviors and exert anti-inflammatory effects, suggesting a promising therapeutic approach for depression.
Collapse
Affiliation(s)
- Francini Arboit
- Center of Health Sciences, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (F.A.); (G.C.P.); (M.M.P.); (G.V.B.)
| | - Gabriele Cheiran Pereira
- Center of Health Sciences, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (F.A.); (G.C.P.); (M.M.P.); (G.V.B.)
| | - Maria Fernanda Pessano Fialho
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (M.F.P.F.); (G.B.); (E.d.S.B.)
| | - Gabriela Becker
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (M.F.P.F.); (G.B.); (E.d.S.B.)
| | - Evelyne da Silva Brum
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (M.F.P.F.); (G.B.); (E.d.S.B.)
| | - Micheli Mainardi Pillat
- Center of Health Sciences, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (F.A.); (G.C.P.); (M.M.P.); (G.V.B.)
| | - Guilherme Vargas Bochi
- Center of Health Sciences, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (F.A.); (G.C.P.); (M.M.P.); (G.V.B.)
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
| | - Luiz Osório Cruz Portela
- Center of Physical Education and Sports, Federal University of Santa Maria, Santa Maria 97105-900, Brazil;
| | - Eliane Maria Zanchet
- Center of Health Sciences, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil; (F.A.); (G.C.P.); (M.M.P.); (G.V.B.)
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
| |
Collapse
|
6
|
Gonzalez-Rothi EJ, Allen LL, Seven YB, Ciesla MC, Holland AE, Santiago JV, Mitchell GS. Prolonged intermittent hypoxia differentially regulates phrenic motor neuron serotonin receptor expression in rats following chronic cervical spinal cord injury. Exp Neurol 2024; 378:114808. [PMID: 38750949 DOI: 10.1016/j.expneurol.2024.114808] [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: 12/22/2023] [Revised: 04/05/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024]
Abstract
Low-dose (< 2 h/day), acute intermittent hypoxia (AIH) elicits multiple forms of serotonin-dependent phrenic motor plasticity and is emerging as a promising therapeutic strategy to restore respiratory and non-respiratory motor function after spinal cord injury (SCI). In contrast, high-dose (> 8 h/day), chronic intermittent hypoxia (CIH) undermines some forms of serotonin-dependent phrenic motor plasticity and elicits pathology. CIH is a hallmark of sleep disordered breathing, which is highly prevalent in individuals with cervical SCI. Interestingly, AIH and CIH preconditioning differentially impact phrenic motor plasticity. Although mechanisms of AIH-induced plasticity in the phrenic motor system are well-described in naïve rats, we know little concerning how these mechanisms are affected by chronic SCI or intermittent hypoxia preconditioning. Thus, in a rat model of chronic, incomplete cervical SCI (lateral spinal hemisection at C2 (C2Hx), we assessed serotonin type 2A, 2B and 7 receptor expression in and near phrenic motor neurons and compared: 1) intact vs. chronically injured rats; and 2) the impact of preconditioning with varied "doses" of intermittent hypoxia (IH). While there were no effects of chronic injury or intermittent hypoxia alone, CIH affected multiple receptors in rats with chronic C2Hx. Specifically, CIH preconditioning (8 h/day; 28 days) increased serotonin 2A and 7 receptor expression exclusively in rats with chronic C2Hx. Understanding the complex, context-specific interactions between chronic SCI and CIH and how this ultimately impacts phrenic motor plasticity is important as we leverage AIH-induced motor plasticity to restore breathing and other non-respiratory motor functions in people with chronic SCI.
Collapse
Affiliation(s)
- Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Latoya L Allen
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Marissa C Ciesla
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashley E Holland
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Juliet V Santiago
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
7
|
Barok R, Grittner JML, Miller S, Dougherty BJ. Sex hormone supplementation improves breathing and restores respiratory neuroplasticity following C2 hemisection in rats. Front Physiol 2024; 15:1390777. [PMID: 38803364 PMCID: PMC11128654 DOI: 10.3389/fphys.2024.1390777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
In addition to loss of sensory and motor function below the level of the lesion, traumatic spinal cord injury (SCI) may reduce circulating steroid hormones that are necessary for maintaining normal physiological function for extended time periods. For men, who comprise nearly 80% of new SCI cases each year, testosterone is the most abundant circulating sex steroid. SCI often results in significantly reduced testosterone production and may result in chronic low testosterone levels. Testosterone plays a role in respiratory function and the expression of respiratory neuroplasticity. When testosterone levels are low, young adult male rats are unable to express phrenic long-term facilitation (pLTF), an inducible form of respiratory neuroplasticity invoked by acute, intermittent hypoxia (AIH). However, testosterone replacement can restore this respiratory neuroplasticity. Complicating the interpretation of this finding is that testosterone may exert its influence in three possible ways: 1) directly through androgen receptor (AR) activation, 2) through conversion to dihydrotestosterone (DHT) by way of the enzyme 5α-reductase, or 3) through conversion to 17β-estradiol (E2) by way of the enzyme aromatase. DHT signals via AR activation similar to testosterone, but with higher affinity, while E2 activates local estrogen receptors. Evidence to date supports the idea that exogenous testosterone supplementation exerts its influence through estrogen receptor signaling under conditions of low circulating testosterone. Here we explored both recovery of breathing function (measured with whole body barometric plethysmography) and the expression of AIH-induced pLTF in male rats following C2-hemisection SCI. One week post injury, rats were supplemented with either E2 or DHT for 7 days. We hypothesized that E2 would enhance ventilation and reveal pLTF following AIH in SCI rats. To our surprise, though E2 did beneficially impact overall breathing recovery following C2-hemisection, both E2 supplementation and DHT restored the expression of AIH-induced pLTF 2 weeks post-SCI.
Collapse
Affiliation(s)
- Rebecca Barok
- Rehabilitation Science Graduate Program, Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Jessica M. L. Grittner
- Rehabilitation Science Graduate Program, Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Shawn Miller
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Brendan J. Dougherty
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
8
|
Jacobs M, Evans E, Ellis C. Exploring the association between social determinants and aphasia impairment: A retrospective data integration approach. PLoS One 2024; 19:e0299979. [PMID: 38512886 PMCID: PMC10956803 DOI: 10.1371/journal.pone.0299979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Traditionally, the study of aphasia focused on brain trauma, clinical biomarkers, and cognitive processes, rarely considering the social determinants of health. This study evaluates the relationship between aphasia impairment and demographic, socioeconomic, and contextual determinants among people with aphasia (PWA). METHODS PWA indexed within AphasiaBank-a database populated by multiple clinical aphasiology centers with standardized protocols characterizing language, neuropsychological functioning, and demographic information-were matched with respondents in the Medical Expenditure Panel Survey based on response year, age, sex, race, ethnicity, time post stroke, and mental health status. Generalized log-linear regression models with bootstrapped standard errors evaluated the association between scores on the Western Aphasia Battery-Revised Aphasia Quotient (WAB-R AQ) and demographic, economic, and contextual characteristics accounting for clustering of respondents and the stratification of data collection. Region, age, and income specific models tested the sensitivity of results. RESULTS PWA over age 60 had 2.4% (SE = 0.020) lower WAB-R AQ scores compared with younger PWA. Compared to White PWA, Black and Hispanic PWA had 4.7% (SE = 0.03) and 0.81% (SE = 0.06) lower WAB-R AQ scores, respectively, as did those and living in the Southern US (-2.2%, SE = 0.03) even after controlling for age, family size, and aphasia type. Those living in larger families (β = 0.005, SE = 0.008), with income over $30,000 (β = 0.017, SE = 0.022), and a college degree (β = 0.030, SE = 0.035) had higher WAB-R AQ relative to their counterparts. Region-specific models showed that racial differences were only significant in the South and Midwest, while ethnic differences are only significant in the West. Sex differences only appeared in age-specific models. Racial and ethnic differences were not significant in the high-income group regression. CONCLUSION These findings support evidence that circumstances in which individuals live, work, and age are significantly associated with their health outcomes including aphasia impairment.
Collapse
Affiliation(s)
- Molly Jacobs
- Department of Health Services Research, Management and Policy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States of America
| | - Elizabeth Evans
- Department of Speech, Language and Hearing Sciences, Communication Equity and Outcomes Laboratory, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States of America
| | - Charles Ellis
- Department of Speech, Language and Hearing Sciences, Communication Equity and Outcomes Laboratory, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States of America
| |
Collapse
|
9
|
George SZ, Horn ME. Acute Intermittent Hypoxia Did Not Alter Pain Sensitivity or Pain Intensity Ratings for Individuals with Chronic Low Back Pain: A Pilot Study. J Pain Res 2024; 17:421-429. [PMID: 38328018 PMCID: PMC10848823 DOI: 10.2147/jpr.s433685] [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: 08/03/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
Aims and Objective The purpose of this pilot study was to explore whether AIH produces changes in pain sensitivity or in reports of self-reported pain intensity for individuals with low back pain. Methods In a quasi-experimental, cross-over design we compared participants (n = 9) exposed to normal room air and hypoxia using a commercially available gas blender. The treatment period consisted of 5 consecutive days of randomly assigned to AIH or room air. For the participants initially randomized to AIH there was cross-over to receive 5 more consecutive days of room air. Therefore, this design allowed for between group and within subject assessment of AIH effects. Pain sensitivity was assessed with quantitative sensory testing (QST) for posterior superior iliac spine pressure threshold, plantar thermal threshold, and peak pain ratings. Self-reported pain intensity for low back pain was assessed via the Brief Pain Inventory. Results There were no between group differences for AIH and room air in pain sensitivity or self-reported pain intensity. In the within subject analyses larger effect sizes favoring AIH were detected for plantar measures of pain sensitivity but not for self-reported pain intensity. Conclusion This study, while presenting null findings, describes an initial step in determining whether AIH can be used to increase pain relief. Based on this pilot study we offer guidance for future research including study design, AIH dosage, participant selection, and using AIH in combination with non-pharmacologic treatments.
Collapse
Affiliation(s)
- Steven Z George
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
- Departments of Orthopaedic Surgery and Population Health Science, Duke University, Durham, NC, USA
| | - Maggie E Horn
- Departments of Orthopaedic Surgery and Population Health Science, Duke University, Durham, NC, USA
| |
Collapse
|
10
|
Miller S, Lopez EJ, Grittner JML, Dougherty BJ. Low level CO 2 supplementation maintains isocapnia and reveals ventilatory long-term facilitation in rats. Respir Physiol Neurobiol 2024; 320:104185. [PMID: 37935342 PMCID: PMC10842720 DOI: 10.1016/j.resp.2023.104185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
Acute, intermittent hypoxia (AIH) induces ventilatory long-term facilitation (vLTF) in awake, freely behaving rats under poikilocapnic and isocapnic experimental conditions. Establishing pre-clinical methods for vLTF induction that more closely align with successful protocols in humans and anesthetized rats would minimize dissonance in experimental findings and improve translational aspects of vLTF. Here, we tested several levels of low-dose CO2 supplementation during and after AIH to determine 1) the lowest amount of inspired CO2 that would maintain isocapnia in rats during a vLTF protocol, and 2) the net impact of supplemental CO2 on vLTF expression. Rats received one of four levels of inspired CO2 (0%, 0.5%, 1% or 2%) administered during AIH and for the 60 min following AIH to quantify vLTF. Our findings indicated that 2% inspired CO2 was sufficient to maintain isocapnia across the AIH protocol and reveal significant vLTF. These findings provide evidence-based support for using 2% supplemental CO2 during and after AIH when assessing vLTF in rats.
Collapse
Affiliation(s)
- Shawn Miller
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Edgar Juarez Lopez
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jessica M L Grittner
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, MN, USA; Graduate Program in Rehabilitation Science, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Brendan J Dougherty
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
11
|
Zhang G, Yang G, Zhou Y, Cao Z, Yin M, Ma L, Fan M, Zhao YQ, Zhu L. Intermittent hypoxia training effectively protects against cognitive decline caused by acute hypoxia exposure. Pflugers Arch 2024; 476:197-210. [PMID: 37994929 DOI: 10.1007/s00424-023-02885-x] [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: 07/16/2023] [Revised: 10/24/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023]
Abstract
Intermittent hypoxia training (IHT) is a promising approach that has been used to induce acclimatization to hypoxia and subsequently lower the risk of developing acute mountain sickness (AMS). However, the effects of IHT on cognitive and cerebrovascular function after acute hypoxia exposure have not been characterized. In the present study, we first confirmed that the simplified IHT paradigm was effective at relieving AMS at 4300 m. Second, we found that IHT improved participants' cognitive and neural alterations when they were exposed to hypoxia. Specifically, impaired working memory performance, decreased conflict control function, impaired cognitive control, and aggravated mental fatigue induced by acute hypoxia exposure were significantly alleviated in the IHT group. Furthermore, a reversal of brain swelling induced by acute hypoxia exposure was visualized in the IHT group using magnetic resonance imaging. An increase in cerebral blood flow (CBF) was observed in multiple brain regions of the IHT group after hypoxia exposure as compared with the control group. Based on these findings, the simplified IHT paradigm might facilitate hypoxia acclimatization, alleviate AMS symptoms, and increase CBF in multiple brain regions, thus ameliorating brain swelling and cognitive dysfunction.
Collapse
Affiliation(s)
- Guangbo Zhang
- Department of Cognition Sciences and Stress Medicine, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, China
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Guochun Yang
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing, China
| | - Yanzhao Zhou
- Department of Cognition Sciences and Stress Medicine, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, China
| | | | - Ming Yin
- The First Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Lin Ma
- The First Medical Center of Chinese, PLA General Hospital, Beijing, China
| | - Ming Fan
- Department of Cognition Sciences and Stress Medicine, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, China
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong-Qi Zhao
- Department of Cognition Sciences and Stress Medicine, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, China.
- Anhui Medical University, Hefei, China.
| | - Lingling Zhu
- Department of Cognition Sciences and Stress Medicine, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.
- Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
Janssen Daalen JM, Koopman WJH, Saris CGJ, Meinders MJ, Thijssen DHJ, Bloem BR. The Hypoxia Response Pathway: A Potential Intervention Target in Parkinson's Disease? Mov Disord 2024; 39:273-293. [PMID: 38140810 DOI: 10.1002/mds.29688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder for which only symptomatic treatments are available. Both preclinical and clinical studies suggest that moderate hypoxia induces evolutionarily conserved adaptive mechanisms that enhance neuronal viability and survival. Therefore, targeting the hypoxia response pathway might provide neuroprotection by ameliorating the deleterious effects of mitochondrial dysfunction and oxidative stress, which underlie neurodegeneration in PD. Here, we review experimental studies regarding the link between PD pathophysiology and neurophysiological adaptations to hypoxia. We highlight the mechanistic differences between the rescuing effects of chronic hypoxia in neurodegeneration and short-term moderate hypoxia to improve neuronal resilience, termed "hypoxic conditioning". Moreover, we interpret these preclinical observations regarding the pharmacological targeting of the hypoxia response pathway. Finally, we discuss controversies with respect to the differential effects of hypoxia response pathway activation across the PD spectrum, as well as intervention dosing in hypoxic conditioning and potential harmful effects of such interventions. We recommend that initial clinical studies in PD should focus on the safety, physiological responses, and mechanisms of hypoxic conditioning, as well as on repurposing of existing pharmacological compounds. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jules M Janssen Daalen
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Christiaan G J Saris
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjan J Meinders
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Thakre PP, Fuller DD. Pattern sensitivity of ampakine-hypoxia interactions for evoking phrenic motor facilitation in anesthetized rat. J Neurophysiol 2024; 131:216-224. [PMID: 38116608 PMCID: PMC11286303 DOI: 10.1152/jn.00315.2023] [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: 08/21/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
Repeated hypoxic episodes can produce a sustained (>60 min) increase in neural drive to the diaphragm. The requirement of repeated hypoxic episodes (vs. a single episode) to produce phrenic motor facilitation (pMF) can be removed by allosteric modulation of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors using ampakines. We hypothesized that the ampakine-hypoxia interaction resulting in pMF requires that ampakine dosing precedes the onset of hypoxia. Phrenic nerve recordings were made from urethane-anesthetized, mechanically ventilated, and vagotomized adult male Sprague-Dawley rats during isocapnic conditions. Ampakine CX717 (15 mg/kg iv) was given immediately before (n = 8), during (n = 8), or immediately after (n = 8) a 5-min hypoxic episode (arterial oxygen partial pressure 40-45 mmHg). Ampakine before hypoxia (Aprior) resulted in a sustained increase in inspiratory phrenic burst amplitude (i.e., pMF) reaching +70 ± 21% above baseline (BL) after 60 min. This was considerably greater than corresponding values in the groups receiving ampakine during hypoxia (+28 ± 47% above BL, P = 0.005 vs. Aprior) or after hypoxia (+23 ± 40% above BL, P = 0.005 vs. Aprior). Phrenic inspiratory burst rate, heart rate, and systolic, diastolic, and mean arterial pressure (mmHg) were similar across the three treatment groups (all P > 0.3, treatment effect). We conclude that the presentation order of ampakine and hypoxia impacts the magnitude of pMF, with ampakine pretreatment evoking the strongest response. Ampakine pretreatment may have value in the context of hypoxia-based neurorehabilitation strategies.NEW & NOTEWORTHY Phrenic motor facilitation (pMF) is evoked after repeated episodes of brief hypoxia. pMF can also be induced when an allosteric modulator of AMPA receptors (ampakine) is intravenously delivered immediately before a single brief hypoxic episode. Here we show that ampakine delivery before hypoxia (vs. during or after hypoxia) evokes the largest pMF with minimal impact on arterial blood pressure and heart rate. Ampakine pretreatment may have value in the context of hypoxia-based neurorehabilitation strategies.
Collapse
Affiliation(s)
- Prajwal P Thakre
- Department of Physical Therapy, University of Florida, Gainesville, Florida, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, United States
- McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
14
|
Wu YK, Wecht JM, Bloom OE, Panza GS, Harel NY. Remote Ischemic conditioning as an emerging tool to improve corticospinal transmission in individuals with chronic spinal cord injury. Curr Opin Neurol 2023; 36:523-530. [PMID: 37865833 DOI: 10.1097/wco.0000000000001216] [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] [Indexed: 10/23/2023]
Abstract
PURPOSE OF REVIEW Remote ischemic conditioning (RIC) involves transient blood flow restriction to one limb leading to systemic tissue-protective effects. RIC shares some potential underlying mechanisms with intermittent hypoxia (IH), in which brief bouts of systemic hypoxia trigger increases in growth factor expression and neural plasticity. RIC has shown promise in acute myocardial infarction and stroke but may be applicable toward chronic neuropathology as well. Consequently, this review discusses similarities and differences between RIC and IH and presents preliminary and ongoing research findings regarding RIC. RECENT FINDINGS Several publications demonstrated that combining RIC with motor training may enhance motor learning in adults with intact nervous systems, though the precise mechanisms were unclear. Our own preliminary data has found that RIC, in conjunction with task specific exercise, can increase corticospinal excitability in a subset of people without neurological injury and in those with chronic cervical spinal cord injury or amyotrophic lateral sclerosis. SUMMARY RIC is a low-cost intervention easy to deliver in a clinical or home setting. Its potential application to facilitate neural plasticity and motor learning during rehabilitation training for individuals with chronic neurological disorders is a novel concept requiring further investigation to characterize mechanisms, safety, and efficacy.
Collapse
Affiliation(s)
- Yu-Kuang Wu
- Icahn School of Medicine at Mount Sinai
- James J. Peters VA Medical Center
| | - Jill M Wecht
- Icahn School of Medicine at Mount Sinai
- James J. Peters VA Medical Center
| | - Ona E Bloom
- James J. Peters VA Medical Center
- The Feinstein Institute for Medical Research
- The Zucker School of Medicine at Hofstra Northwell
| | - Gino S Panza
- The Department of Healthcare Science Program of Occupational Therapy, Wayne State University
- John D. Dingell VA Medical Center, USA
| | - Noam Y Harel
- Icahn School of Medicine at Mount Sinai
- James J. Peters VA Medical Center
| |
Collapse
|
15
|
Bakker ME, Djerourou I, Belanger S, Lesage F, Vanni MP. Alteration of functional connectivity despite preserved cerebral oxygenation during acute hypoxia. Sci Rep 2023; 13:13269. [PMID: 37582847 PMCID: PMC10427674 DOI: 10.1038/s41598-023-40321-3] [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: 04/06/2023] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
Resting state networks (RSN), which show the connectivity in the brain in the absence of any stimuli, are increasingly important to assess brain function. Here, we investigate the changes in RSN as well as the hemodynamic changes during acute, global hypoxia. Mice were imaged at different levels of oxygen (21, 12, 10 and 8%) over the course of 10 weeks, with hypoxia and normoxia acquisitions interspersed. Simultaneous GCaMP and intrinsic optical imaging allowed tracking of both neuronal and hemodynamic changes. During hypoxic conditions, we found a global increase of both HbO and HbR in the brain. The saturation levels of blood dropped after the onset of hypoxia, but surprisingly climbed back to levels similar to baseline within the 10-min hypoxia period. Neuronal activity also showed a peak at the onset of hypoxia, but dropped back to baseline as well. Despite regaining baseline sO2 levels, changes in neuronal RSN were observed. In particular, the connectivity as measured with GCaMP between anterior and posterior parts of the brain decreased. In contrast, when looking at these same connections with HbO measurements, an increase in connectivity in anterior-posterior brain areas was observed suggesting a potential neurovascular decoupling.
Collapse
Affiliation(s)
- Marleen E Bakker
- École d'Optométrie, Université de Montréal, 2500 Chem. De Polytechnique, Montréal, QC, H3T 1J4, Canada.
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, Canada.
| | - Ismaël Djerourou
- École d'Optométrie, Université de Montréal, 2500 Chem. De Polytechnique, Montréal, QC, H3T 1J4, Canada
| | | | - Frédéric Lesage
- Institute of Biomedical Engineering, École Polytechnique de Montréal, Montréal, Canada
- Montréal Heart Institute, Montréal, Canada
| | - Matthieu P Vanni
- École d'Optométrie, Université de Montréal, 2500 Chem. De Polytechnique, Montréal, QC, H3T 1J4, Canada
| |
Collapse
|
16
|
Marciante AB, Seven YB, Kelly MN, Perim RR, Mitchell GS. Magnitude and Mechanism of Phrenic Long-term Facilitation Shift Between Daily Rest Versus Active Phase. FUNCTION 2023; 4:zqad041. [PMID: 37753182 PMCID: PMC10519274 DOI: 10.1093/function/zqad041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 09/28/2023] Open
Abstract
Plasticity is a fundamental property of the neural system controlling breathing. One key example of respiratory motor plasticity is phrenic long-term facilitation (pLTF), a persistent increase in phrenic nerve activity elicited by acute intermittent hypoxia (AIH). pLTF can arise from distinct cell signaling cascades initiated by serotonin versus adenosine receptor activation, respectively, and interact via powerful cross-talk inhibition. Here, we demonstrate that the daily rest/active phase and the duration of hypoxic episodes within an AIH protocol have profound impact on the magnitude and mechanism of pLTF due to shifts in serotonin/adenosine balance. Using the historical "standard" AIH protocol (3, 5-min moderate hypoxic episodes), we demonstrate that pLTF magnitude is unaffected by exposure in the midactive versus midrest phase, yet the mechanism driving pLTF shifts from serotonin-dominant (midrest) to adenosine-dominant (midactive). This mechanistic "flip" results from combined influences of hypoxia-evoked adenosine release and daily fluctuations in basal spinal adenosine. Since AIH evokes less adenosine with shorter (15, 1-min) hypoxic episodes, midrest pLTF is amplified due to diminished adenosine constraint on serotonin-driven plasticity; in contrast, elevated background adenosine during the midactive phase suppresses serotonin-dominant pLTF. These findings demonstrate the importance of the serotonin/adenosine balance in regulating the amplitude and mechanism of AIH-induced pLTF. Since AIH is emerging as a promising therapeutic modality to restore respiratory and nonrespiratory movements in people with spinal cord injury or ALS, knowledge of how time-of-day and hypoxic episode duration impact the serotonin/adenosine balance and the magnitude and mechanism of pLTF has profound biological, experimental, and translational implications.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Mia N Kelly
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Raphael R Perim
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
17
|
Kang J, Lu N, Yang S, Guo B, Zhu Y, Wu S, Huang X, Wong-Riley MTT, Liu YY. Alterations in synapses and mitochondria induced by acute or chronic intermittent hypoxia in the pre-Bötzinger complex of rats: an ultrastructural triple-labeling study with immunocytochemistry and histochemistry. Front Cell Neurosci 2023; 17:1132241. [PMID: 37396926 PMCID: PMC10312010 DOI: 10.3389/fncel.2023.1132241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction The pre-Bötzinger complex (pre-BötC), a kernel of inspiratory rhythmogenesis, is a heterogeneous network with excitatory glutamatergic and inhibitory GABAergic and glycinergic neurons. Inspiratory rhythm generation relies on synchronous activation of glutamatergic neuron, whilst inhibitory neurons play a critical role in shaping the breathing pattern, endowing the rhythm with flexibility in adapting to environmental, metabolic, and behavioral needs. Here we report ultrastructural alterations in excitatory, asymmetric synapses (AS) and inhibitory, symmetric synapses (SS), especially perforated synapses with discontinuous postsynaptic densities (PSDs) in the pre-BötC in rats exposed to daily acute intermittent hypoxia (dAIH) or chronic (C) IH. Methods We utilized for the first time a combination of somatostatin (SST) and neurokinin 1 receptor (NK1R) double immunocytochemistry with cytochrome oxidase histochemistry, to reveal synaptic characteristics and mitochondrial dynamic in the pre-BötC. Results We found perforated synapses with synaptic vesicles accumulated in distinct pools in apposition to each discrete PSD segments. dAIH induced significant increases in the PSD size of macular AS, and the proportion of perforated synapses. AS were predominant in the dAIH group, whereas SS were in a high proportion in the CIH group. dAIH significantly increased SST and NK1R expressions, whereas CIH led to a decrease. Desmosome-like contacts (DLC) were characterized for the first time in the pre-BötC. They were distributed alongside of synapses, especially SS. Mitochondria appeared in more proximity to DLC than synapses, suggestive of a higher energy demand of the DLC. Findings of single spines with dual AS and SS innervation provide morphological evidence of excitation-inhibition interplay within a single spine in the pre-BötC. In particular, we characterized spine-shaft microdomains of concentrated synapses coupled with mitochondrial positioning that could serve as a structural basis for synchrony of spine-shaft communication. Mitochondria were found within spines and ultrastructural features of mitochondrial fusion and fission were depicted for the first time in the pre-BötC. Conclusion We provide ultrastructural evidence of excitation-inhibition synapses in shafts and spines, and DLC in association with synapses that coincide with mitochondrial dynamic in their contribution to respiratory plasticity in the pre-BötC.
Collapse
Affiliation(s)
- Junjun Kang
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Naining Lu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Shoujing Yang
- Department of Pathology, The Fourth Military Medical University, Xi’an, China
| | - Baolin Guo
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Yuanyuan Zhu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Shengxi Wu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Xiaofeng Huang
- Department of Pathology, Xi’an Gaoxin Hospital, Xi’an, China
| | - Margaret T. T. Wong-Riley
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ying-Ying Liu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
18
|
Nair J, Welch JF, Marciante AB, Hou T, Lu Q, Fox EJ, Mitchell GS. APOE4, Age, and Sex Regulate Respiratory Plasticity Elicited by Acute Intermittent Hypercapnic-Hypoxia. FUNCTION 2023; 4:zqad026. [PMID: 37575478 PMCID: PMC10413930 DOI: 10.1093/function/zqad026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 08/15/2023] Open
Abstract
Rationale Acute intermittent hypoxia (AIH) shows promise for enhancing motor recovery in chronic spinal cord injuries and neurodegenerative diseases. However, human trials of AIH have reported significant variability in individual responses. Objectives Identify individual factors (eg, genetics, age, and sex) that determine response magnitude of healthy adults to an optimized AIH protocol, acute intermittent hypercapnic-hypoxia (AIHH). Methods In 17 healthy individuals (age = 27 ± 5 yr), associations between individual factors and changes in the magnitude of AIHH (15, 1-min O2 = 9.5%, CO2 = 5% episodes) induced changes in diaphragm motor-evoked potential (MEP) amplitude and inspiratory mouth occlusion pressures (P0.1) were evaluated. Single nucleotide polymorphisms (SNPs) in genes linked with mechanisms of AIH induced phrenic motor plasticity (BDNF, HTR2A, TPH2, MAOA, NTRK2) and neuronal plasticity (apolipoprotein E, APOE) were tested. Variations in AIHH induced plasticity with age and sex were also analyzed. Additional experiments in humanized (h)ApoE knock-in rats were performed to test causality. Results AIHH-induced changes in diaphragm MEP amplitudes were lower in individuals heterozygous for APOE4 (i.e., APOE3/4) compared to individuals with other APOE genotypes (P = 0.048) and the other tested SNPs. Males exhibited a greater diaphragm MEP enhancement versus females, regardless of age (P = 0.004). Additionally, age was inversely related with change in P0.1 (P = 0.007). In hApoE4 knock-in rats, AIHH-induced phrenic motor plasticity was significantly lower than hApoE3 controls (P < 0.05). Conclusions APOE4 genotype, sex, and age are important biological determinants of AIHH-induced respiratory motor plasticity in healthy adults. Addition to Knowledge Base AIH is a novel rehabilitation strategy to induce functional recovery of respiratory and non-respiratory motor systems in people with chronic spinal cord injury and/or neurodegenerative disease. Figure 5 Since most AIH trials report considerable inter-individual variability in AIH outcomes, we investigated factors that potentially undermine the response to an optimized AIH protocol, AIHH, in healthy humans. We demonstrate that genetics (particularly the lipid transporter, APOE), age and sex are important biological determinants of AIHH-induced respiratory motor plasticity.
Collapse
Affiliation(s)
- Jayakrishnan Nair
- Breathing Research and Therapeutics Center, Department of Physical Therapy, University of Florida, Gainesville, 32603, USA
- Department of Physical Therapy, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Joseph F Welch
- Breathing Research and Therapeutics Center, Department of Physical Therapy, University of Florida, Gainesville, 32603, USA
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, 3- B15 2TT, UK
| | - Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy, University of Florida, Gainesville, 32603, USA
| | - Tingting Hou
- Department of Biostatistics, University of Florida, Gainesville, 32603, USA
| | - Qing Lu
- Department of Biostatistics, University of Florida, Gainesville, 32603, USA
| | - Emily J Fox
- Breathing Research and Therapeutics Center, Department of Physical Therapy, University of Florida, Gainesville, 32603, USA
- Brooks Rehabilitation, Jacksonville, FL, 32216, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy, University of Florida, Gainesville, 32603, USA
| |
Collapse
|
19
|
Michel-Flutot P, Lane MA, Lepore AC, Vinit S. Therapeutic Strategies Targeting Respiratory Recovery after Spinal Cord Injury: From Preclinical Development to Clinical Translation. Cells 2023; 12:1519. [PMID: 37296640 PMCID: PMC10252981 DOI: 10.3390/cells12111519] [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: 04/14/2023] [Revised: 05/15/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
High spinal cord injuries (SCIs) lead to permanent functional deficits, including respiratory dysfunction. Patients living with such conditions often rely on ventilatory assistance to survive, and even those that can be weaned continue to suffer life-threatening impairments. There is currently no treatment for SCI that is capable of providing complete recovery of diaphragm activity and respiratory function. The diaphragm is the main inspiratory muscle, and its activity is controlled by phrenic motoneurons (phMNs) located in the cervical (C3-C5) spinal cord. Preserving and/or restoring phMN activity following a high SCI is essential for achieving voluntary control of breathing. In this review, we will highlight (1) the current knowledge of inflammatory and spontaneous pro-regenerative processes occurring after SCI, (2) key therapeutics developed to date, and (3) how these can be harnessed to drive respiratory recovery following SCIs. These therapeutic approaches are typically first developed and tested in relevant preclinical models, with some of them having been translated into clinical studies. A better understanding of inflammatory and pro-regenerative processes, as well as how they can be therapeutically manipulated, will be the key to achieving optimal functional recovery following SCIs.
Collapse
Affiliation(s)
- Pauline Michel-Flutot
- END-ICAP, UVSQ, Inserm, Université Paris-Saclay, 78000 Versailles, France;
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Michael A. Lane
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA;
| | - Angelo C. Lepore
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Stéphane Vinit
- END-ICAP, UVSQ, Inserm, Université Paris-Saclay, 78000 Versailles, France;
| |
Collapse
|
20
|
Marciante AB, Mitchell GS. Mild inflammation impairs acute intermittent hypoxia-induced phrenic long-term facilitation by a spinal adenosine-dependent mechanism. J Neurophysiol 2023; 129:799-806. [PMID: 36883762 PMCID: PMC10069977 DOI: 10.1152/jn.00035.2023] [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: 01/20/2023] [Revised: 02/23/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Inflammation undermines neuroplasticity, including serotonin-dependent phrenic long-term facilitation (pLTF) following moderate acute intermittent hypoxia (mAIH: 3, 5-min episodes, arterial Po2: 40-50 mmHg; 5-min intervals). Mild inflammation elicited by a low dose of the TLR-4 receptor agonist, lipopolysaccharide (LPS; 100 µg/kg, ip), abolishes mAIH-induced pLTF by unknown mechanisms. In the central nervous system, neuroinflammation primes glia, triggering ATP release and extracellular adenosine accumulation. As spinal adenosine 2 A (A2A) receptor activation impairs mAIH-induced pLTF, we hypothesized that spinal adenosine accumulation and A2A receptor activation are necessary in the mechanism whereby LPS impairs pLTF. We report that 24 h after LPS injection in adult male Sprague Dawley rats: 1) adenosine levels increase in ventral spinal segments containing the phrenic motor nucleus (C3-C5; P = 0.010; n = 7/group) and 2) cervical spinal A2A receptor inhibition (MSX-3, 10 µM, 12 µL intrathecal) rescues mAIH-induced pLTF. In LPS vehicle-treated rats (saline, ip), MSX-3 enhanced pLTF versus controls (LPS: 110 ± 16% baseline; controls: 53 ± 6%; P = 0.002; n = 6/group). In LPS-treated rats, pLTF was abolished as expected (4 ± 6% baseline; n = 6), but intrathecal MSX-3 restored pLTF to levels equivalent to MSX-3-treated control rats (120 ± 14% baseline; P < 0.001; n = 6; vs. LPS controls with MSX-3: P = 0.539). Thus, inflammation abolishes mAIH-induced pLTF by a mechanism that requires increased spinal adenosine levels and A2A receptor activation. As repetitive mAIH is emerging as a treatment to improve breathing and nonrespiratory movements in people with spinal cord injury or ALS, A2A inhibition may offset undermining effects of neuroinflammation associated with these neuromuscular disorders.NEW & NOTEWORTHY Mild inflammation undermines motor plasticity elicited by mAIH. In a model of mAIH-induced respiratory motor plasticity (phrenic long-term facilitation; pLTF), we report that inflammation induced by low-dose lipopolysaccharide undermines mAIH-induced pLTF by a mechanism requiring increased cervical spinal adenosine and adenosine 2 A receptor activation. This finding advances the understanding of mechanisms impairing neuroplasticity, potentially undermining the ability to compensate for the onset of lung/neural injury or to harness mAIH as a therapeutic modality.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy & McKnight Brain Institute, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
21
|
Brumă E, Onose G, Ciobanu V. Research on clinical-paraclinical and evolutive aspects in pati-ents with post spinal cord injury (SCI) statuses and Covid-19 – a systematic literature review. BALNEO AND PRM RESEARCH JOURNAL 2023. [DOI: 10.12680/balneo.2023.538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
The COVID-19 pandemic has generated a lot of interest among doctors as well as scientists around the world. Studies on the impact of the Covid-19 pandemic, including in people with post SCI sufferance, are ongoing, aiming to understand the pathophysiological mechanisms of SARS-CoV2 in target tissues, to optimize related methods of diagnosis and treatment in both, in-itial and later phases of the disease – e.g.: ”long Covid” status – and thus, to make a substantial contribution to the quality of life improvement of the affected patients.
After using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (acronym PRISMA) method to quest for afferent knowledge, it resulted a quite small number (12) of arti-cles, most of them indirectly approaching this topic. Therefore, is important to deepen this niche – which is scarcely approached – in order to find new therapeutic approaches able to combat COVID-19-associated to SCI manifestations – like, for instance, to check whether the principle of intermittent hypoxia treatment is effective and worth to be included in the rehabilitation treat-ment protocols – as neither an indubitable effective drug or vaccine, or respectively, cure for SCI, has been provided so far.
Collapse
Affiliation(s)
- Elena Brumă
- Neuromuscular Rehabilitation Clinic Division – the Teaching Emergency Hospital “Bagdasar-Arseni”, Bucharest, Romania
| | - Gelu Onose
- Faculty of Medicine – the University of Medicine and Pharmacy “Carol Davila”, in Bucharest, Romania
| | - Vlad Ciobanu
- Computer Science Department, ”Politehnica” University of Bucharest, Romania
| |
Collapse
|
22
|
Nair J, Welch JF, Marciante AB, Hou T, Lu Q, Fox EJ, Mitchell GS. APOE4, Age & Sex Regulate Respiratory Plasticity Elicited By Acute Intermittent Hypercapnic-Hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522840. [PMID: 36711653 PMCID: PMC9881941 DOI: 10.1101/2023.01.06.522840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Rationale Acute intermittent hypoxia (AIH) is a promising strategy to induce functional motor recovery following chronic spinal cord injuries and neurodegenerative diseases. Although significant results are obtained, human AIH trials report considerable inter-individual response variability. Objectives Identify individual factors ( e.g. , genetics, age, and sex) that determine response magnitude of healthy adults to an optimized AIH protocol, acute intermittent hypercapnic-hypoxia (AIHH). Methods Associations of individual factors with the magnitude of AIHH (15, 1-min O 2 =9.5%, CO 2 =5% episodes) induced changes in diaphragm motor-evoked potential amplitude (MEP) and inspiratory mouth occlusion pressures (P 0.1 ) were evaluated in 17 healthy individuals (age=27±5 years) compared to Sham. Single nucleotide polymorphisms (SNPs) in genes linked with mechanisms of AIH induced phrenic motor plasticity ( BDNF, HTR 2A , TPH 2 , MAOA, NTRK 2 ) and neuronal plasticity (apolipoprotein E, APOE ) were tested. Variations in AIHH induced plasticity with age and sex were also analyzed. Additional experiments in humanized ( h ) ApoE knock-in rats were performed to test causality. Results AIHH-induced changes in diaphragm MEP amplitudes were lower in individuals heterozygous for APOE 4 ( i.e., APOE 3/4 ) allele versus other APOE genotypes (p=0.048). No significant differences were observed between any other SNPs investigated, notably BDNFval/met ( all p>0.05 ). Males exhibited a greater diaphragm MEP enhancement versus females, regardless of age (p=0.004). Age was inversely related with change in P 0.1 within the limited age range studied (p=0.007). In hApoE 4 knock-in rats, AIHH-induced phrenic motor plasticity was significantly lower than hApoE 3 controls (p<0.05). Conclusions APOE 4 genotype, sex and age are important biological determinants of AIHH-induced respiratory motor plasticity in healthy adults. ADDITION TO KNOWLEDGE BASE Acute intermittent hypoxia (AIH) is a novel rehabilitation strategy to induce functional recovery of respiratory and non-respiratory motor systems in people with chronic spinal cord injury and/or neurodegenerative diseases. Since most AIH trials report considerable inter-individual variability in AIH outcomes, we investigated factors that potentially undermine the response to an optimized AIH protocol, acute intermittent hypercapnic-hypoxia (AIHH), in healthy humans. We demonstrate that genetics (particularly the lipid transporter, APOE ), age and sex are important biological determinants of AIHH-induced respiratory motor plasticity.
Collapse
Affiliation(s)
- Jayakrishnan Nair
- Breathing Research and Therapeutics Center Department of Physical Therapy, University of Florida
- Current address: Department of Physical Therapy, Thomas Jefferson University, PA
| | - Joseph F. Welch
- Breathing Research and Therapeutics Center Department of Physical Therapy, University of Florida
- Current address: School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alexandria B. Marciante
- Breathing Research and Therapeutics Center Department of Physical Therapy, University of Florida
| | - Tingting Hou
- Department of Biostatistics, University of Florida
| | - Qing Lu
- Department of Biostatistics, University of Florida
| | - Emily J. Fox
- Breathing Research and Therapeutics Center Department of Physical Therapy, University of Florida
- Brooks Rehabilitation, Jacksonville, Florida
| | - Gordon S. Mitchell
- Breathing Research and Therapeutics Center Department of Physical Therapy, University of Florida
| |
Collapse
|
23
|
Sales de Campos P, Olsen WL, Wymer JP, Smith BK. Respiratory therapies for Amyotrophic Lateral Sclerosis: A state of the art review. Chron Respir Dis 2023; 20:14799731231175915. [PMID: 37219417 PMCID: PMC10214054 DOI: 10.1177/14799731231175915] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/14/2023] [Indexed: 05/24/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative condition noteworthy for upper and lower motor neuron death. Involvement of respiratory motor neuron pools leads to progressive pathology. These impairments include decreases in neural activation and muscle coordination, progressive airway obstruction, weakened airway defenses, restrictive lung disease, increased risk of pulmonary infections, and weakness and atrophy of respiratory muscles. These neural, airway, pulmonary, and neuromuscular changes deteriorate integrated respiratory-related functions including sleep, cough, swallowing, and breathing. Ultimately, respiratory complications account for a large portion of morbidity and mortality in ALS. This state-of-the-art review highlights applications of respiratory therapies for ALS, including lung volume recruitment, mechanical insufflation-exsufflation, non-invasive ventilation, and respiratory strength training. Therapeutic acute intermittent hypoxia, an emerging therapeutic tool for inducing respiratory plasticity will also be introduced. A focus on emerging evidence and future work underscores the common goal to continue to improve survival for patients living with ALS.
Collapse
Affiliation(s)
- Priscila Sales de Campos
- Breathing Research and Therapeutics
Center, University of Florida, Gainesville, FL, USA
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Wendy L Olsen
- Breathing Research and Therapeutics
Center, University of Florida, Gainesville, FL, USA
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Electrical and
Computer Engineering, University of Florida, Gainesville, FL, USA
| | - James P Wymer
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Barbara K Smith
- Breathing Research and Therapeutics
Center, University of Florida, Gainesville, FL, USA
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
24
|
Seven YB, Allen LL, Ciesla MC, Smith KN, Zwick A, Simon AK, Holland AE, Santiago JV, Stefan K, Ross A, Gonzalez-Rothi EJ, Mitchell GS. Intermittent Hypoxia Differentially Regulates Adenosine Receptors in Phrenic Motor Neurons with Spinal Cord Injury. Neuroscience 2022; 506:38-50. [PMID: 36273657 DOI: 10.1016/j.neuroscience.2022.10.007] [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: 06/28/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
Cervical spinal cord injury (cSCI) impairs neural drive to the respiratory muscles, causing life- threatening complications such as respiratory insufficiency and diminished airway protection. Repetitive "low dose" acute intermittent hypoxia (AIH) is a promising strategy to restore motor function in people with chronic SCI. Conversely, "high dose" chronic intermittent hypoxia (CIH; ∼8 h/night), such as experienced during sleep apnea, causes pathology. Sleep apnea, spinal ischemia, hypoxia and neuroinflammation associated with cSCI increase extracellular adenosine concentrations and activate spinal adenosine receptors which in turn constrains the functional benefits of therapeutic AIH. Adenosine 1 and 2A receptors (A1, A2A) compete to determine net cAMP signaling and likely the tAIH efficacy with chronic cSCI. Since cSCI and intermittent hypoxia may regulate adenosine receptor expression in phrenic motor neurons, we tested the hypotheses that: 1) daily AIH (28 days) downregulates A2A and upregulates A1 receptor expression; 2) CIH (28 days) upregulates A2A and downregulates A1 receptor expression; and 3) cSCI alters the impact of CIH on adenosine receptor expression. Daily AIH had no effect on either adenosine receptor in intact or injured rats. However, CIH exerted complex effects depending on injury status. Whereas CIH increased A1 receptor expression in intact (not injured) rats, it increased A2A receptor expression in spinally injured (not intact) rats. The differential impact of CIH reinforces the concept that the injured spinal cord behaves in distinct ways from intact spinal cords, and that these differences should be considered in the design of experiments and/or new treatments for chronic cSCI.
Collapse
Affiliation(s)
- Yasin B Seven
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Latoya L Allen
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Marissa C Ciesla
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Kristin N Smith
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Amanda Zwick
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Alec K Simon
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashley E Holland
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Juliet V Santiago
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Kelsey Stefan
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashley Ross
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
25
|
Locke KC, Randelman ML, Hoh DJ, Zholudeva LV, Lane MA. Respiratory plasticity following spinal cord injury: perspectives from mouse to man. Neural Regen Res 2022; 17:2141-2148. [PMID: 35259820 PMCID: PMC9083159 DOI: 10.4103/1673-5374.335839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 10/20/2021] [Indexed: 12/03/2022] Open
Abstract
The study of respiratory plasticity in animal models spans decades. At the bench, researchers use an array of techniques aimed at harnessing the power of plasticity within the central nervous system to restore respiration following spinal cord injury. This field of research is highly clinically relevant. People living with cervical spinal cord injury at or above the level of the phrenic motoneuron pool at spinal levels C3-C5 typically have significant impairments in breathing which may require assisted ventilation. Those who are ventilator dependent are at an increased risk of ventilator-associated co-morbidities and have a drastically reduced life expectancy. Pre-clinical research examining respiratory plasticity in animal models has laid the groundwork for clinical trials. Despite how widely researched this injury is in animal models, relatively few treatments have broken through the preclinical barrier. The three goals of this present review are to define plasticity as it pertains to respiratory function post-spinal cord injury, discuss plasticity models of spinal cord injury used in research, and explore the shift from preclinical to clinical research. By investigating current targets of respiratory plasticity research, we hope to illuminate preclinical work that can influence future clinical investigations and the advancement of treatments for spinal cord injury.
Collapse
Affiliation(s)
- Katherine C. Locke
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Margo L. Randelman
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Daniel J. Hoh
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Lyandysha V. Zholudeva
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
- Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Michael A. Lane
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| |
Collapse
|
26
|
Yu JJ, Non AL, Heinrich EC, Gu W, Alcock J, Moya EA, Lawrence ES, Tift MS, O'Brien KA, Storz JF, Signore AV, Khudyakov JI, Milsom WK, Wilson SM, Beall CM, Villafuerte FC, Stobdan T, Julian CG, Moore LG, Fuster MM, Stokes JA, Milner R, West JB, Zhang J, Shyy JY, Childebayeva A, Vázquez-Medina JP, Pham LV, Mesarwi OA, Hall JE, Cheviron ZA, Sieker J, Blood AB, Yuan JX, Scott GR, Rana BK, Ponganis PJ, Malhotra A, Powell FL, Simonson TS. Time Domains of Hypoxia Responses and -Omics Insights. Front Physiol 2022; 13:885295. [PMID: 36035495 PMCID: PMC9400701 DOI: 10.3389/fphys.2022.885295] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The ability to respond rapidly to changes in oxygen tension is critical for many forms of life. Challenges to oxygen homeostasis, specifically in the contexts of evolutionary biology and biomedicine, provide important insights into mechanisms of hypoxia adaptation and tolerance. Here we synthesize findings across varying time domains of hypoxia in terms of oxygen delivery, ranging from early animal to modern human evolution and examine the potential impacts of environmental and clinical challenges through emerging multi-omics approaches. We discuss how diverse animal species have adapted to hypoxic environments, how humans vary in their responses to hypoxia (i.e., in the context of high-altitude exposure, cardiopulmonary disease, and sleep apnea), and how findings from each of these fields inform the other and lead to promising new directions in basic and clinical hypoxia research.
Collapse
Affiliation(s)
- James J. Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Amy L. Non
- Department of Anthropology, Division of Social Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Erica C. Heinrich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Herbert Wertheim School of Public Health and Longevity Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joe Alcock
- Department of Emergency Medicine, University of New Mexico, Albuquerque, MX, United States
| | - Esteban A. Moya
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elijah S. Lawrence
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michael S. Tift
- Department of Biology and Marine Biology, College of Arts and Sciences, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Katie A. O'Brien
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Physiology, Development and Neuroscience, Faculty of Biology, School of Biological Sciences, University of Cambridge, Cambridge, ENG, United Kingdom
| | - Jay F. Storz
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Anthony V. Signore
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Jane I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | | | - Sean M. Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda, CA, United States
| | | | | | | | - Colleen G. Julian
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lorna G. Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Aurora, CO, United States
| | - Mark M. Fuster
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jennifer A. Stokes
- Department of Kinesiology, Southwestern University, Georgetown, TX, United States
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA, United States
| | - John B. West
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jiao Zhang
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - John Y. Shyy
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - Ainash Childebayeva
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - José Pablo Vázquez-Medina
- Department of Integrative Biology, College of Letters and Science, University of California, Berkeley, Berkeley, CA, United States
| | - Luu V. Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - James E. Hall
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Zachary A. Cheviron
- Division of Biological Sciences, College of Humanities and Sciences, University of Montana, Missoula, MT, United States
| | - Jeremy Sieker
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Arlin B. Blood
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jason X. Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Graham R. Scott
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brinda K. Rana
- Moores Cancer Center, UC San Diego, La Jolla, CA, United States
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - Paul J. Ponganis
- Center for Marine Biotechnology and Biomedicine, La Jolla, CA, United States
| | - Atul Malhotra
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Frank L. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
27
|
Effects of acute intermittent hypoxia on corticospinal excitability within the primary motor cortex. Eur J Appl Physiol 2022; 122:2111-2123. [PMID: 35752660 PMCID: PMC9381468 DOI: 10.1007/s00421-022-04982-8] [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: 12/07/2021] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
Purpose Acute intermittent hypoxia (AIH) is a safe and non-invasive treatment approach that uses brief, repetitive periods of breathing reduced oxygen air alternated with normoxia. While AIH is known to affect spinal circuit excitability, the effects of AIH on cortical excitability remain largely unknown. We investigated the effects of AIH on cortical excitability within the primary motor cortex. Methods Eleven healthy, right-handed participants completed two testing sessions: (1) AIH (comprising 3 min in hypoxia [fraction of inspired oxygen ~ 10%] and 2 min in normoxia repeated over five cycles) and (2) normoxia (NOR) (equivalent duration to AIH). Single- and paired-pulse transcranial magnetic stimulations were delivered to the primary motor cortex, before and 0, 25, and 50 min after AIH and normoxia. Results The mean nadir in arterial oxygen saturation was lower (p < 0.001) during the cycles of AIH (82.5 ± 4.9%) than NOR (97.8 ± 0.6%). There was no significant difference in corticospinal excitability, intracortical facilitation, or intracortical inhibition between AIH and normoxia conditions at any time point (all p > 0.05). There was no association between arterial oxygen saturation and changes in corticospinal excitability after AIH (r = 0.05, p = 0.87). Conclusion Overall, AIH did not modify either corticospinal excitability or excitability of intracortical facilitatory and inhibitory circuits within the primary motor cortex. Future research should explore whether a more severe or individualised AIH dose would induce consistent, measurable changes in corticospinal excitability. Supplementary Information The online version contains supplementary material available at 10.1007/s00421-022-04982-8.
Collapse
|
28
|
Rybnikova EA, Nalivaeva NN, Zenko MY, Baranova KA. Intermittent Hypoxic Training as an Effective Tool for Increasing the Adaptive Potential, Endurance and Working Capacity of the Brain. Front Neurosci 2022; 16:941740. [PMID: 35801184 PMCID: PMC9254677 DOI: 10.3389/fnins.2022.941740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
This review is devoted to the phenomenon of intermittent hypoxic training and is aimed at drawing the attention of researchers to the necessity of studying the mechanisms mediating the positive, particularly neuroprotective, effects of hypoxic training at the molecular level. The review briefly describes the historical aspects of studying the beneficial effects of mild hypoxia, as well as the use of hypoxic training in medicine and sports. The physiological mechanisms of hypoxic adaptation, models of hypoxic training and their effectiveness are summarized, giving examples of their beneficial effects in various organs including the brain. The review emphasizes a high, far from being realized at present, potential of hypoxic training in preventive and clinical medicine especially in the area of neurodegeneration and age-related cognitive decline.
Collapse
Affiliation(s)
- Elena A. Rybnikova
- Pavlov Institute Physiology of Russian Academy of Sciences, St. Petersburg, Russia
- *Correspondence: Elena A. Rybnikova,
| | - Natalia N. Nalivaeva
- Pavlov Institute Physiology of Russian Academy of Sciences, St. Petersburg, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, St. Petersburg, Russia
| | - Mikhail Y. Zenko
- Pavlov Institute Physiology of Russian Academy of Sciences, St. Petersburg, Russia
| | - Ksenia A. Baranova
- Pavlov Institute Physiology of Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
29
|
Barnes LA, Mesarwi OA, Sanchez-Azofra A. The Cardiovascular and Metabolic Effects of Chronic Hypoxia in Animal Models: A Mini-Review. Front Physiol 2022; 13:873522. [PMID: 35432002 PMCID: PMC9008331 DOI: 10.3389/fphys.2022.873522] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Animal models are useful to understand the myriad physiological effects of hypoxia. Such models attempt to recapitulate the hypoxemia of human disease in various ways. In this mini-review, we consider the various animal models which have been deployed to understand the effects of chronic hypoxia on pulmonary and systemic blood pressure, glucose and lipid metabolism, atherosclerosis, and stroke. Chronic sustained hypoxia (CSH)-a model of chronic lung or heart diseases in which hypoxemia may be longstanding and persistent, or of high altitude, in which effective atmospheric oxygen concentration is low-reliably induces pulmonary hypertension in rodents, and appears to have protective effects on glucose metabolism. Chronic intermittent hypoxia (CIH) has long been used as a model of obstructive sleep apnea (OSA), in which recurrent airway occlusion results in intermittent reductions in oxyhemoglobin saturations throughout the night. CIH was first shown to increase systemic blood pressure, but has also been associated with other maladaptive physiological changes, including glucose dysregulation, atherosclerosis, progression of nonalcoholic fatty liver disease, and endothelial dysfunction. However, models of CIH have generally been implemented so as to mimic severe human OSA, with comparatively less focus on milder hypoxic regimens. Here we discuss CSH and CIH conceptually, the effects of these stimuli, and limitations of the available data.
Collapse
Affiliation(s)
- Laura A. Barnes
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Ana Sanchez-Azofra
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, Department of Medicine, University of California, San Diego, San Diego, CA, United States
- Servicio de Neumología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
30
|
Su J, Meng Y, Fang Y, Sun L, Wang M, Liu Y, Zhao C, Dai L, Ouyang S. Role of raphe magnus 5-HT 1A receptor in increased ventilatory responses induced by intermittent hypoxia in rats. Respir Res 2022; 23:42. [PMID: 35241072 PMCID: PMC8892800 DOI: 10.1186/s12931-022-01970-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/22/2022] [Indexed: 11/11/2022] Open
Abstract
Background Intermittent hypoxia induces increased ventilatory responses in a 5-HT-dependent manner. This study aimed to explore that effect of raphe magnus serotonin 1A receptor (5-HT1A) receptor on the increased ventilatory responses induced by intermittent hypoxia. Methods Stereotaxic surgery was performed in adult male rats, and acute and chronic intermittent hypoxia models were established after recovery from surgery. The experimental group received microinjections of 5-HT1A receptor agonist 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT) into the raphe magnus nucleus (RMg). Meanwhile, the control group received microinjections of artificial cerebrospinal fluid instead of 8-OH-DPAT. Ventilatory responses were compared among the different groups of oxygen status. 5-HT expressions in the RMg region were assessed by immunohistochemistry after chronic intermittent hypoxia. Results Compared with the normoxia group, the acute intermittent hypoxia group exhibited higher ventilatory responses (e.g., shorter inspiratory time and higher tidal volume, frequency of breathing, minute ventilation, and mean inspiratory flow) (P < 0.05). 8-OH-DPAT microinjection partly weakened these changes in the acute intermittent hypoxia group. Further, compared with the acute intermittent hypoxia group, rats in chronic intermittent hypoxia group exhibited higher measures of ventilatory responses after 1 day of intermittent hypoxia (P < 0.05). These effects peaked after 3 days of intermittent hypoxia treatment and then decreased gradually. Moreover, these changes were diminished in the experimental group. 5-HT expression in the RMg region increased after chronic intermittent hypoxia, which was consistent with the changing trend of ventilatory responses. While activation of the 5-HT1A receptor in the RMg region alleviated this phenomenon. Conclusions The results indicate that RMg 5-HT1A receptor, via changing the expression level of 5-HT in the RMg region, is involved in the modulation of the increased ventilatory responses induced by intermittent hypoxia. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01970-6.
Collapse
Affiliation(s)
- Jiao Su
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yang Meng
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yifei Fang
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Linge Sun
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Mengge Wang
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yanjun Liu
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Chunling Zhao
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Henan, 450052, China
| | - Songyun Ouyang
- Department of Respiratory and Sleep Medicine, First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
31
|
Stoica SI, Bleotu C, Ciobanu V, Ionescu AM, Albadi I, Onose G, Munteanu C. Considerations about Hypoxic Changes in Neuraxis Tissue Injuries and Recovery. Biomedicines 2022; 10:481. [PMID: 35203690 PMCID: PMC8962344 DOI: 10.3390/biomedicines10020481] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/06/2022] [Accepted: 02/13/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxia represents the temporary or longer-term decrease or deprivation of oxygen in organs, tissues, and cells after oxygen supply drops or its excessive consumption. Hypoxia can be (para)-physiological-adaptive-or pathological. Thereby, the mechanisms of hypoxia have many implications, such as in adaptive processes of normal cells, but to the survival of neoplastic ones, too. Ischemia differs from hypoxia as it means a transient or permanent interruption or reduction of the blood supply in a given region or tissue and consequently a poor provision with oxygen and energetic substratum-inflammation and oxidative stress damages generating factors. Considering the implications of hypoxia on nerve tissue cells that go through different ischemic processes, in this paper, we will detail the molecular mechanisms by which such structures feel and adapt to hypoxia. We will present the hypoxic mechanisms and changes in the CNS. Also, we aimed to evaluate acute, subacute, and chronic central nervous hypoxic-ischemic changes, hoping to understand better and systematize some neuro-muscular recovery methods necessary to regain individual independence. To establish the link between CNS hypoxia, ischemic-lesional mechanisms, and neuro-motor and related recovery, we performed a systematic literature review following the" Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA") filtering method by interrogating five international medical renown databases, using, contextually, specific keywords combinations/"syntaxes", with supplementation of the afferent documentation through an amount of freely discovered, also contributive, bibliographic resources. As a result, 45 papers were eligible according to the PRISMA-inspired selection approach, thus covering information on both: intimate/molecular path-physiological specific mechanisms and, respectively, consequent clinical conditions. Such a systematic process is meant to help us construct an article structure skeleton giving a primary objective input about the assembly of the literature background to be approached, summarised, and synthesized. The afferent contextual search (by keywords combination/syntaxes) we have fulfilled considerably reduced the number of obtained articles. We consider this systematic literature review is warranted as hypoxia's mechanisms have opened new perspectives for understanding ischemic changes in the CNS neuraxis tissue/cells, starting at the intracellular level and continuing with experimental research to recover the consequent clinical-functional deficits better.
Collapse
Affiliation(s)
- Simona Isabelle Stoica
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
| | - Coralia Bleotu
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania;
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest (PUB), 060042 Bucharest, Romania;
| | - Anca Mirela Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
| | - Irina Albadi
- Teaching Emergency County Hospital “Sf. Apostol Andrei”, 900591 Constanta, Romania;
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila” (UMPCD), 020022 Bucharest, Romania; (S.I.S.); (A.M.I.)
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
| | - Constantin Munteanu
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), 041915 Bucharest, Romania
- Department of Research, Romanian Association of Balneology, 022251 Bucharest, Romania
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania
| |
Collapse
|
32
|
Sajjadi E, Seven YB, Ehrbar JG, Wymer JP, Mitchell GS, Smith BK. Acute intermittent hypoxia and respiratory muscle recruitment in people with amyotrophic lateral sclerosis: A preliminary study. Exp Neurol 2022; 347:113890. [PMID: 34624328 PMCID: PMC9488543 DOI: 10.1016/j.expneurol.2021.113890] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 01/03/2023]
Abstract
Respiratory failure is the main cause of death in amyotrophic lateral sclerosis (ALS). Since no effective treatments to preserve independent breathing are available, there is a critical need for new therapies to preserve or restore breathing ability. Since acute intermittent hypoxia (AIH) elicits spinal respiratory motor plasticity in rodent ALS models, and may restore breathing ability in people with ALS, we performed a proof-of-principle study to investigate this possibility in ALS patients. Quiet breathing, sniff nasal inspiratory pressure (SNIP) and maximal inspiratory pressure (MIP) were tested in 13 persons with ALS and 10 age-matched controls, before and 60 min post-AIH (15, 1 min episodes of 10% O2, 2 min normoxic intervals) or sham AIH (continuous normoxia). The root mean square (RMS) of the right and left diaphragm, 2nd parasternal, scalene and sternocleidomastoid muscles were monitored. A vector analysis was used to calculate summated vector magnitude (Mag) and similarity index (SI) of collective EMG activity during quiet breathing, SNIP and MIP maneuvers. AIH facilitated tidal volume and minute ventilation (treatment main effects: p < 0.05), and Mag (ie. collective respiratory muscle activity; p < 0.001) during quiet breathing in ALS and control subjects, but there was no effect on SI during quiet breathing. SNIP SI decreased in both groups post-AIH (p < 0.005), whereas Mag was unchanged (p = 0.09). No differences were observed in SNIP or MIP post AIH in either group. Discomfort was not reported during AIH by any subject, nor were adverse events observed. Thus, AIH may be a safe way to increase collective inspiratory muscle activity during quiet breathing in ALS patients, although a single AIH presentation was not sufficient to significantly increase peak inspiratory pressure generation. These preliminary results provide evidence that AIH may improve breathing function in people with ALS, and that future studies of prolonged, repetitive AIH protocols are warranted.
Collapse
Affiliation(s)
- Elaheh Sajjadi
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA, 32610,McKnight Brain Institute, University of Florida, Gainesville, FL, USA, 32610,Department of Physical Therapy, University of Florida, Gainesville, FL, USA, 32610
| | - Yasin B. Seven
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA, 32610,McKnight Brain Institute, University of Florida, Gainesville, FL, USA, 32610,Department of Physical Therapy, University of Florida, Gainesville, FL, USA, 32610
| | - Jessica G Ehrbar
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA, 32610
| | - James P. Wymer
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA, 32610,McKnight Brain Institute, University of Florida, Gainesville, FL, USA, 32610,Neurology, University of Florida, Gainesville, FL, USA, 32610
| | - Gordon S. Mitchell
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA, 32610,McKnight Brain Institute, University of Florida, Gainesville, FL, USA, 32610,Department of Physical Therapy, University of Florida, Gainesville, FL, USA, 32610
| | - Barbara K. Smith
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA, 32610,Department of Physical Therapy, University of Florida, Gainesville, FL, USA, 32610,Pediatrics, University of Florida, Gainesville, FL, USA, 32610
| |
Collapse
|
33
|
Vose AK, Welch JF, Nair J, Dale EA, Fox EJ, Muir GD, Trumbower RD, Mitchell GS. Therapeutic acute intermittent hypoxia: A translational roadmap for spinal cord injury and neuromuscular disease. Exp Neurol 2022; 347:113891. [PMID: 34637802 PMCID: PMC8820239 DOI: 10.1016/j.expneurol.2021.113891] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 10/03/2021] [Indexed: 01/03/2023]
Abstract
We review progress towards greater mechanistic understanding and clinical translation of a strategy to improve respiratory and non-respiratory motor function in people with neuromuscular disorders, therapeutic acute intermittent hypoxia (tAIH). In 2016 and 2020, workshops to create and update a "road map to clinical translation" were held to help guide future research and development of tAIH to restore movement in people living with chronic, incomplete spinal cord injuries. After briefly discussing the pioneering, non-targeted basic research inspiring this novel therapeutic approach, we then summarize workshop recommendations, emphasizing critical knowledge gaps, priorities for future research effort, and steps needed to accelerate progress as we evaluate the potential of tAIH for routine clinical use. Highlighted areas include: 1) greater mechanistic understanding, particularly in non-respiratory motor systems; 2) optimization of tAIH protocols to maximize benefits; 3) identification of combinatorial treatments that amplify plasticity or remove plasticity constraints, including task-specific training; 4) identification of biomarkers for individuals most/least likely to benefit from tAIH; 5) assessment of long-term tAIH safety; and 6) development of a simple, safe and effective device to administer tAIH in clinical and home settings. Finally, we update ongoing clinical trials and recent investigations of tAIH in SCI and other clinical disorders that compromise motor function, including ALS, multiple sclerosis, and stroke.
Collapse
Affiliation(s)
- Alicia K Vose
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Joseph F Welch
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Jayakrishnan Nair
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Erica A Dale
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Emily J Fox
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Gillian D Muir
- Department of Biomedical Sciences, WCVM, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Randy D Trumbower
- Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
34
|
Mitchell GS, Baker TL. Respiratory neuroplasticity: Mechanisms and translational implications of phrenic motor plasticity. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:409-432. [PMID: 35965036 DOI: 10.1016/b978-0-323-91534-2.00016-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Widespread appreciation that neuroplasticity is an essential feature of the neural system controlling breathing has emerged only in recent years. In this chapter, we focus on respiratory motor plasticity, with emphasis on the phrenic motor system. First, we define related but distinct concepts: neuromodulation and neuroplasticity. We then focus on mechanisms underlying two well-studied models of phrenic motor plasticity: (1) phrenic long-term facilitation following brief exposure to acute intermittent hypoxia; and (2) phrenic motor facilitation after prolonged or recurrent bouts of diminished respiratory neural activity. Advances in our understanding of these novel and important forms of plasticity have been rapid and have already inspired translation in multiple respects: (1) development of novel therapeutic strategies to preserve/restore breathing function in humans with severe neurological disorders, such as spinal cord injury and amyotrophic lateral sclerosis; and (2) the discovery that similar plasticity also occurs in nonrespiratory motor systems. Indeed, the realization that similar plasticity occurs in respiratory and nonrespiratory motor neurons inspired clinical trials to restore leg/walking and hand/arm function in people living with chronic, incomplete spinal cord injury. Similar application may be possible to other clinical disorders that compromise respiratory and non-respiratory movements.
Collapse
Affiliation(s)
- Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| | - Tracy L Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
35
|
Sandhu MS, Rymer WZ. Brief exposure to systemic hypoxia enhances plasticity of the central nervous system in spinal cord injured animals and man. Curr Opin Neurol 2021; 34:819-824. [PMID: 34545014 DOI: 10.1097/wco.0000000000000990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW We have known for many decades that animals that sustain injuries to the neuraxis, which result in respiratory impairment, are able to develop rapid neural compensation for these injuries. This compensation, which is linked to the systemic hypoxia resulting from damage to the respiratory apparatus, is a potent manifestation of neural plasticity. Hypoxia-induced plasticity is also applicable to somatic neural systems that regulate motor activity in extremity muscles. We report on recent developments in our understanding of the mechanisms underlying this seemingly beneficial action of acute intermittent hypoxia (AIH). RECENT FINDINGS AIH improves breathing in animal models of spinal cord injury, and increases strength and endurance in individuals with incomplete spinal injuries. The role of AIH as a therapeutic intervention remains to be confirmed but it has proved to be well tolerated for use in humans with no adverse effects reported to date. The effects of AIH emerge rapidly and persist for several hours raising the possibility that the intervention may serve as a priming mechanism for facilitating rehabilitation and promoting recovery after neurologic injury in man. SUMMARY AIH is emerging as a potent and relatively inexpensive modality for inducing neuroplasticity, so it may prove feasible to use AIH in a clinical setting.
Collapse
Affiliation(s)
- Milap S Sandhu
- Shirley Ryan AbilityLab
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, USA
| | - William Z Rymer
- Shirley Ryan AbilityLab
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
36
|
Rana S, Sunshine MD, Greer JJ, Fuller DD. Ampakines Stimulate Diaphragm Activity after Spinal Cord Injury. J Neurotrauma 2021; 38:3467-3482. [PMID: 34806433 PMCID: PMC8713281 DOI: 10.1089/neu.2021.0301] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Respiratory compromise after cervical spinal cord injury (SCI) is a leading cause of mortality and morbidity. Most SCIs are incomplete, and spinal respiratory motoneurons as well as proprio- and bulbospinal synaptic pathways provide a neurological substrate to enhance respiratory output. Ampakines are allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, which are prevalent on respiratory neurons. We hypothesized that low dose ampakine treatment could safely and effectively increase diaphragm electromyography (EMG) activity that has been impaired as a result of acute- or sub-acute cervical SCI. Diaphragm EMG was recorded using chronic indwelling electrodes in unanesthetized, freely moving rats. A spinal hemi-lesion was induced at C2 (C2Hx), and rats were studied at 4 and 14 days post-injury during room air breathing and acute respiratory challenge accomplished by inspiring a 10% O2, 7% CO2 gas mixture. Once a stable baseline recording was established, one of two different ampakines (CX717 or CX1739, 5 mg/kg, intravenous) or a vehicle (2-hydroxypropyl-beta-cyclodextrin [HPCD]) was delivered. At 4 days post-injury, both ampakines increased diaphragm EMG output ipsilateral to C2Hx during both baseline breathing and acute respiratory challenge. Only CX1739 treatment also led to a sustained (15 min) increase in ipsilateral EMG output. At 14 days post-injury, both ampakines produced sustained increases in ipsilateral diaphragm EMG output and enabled increased output during the respiratory challenge. We conclude that low dose ampakine treatment can increase diaphragm EMG activity after cervical SCI, and therefore may provide a pharmacological strategy that could be useful in the context of respiratory rehabilitation.
Collapse
Affiliation(s)
- Sabhya Rana
- Department of Physical Therapy and University of Florida, Gainesville, Florida, USA
- McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, Gainesville, Florida, USA
| | - Michael D. Sunshine
- Department of Physical Therapy and University of Florida, Gainesville, Florida, USA
- McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, Gainesville, Florida, USA
| | - John J. Greer
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - David D. Fuller
- Department of Physical Therapy and University of Florida, Gainesville, Florida, USA
- McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, Gainesville, Florida, USA
| |
Collapse
|
37
|
Allen LL, Nichols NL, Asa ZA, Emery AT, Ciesla MC, Santiago JV, Holland AE, Mitchell GS, Gonzalez-Rothi EJ. Phrenic motor neuron survival below cervical spinal cord hemisection. Exp Neurol 2021; 346:113832. [PMID: 34363808 PMCID: PMC9065093 DOI: 10.1016/j.expneurol.2021.113832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 02/04/2023]
Abstract
Cervical spinal cord injury (cSCI) severs bulbospinal projections to respiratory motor neurons, paralyzing respiratory muscles below the injury. C2 spinal hemisection (C2Hx) is a model of cSCI often used to study spontaneous and induced plasticity and breathing recovery post-injury. One key assumption is that C2Hx dennervates motor neurons below the injury, but does not affect their survival. However, a recent study reported substantial bilateral motor neuron death caudal to C2Hx. Since phrenic motor neuron (PMN) death following C2Hx would have profound implications for therapeutic strategies designed to target spared neural circuits, we tested the hypothesis that C2Hx minimally impacts PMN survival. Using improved retrograde tracing methods, we observed no loss of PMNs at 2- or 8-weeks post-C2Hx. We also observed no injury-related differences in ChAT or NeuN immunolabeling within labelled PMNs. Although we found no evidence of PMN loss following C2Hx, we cannot rule out neuronal loss in other motor pools. These findings address an essential prerequisite for studies that utilize C2Hx as a model to explore strategies for inducing plasticity and/or regeneration within the phrenic motor system, as they provide important insights into the viability of phrenic motor neurons as therapeutic targets after high cervical injury.
Collapse
Affiliation(s)
- Latoya L Allen
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nicole L Nichols
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Zachary A Asa
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | | | - Marissa C Ciesla
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Juliet V Santiago
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ashley E Holland
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
38
|
Li G, Liu J, Guan Y, Ji X. The role of hypoxia in stem cell regulation of the central nervous system: From embryonic development to adult proliferation. CNS Neurosci Ther 2021; 27:1446-1457. [PMID: 34817133 PMCID: PMC8611781 DOI: 10.1111/cns.13754] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is involved in the regulation of various cell functions in the body, including the regulation of stem cells. The hypoxic microenvironment is indispensable from embryonic development to the regeneration and repair of adult cells. In addition to embryonic stem cells, which need to maintain their self-renewal properties and pluripotency in a hypoxic environment, adult stem cells, including neural stem cells (NSCs), also exist in a hypoxic microenvironment. The subventricular zone (SVZ) and hippocampal dentate gyrus (DG) are the main sites of adult neurogenesis in the brain. Hypoxia can promote the proliferation, migration, and maturation of NSCs in these regions. Also, because most neurons in the brain are non-regenerative, stem cell transplantation is considered as a promising strategy for treating central nervous system (CNS) diseases. Hypoxic treatment also increases the effectiveness of stem cell therapy. In this review, we firstly describe the role of hypoxia in different stem cells, such as embryonic stem cells, NSCs, and induced pluripotent stem cells, and discuss the role of hypoxia-treated stem cells in CNS diseases treatment. Furthermore, we highlight the role and mechanisms of hypoxia in regulating adult neurogenesis in the SVZ and DG and adult proliferation of other cells in the CNS.
Collapse
Affiliation(s)
- Gaifen Li
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jia Liu
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
| | - Yuying Guan
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Laboratory of Brain DisordersMinistry of Science and TechnologyCollaborative Innovation Center for Brain DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
39
|
Randelman M, Zholudeva LV, Vinit S, Lane MA. Respiratory Training and Plasticity After Cervical Spinal Cord Injury. Front Cell Neurosci 2021; 15:700821. [PMID: 34621156 PMCID: PMC8490715 DOI: 10.3389/fncel.2021.700821] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/11/2021] [Indexed: 12/30/2022] Open
Abstract
While spinal cord injuries (SCIs) result in a vast array of functional deficits, many of which are life threatening, the majority of SCIs are anatomically incomplete. Spared neural pathways contribute to functional and anatomical neuroplasticity that can occur spontaneously, or can be harnessed using rehabilitative, electrophysiological, or pharmacological strategies. With a focus on respiratory networks that are affected by cervical level SCI, the present review summarizes how non-invasive respiratory treatments can be used to harness this neuroplastic potential and enhance long-term recovery. Specific attention is given to "respiratory training" strategies currently used clinically (e.g., strength training) and those being developed through pre-clinical and early clinical testing [e.g., intermittent chemical stimulation via altering inhaled oxygen (hypoxia) or carbon dioxide stimulation]. Consideration is also given to the effect of training on non-respiratory (e.g., locomotor) networks. This review highlights advances in this area of pre-clinical and translational research, with insight into future directions for enhancing plasticity and improving functional outcomes after SCI.
Collapse
Affiliation(s)
- Margo Randelman
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Lyandysha V Zholudeva
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States.,Gladstone Institutes, San Francisco, CA, United States
| | - Stéphane Vinit
- INSERM, END-ICAP, Université Paris-Saclay, UVSQ, Versailles, France
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
40
|
Thakre PP, Sunshine MD, Fuller DD. Ampakine pretreatment enables a single hypoxic episode to produce phrenic motor facilitation with no added benefit of additional episodes. J Neurophysiol 2021; 126:1420-1429. [PMID: 34495779 DOI: 10.1152/jn.00307.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Repeated short episodes of hypoxia produce a sustained increase in phrenic nerve output lasting well beyond acute intermittent hypoxia (AIH) exposure (i.e., phrenic long-term facilitation; pLTF). Pretreatment with ampakines, drugs which allosterically modulate AMPA receptors, enables a single brief episode of hypoxia to produce pLTF, lasting up to 90 min after hypoxia. Here, we tested the hypothesis that ampakine pretreatment would enhance the magnitude of pLTF evoked by repeated bouts of hypoxia. Phrenic nerve output was recorded in urethane-anesthetized, mechanically ventilated, and vagotomized adult male Sprague-Dawley rats. Initial experiments demonstrated that ampakine CX717 (15 mg/kg iv) caused an acute increase in phrenic nerve inspiratory burst amplitude reaching 70 ± 48% baseline (BL) after 2 min (P = 0.01). This increased bursting was not sustained (2 ± 32% BL at 60 min, P = 0.9). When CX717 was delivered 2 min before a single episode of isocapnic hypoxia (5 min, [Formula: see text] = 44 ± 9 mmHg), facilitation of phrenic nerve burst amplitude occurred (96 ± 62% BL at 60 min, P < 0.001). However, when CX717 was given 2 min before three, 5-min hypoxic episodes ([Formula: see text] = 45 ± 6 mmHg) pLTF was attenuated and did not reach statistical significance (24 ± 29% BL, P = 0.08). In the absence of CX717 pretreatment, pLTF was observed after three (74 ± 33% BL at 60 min, P < 0.001) but not one episode of hypoxia (1 ± 8% BL at 60 min, P = 0.9). We conclude that pLTF is not enhanced when ampakine pretreatment is followed by repeated bouts of hypoxia. Rather, the combination of ampakine and a single hypoxic episode appears to be ideal for producing sustained increase in phrenic motor output.NEW & NOTEWORTHY Pretreatment with ampakine CX717 created conditions that enabled an acute bout of moderate hypoxia to evoke phrenic motor facilitation, but this response was not observed when ampakine pretreatment was followed by intermittent hypoxia. Thus, in anesthetized and spinal intact rats, the combination of ampakine and one bout of hypoxia appears ideal for triggering respiratory neuroplasticity.
Collapse
Affiliation(s)
- Prajwal P Thakre
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Michael D Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida
| |
Collapse
|
41
|
Barok R, Grittner JML, Dougherty BJ. The long-term impact of ovariectomy on ventilation and expression of phrenic long-term facilitation in female rats. Exp Physiol 2021; 106:2002-2012. [PMID: 34180081 PMCID: PMC8410681 DOI: 10.1113/ep089546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/18/2021] [Indexed: 01/04/2023]
Abstract
NEW FINDINGS What is the central question of this study? Would ovariectomy cause prolonged changes in ventilation and sustained loss of acute, intermittent hypoxia-induced neuroplasticity or would these outcomes be restored with time? What is the main finding and its importance? Our main findings demonstrate that ovariectomy elicits minimal alteration in overall breathing function but impairs acute, intermittent hypoxia-induced plasticity for ≤ 12 weeks. ABSTRACT Sex hormones are necessary to enable respiratory neuroplasticity, including phrenic long-term facilitation (pLTF), a form of respiratory motor plasticity elicited by acute, intermittent hypoxia (AIH). Female rats exhibit a progressive increase in phrenic nerve amplitude after AIH characteristic of pLTF only during pro-oestrus, the stage of the oestrous cycle notable for elevated circulating oestradiol levels. Removal of the ovaries [ovariectomy (OVX)], the primary source of circulating oestradiol, also eliminates AIH-induced pLTF after 1 week. Ovariectomy is used routinely as a model to examine the impact of sex hormones on CNS structure and function, but the long-term impact of OVX is rarely examined. Extra-ovarian sites of oestradiol synthesis, including multiple CNS sites, have been identified and might possess the capacity to restore oestradiol levels, in part, over time, impacting respiratory function and the expression of respiratory neuroplasticity. We examined both ventilation in awake, freely behaving female rats, using barometric plethysmography, and the expression of AIH-induced pLTF in anaesthetized, ventilated female rats 2 and 12 weeks after OVX and compared them with age-matched ovarian-intact female rats. Our findings indicate that chronic OVX had little impact on baseline breathing or in the response to respiratory challenge (10% O2 , 5% CO2 , balance N2 ) during plethysmography. However, OVX rats at both 2 and 12 weeks demonstrated a persistent loss of AIH-induced pLTF relative to control animals (P < 0.01), suggesting that other sources of oestradiol synthesis were insufficient to restore pLTF. These data are consistent with our previous work indicating that oestradiol plays a key role in expression of AIH-induced respiratory neuroplasticity.
Collapse
Affiliation(s)
- Rebecca Barok
- Rehabilitation Science Graduate Program, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jessica M L Grittner
- Rehabilitation Science Graduate Program, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Brendan J Dougherty
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
42
|
Tadjalli A, Seven YB, Sharma A, McCurdy CR, Bolser DC, Levitt ES, Mitchell GS. Acute morphine blocks spinal respiratory motor plasticity via long-latency mechanisms that require toll-like receptor 4 signalling. J Physiol 2021; 599:3771-3797. [PMID: 34142718 DOI: 10.1113/jp281362] [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: 03/14/2021] [Accepted: 06/15/2021] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS While respiratory complications following opioid use are mainly mediated via activation of mu opioid receptors, long-latency off-target signalling via innate immune toll-like receptor 4 (TLR4) may impair other essential elements of breathing control such as respiratory motor plasticity. In adult rats, pre-treatment with a single dose of morphine blocked long-term facilitation (LTF) of phrenic motor output via a long-latency TLR4-dependent mechanism. In the phrenic motor nucleus, morphine triggered TLR4-dependent activation of microglial p38 MAPK - a key enzyme that orchestrates inflammatory signalling and is known to undermine phrenic LTF. Morphine-induced LTF loss may destabilize breathing, potentially contributing to respiratory side effects. Therefore, we suggest minimizing TLR-4 signalling may improve breathing stability during opioid therapy. ABSTRACT Opioid-induced respiratory dysfunction is a significant public health burden. While respiratory effects are mediated via mu opioid receptors, long-latency off-target opioid signalling through innate immune toll-like receptor 4 (TLR4) may modulate essential elements of breathing control, particularly respiratory motor plasticity. Plasticity in respiratory motor circuits contributes to the preservation of breathing in the face of destabilizing influences. For example, respiratory long-term facilitation (LTF), a well-studied model of respiratory motor plasticity triggered by acute intermittent hypoxia, promotes breathing stability by increasing respiratory motor drive to breathing muscles. Some forms of respiratory LTF are exquisitely sensitive to inflammation and are abolished by even a mild inflammation triggered by TLR4 activation (e.g. via systemic lipopolysaccharides). Since opioids induce inflammation and TLR4 activation, we hypothesized that opioids would abolish LTF through a TLR4-dependent mechanism. In adult Sprague Dawley rats, pre-treatment with a single systemic injection of the prototypical opioid agonist morphine blocks LTF expression several hours later in the phrenic motor system - the motor pool driving diaphragm muscle contractions. Morphine blocked phrenic LTF via TLR4-dependent mechanisms because pre-treatment with (+)-naloxone - the opioid inactive stereoisomer and novel small molecule TLR4 inhibitor - prevented impairment of phrenic LTF in morphine-treated rats. Morphine triggered TLR4-dependent activation of microglial p38 MAPK within the phrenic motor system - a key enzyme that orchestrates inflammatory signalling and undermines phrenic LTF. Morphine-induced LTF loss may destabilize breathing, potentially contributing to respiratory side effects. We suggest minimizing TLR-4 signalling may improve breathing stability during opioid therapy by restoring endogenous mechanisms of plasticity within respiratory motor circuits.
Collapse
Affiliation(s)
- Arash Tadjalli
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Yasin B Seven
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Department of Pharmaceutics, University of Florida, Gainesville, FL, USA
| | | | - Donald C Bolser
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA.,Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Erica S Levitt
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA.,Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
43
|
Gonzalez-Rothi EJ, Lee KZ. Intermittent hypoxia and respiratory recovery in pre-clinical rodent models of incomplete cervical spinal cord injury. Exp Neurol 2021; 342:113751. [PMID: 33974878 DOI: 10.1016/j.expneurol.2021.113751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/24/2021] [Accepted: 05/06/2021] [Indexed: 10/21/2022]
Abstract
Impaired respiratory function is a common and devastating consequence of cervical spinal cord injury. Accordingly, the development of safe and effective treatments to restore breathing function is critical. Acute intermittent hypoxia has emerged as a promising therapeutic strategy to treat respiratory insufficiency in individuals with spinal cord injury. Since the original report by Bach and Mitchell (1996) concerning long-term facilitation of phrenic motor output elicited by brief, episodic exposure to reduced oxygen, a series of studies in animal models have led to the realization that acute intermittent hypoxia may have tremendous potential for inducing neuroplasticity and functional recovery in the injured spinal cord. Advances in our understanding of the neurobiology of acute intermittent hypoxia have prompted us to begin to explore its effects in human clinical studies. Here, we review the basic neurobiology of the control of breathing and the pathophysiology and respiratory consequences of two common experimental models of incomplete cervical spinal cord injury (i.e., high cervical hemisection and mid-cervical contusion). We then discuss the impact of acute intermittent hypoxia on respiratory motor function in these models: work that has laid the foundation for translation of this promising therapeutic strategy to clinical populations. Lastly, we examine the limitations of these animal models and intermittent hypoxia and discuss how future work in animal models may further advance the translation and therapeutic efficacy of this treatment.
Collapse
Affiliation(s)
- Elisa J Gonzalez-Rothi
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kun-Ze Lee
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
44
|
Sutor T, Cavka K, Vose AK, Welch JF, Davenport P, Fuller DD, Mitchell GS, Fox EJ. Single-session effects of acute intermittent hypoxia on breathing function after human spinal cord injury. Exp Neurol 2021; 342:113735. [PMID: 33951477 DOI: 10.1016/j.expneurol.2021.113735] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/14/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
After spinal cord injury (SCI) respiratory complications are a leading cause of morbidity and mortality. Acute intermittent hypoxia (AIH) triggers spinal respiratory motor plasticity in rodent models, and repetitive AIH may have the potential to restore breathing capacity in those with SCI. As an initial approach to provide proof of principle for such effects, we tested single-session AIH effects on breathing function in adults with chronic SCI. 17 adults (13 males; 34.1 ± 14.5 years old; 13 motor complete SCI; >6 months post injury) completed two randomly ordered sessions, AIH versus sham. AIH consisted of 15, 1-min episodes (hypoxia: 10.3% O2; sham: 21% O2) interspersed with room air breathing (1.5 min, 21% oxygen); no attempt was made to regulate arterial CO2 levels. Blood oxygen saturation (SpO2), maximal inspiratory and expiratory pressures (MIP; MEP), forced vital capacity (FVC), and mouth occlusion pressure within 0.1 s (P0.1) were assessed. Outcomes were compared using nonparametric Wilcoxon's tests, or a 2 × 2 ANOVA. Baseline SpO2 was 97.2 ± 1.3% and was unchanged during sham experiments. During hypoxic episodes, SpO2 decreased to 84.7 ± 0.9%, and returned to baseline levels during normoxic intervals. Outcomes were unchanged from baseline post-sham. Greater increases in MIP were evident post AIH vs. sham (median values; +10.8 cmH2O vs. -2.6 cmH2O respectively, 95% confidence interval (-18.7) - (-4.3), p = .006) with a moderate Cohen's effect size (0.68). P0.1, MEP and FVC did not change post-AIH. A single AIH session increased maximal inspiratory pressure generation, but not other breathing functions in adults with SCI. Reasons may include greater spared innervation to inspiratory versus expiratory muscles or differences in the capacity for AIH-induced plasticity in inspiratory motor neuron pools. Based on our findings, the therapeutic potential of AIH on breathing capacity in people with SCI warrants further investigation.
Collapse
Affiliation(s)
- Tommy Sutor
- Hunter Holmes McGuire VA Medical Center, United States.
| | - Kathryn Cavka
- Brooks Rehabilitation, Jacksonville, FL, United States.
| | - Alicia K Vose
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States.
| | - Joseph F Welch
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States.
| | - Paul Davenport
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States; Department of Physiological Sciences, University of Florida, Gainesville, FL, United States.
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States.
| | - Gordon S Mitchell
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States.
| | - Emily J Fox
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States; Brooks Rehabilitation, Jacksonville, FL, United States; Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
45
|
Gonzalez-Rothi EJ, Tadjalli A, Allen LL, Ciesla MC, Chami ME, Mitchell GS. Protocol-Specific Effects of Intermittent Hypoxia Pre-Conditioning on Phrenic Motor Plasticity in Rats with Chronic Cervical Spinal Cord Injury. J Neurotrauma 2021; 38:1292-1305. [PMID: 33446048 PMCID: PMC8182475 DOI: 10.1089/neu.2020.7324] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
"Low-dose" acute intermittent hypoxia (AIH; 3-15 episodes/day) is emerging as a promising therapeutic strategy to improve motor function after incomplete cervical spinal cord injury (cSCI). Conversely, chronic "high-dose" intermittent hypoxia (CIH; > 80-100 episodes/day) elicits multi-system pathology and is a hallmark of sleep apnea, a condition highly prevalent in individuals with cSCI. Whereas daily AIH (dAIH) enhances phrenic motor plasticity in intact rats, it is abolished by CIH. However, there have been no direct comparisons of prolonged dAIH versus CIH on phrenic motor outcomes after chronic cSCI. Thus, phrenic nerve activity and AIH-induced phrenic long-term facilitation (pLTF) were assessed in anesthetized rats. Experimental groups included: 1) intact rats exposed to 28 days of normoxia (Nx28; 21% O2; 8 h/day), and three groups with chronic C2 hemisection (C2Hx) exposed to either: 2) Nx28; 3) dAIH (dAIH28; 10, 5-min episodes of 10.5% O2/day; 5-min intervals); or 4) CIH (IH28-2/2; 2-min episodes; 2-min intervals; 8 h/day). Baseline ipsilateral phrenic nerve activity was reduced in injured versus intact rats but unaffected by dAIH28 or IH28-2/2. There were no group differences in contralateral phrenic activity. pLTF was enhanced bilaterally by dAIH28 versus Nx28 but unaffected by IH28-2/2. Whereas dAIH28 enhanced pLTF after cSCI, it did not improve baseline phrenic output. In contrast, unlike shorter protocols in intact rats, CIH28-2/2 did not abolish pLTF in chronic C2Hx. Mechanisms of differential responses to dAIH versus CIH are not yet known, particularly in the context of cSCI. Further, it remains unclear whether enhanced phrenic motor plasticity can improve breathing after cSCI.
Collapse
Affiliation(s)
| | - Arash Tadjalli
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Latoya L. Allen
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Marissa C. Ciesla
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Mohamad El Chami
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Gordon S. Mitchell
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
46
|
Welch JF, Perim RR, Argento PJ, Sutor TW, Vose AK, Nair J, Mitchell GS, Fox EJ. Effect of acute intermittent hypoxia on cortico-diaphragmatic conduction in healthy humans. Exp Neurol 2021; 339:113651. [PMID: 33607080 PMCID: PMC8678369 DOI: 10.1016/j.expneurol.2021.113651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 01/06/2023]
Abstract
Acute intermittent hypoxia (AIH) is a strategy to improve motor output in humans with neuromotor impairment. A single AIH session increases the amplitude of motor evoked potentials (MEP) in a finger muscle (first dorsal interosseous), demonstrating enhanced corticospinal neurotransmission. Since AIH elicits phrenic/diaphragm long-term facilitation (LTF) in rodent models, we tested the hypothesis that AIH augments diaphragm MEPs in humans. Eleven healthy adults (7 males, age = 29 ± 6 years) were tested. Transcranial and cervical magnetic stimulation were used to induce diaphragm MEPs and compound muscle action potentials (CMAP) recorded by surface EMG, respectively. Stimulus-response curves were generated prior to and 30-60 min after AIH. Diaphragm LTF was assessed by measurement of integrated EMG burst amplitude and frequency during eupnoeic breathing before and after AIH. Following baseline measurements, AIH was delivered from an oxygen generator connected to a facemask under poikilocapnic conditions (15 one minute episodes of 9% inspired oxygen with one minute room air intervals). There were no detectable changes in MEP (-1.5 ± 12.1%, p = 0.96) or CMAP (+0.1 ± 7.8%, p = 0.97) amplitudes across the stimulus-response curve. At stimulation intensities approximating 50% of the difference between minimum and maximum baseline amplitudes, MEP and CMAP amplitudes were also unchanged (p > 0.05). Further, no AIH effect was observed on diaphragm EMG activity during eupnoea post-AIH (p > 0.05). We conclude that unlike hand muscles, poikilocapnic AIH does not enhance diaphragm MEPs or produce diaphragm LTF in healthy humans.
Collapse
Affiliation(s)
- Joseph F Welch
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA.
| | - Raphael R Perim
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Patrick J Argento
- Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Tommy W Sutor
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Alicia K Vose
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Jayakrishnan Nair
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Emily J Fox
- Breathing Research and Therapeutics Centre, Department of Physical Therapy, University of Florida, Gainesville, FL, USA; Brooks Rehabilitation, Jacksonville, FL, USA
| |
Collapse
|
47
|
[Into thin air - Altitude training and hypoxic conditioning: From athlete to patient]. Rev Mal Respir 2021; 38:404-417. [PMID: 33722445 DOI: 10.1016/j.rmr.2021.02.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/15/2020] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Hypoxic exposure should be considered as a continuum, the effects of which depend on the dose and individual response to hypoxia. Hypoxic conditioning (HC) represents an innovative and promising strategy, ranging from improved human performance to therapeutic applications. STATE OF THE ART With the aim of improving sports performance, the effectiveness of hypoxic exposure, whether natural or simulated, is difficult to demonstrate because of the large variability of the protocols used. In therapeutics, the benefits of HC are described in many pathological conditions such as obesity or cardiovascular pathologies. If the HC benefits from a strong preclinical rationale, its application to humans remains limited. PERSPECTIVES Advances in training and acclimation will require greater personalization and precise periodization of hypoxic exposures. For patients, the harmonization of HC protocols, the identification of biomarkers and the development and subsequent validation of devices allowing a precise control of the hypoxic stimulus are necessary steps for the development of HC. CONCLUSIONS From the athlete to the patient, HC represents an innovative and promising field of research, ranging from the improvement of human performance to the prevention and treatment of certain pathologies.
Collapse
|
48
|
Arnold BM, Toosi BM, Caine S, Mitchell GS, Muir GD. Prolonged acute intermittent hypoxia improves forelimb reach-to-grasp function in a rat model of chronic cervical spinal cord injury. Exp Neurol 2021; 340:113672. [PMID: 33652030 DOI: 10.1016/j.expneurol.2021.113672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/13/2021] [Accepted: 02/25/2021] [Indexed: 12/20/2022]
Abstract
Repetitive acute intermittent hypoxia (AIH - brief, episodes of low inspired oxygen) elicits spinal motor plasticity, resulting in sustained improvements of respiratory and non-respiratory motor function in both animal models and humans with chronic spinal cord injury (SCI). We previously demonstrated that 7 days of AIH combined with task-specific training improves performance on a skilled locomotor task for at least 3 weeks post-treatment in rats with incomplete SCI. Here we investigated the effect of repetitive AIH administered for 12 wks on a forelimb reach-to-grasp task in a rat model of chronic, incomplete cervical SCI. In a replicated, sham-controlled, randomized and blinded study, male Spraque-Dawley rats were subject to partial hemisection at the 3rd cervical spinal segment, and exposed to daily AIH (10, 5 min episodes of 11% inspired O2; 5 min intervals of 21% O2) or sham normoxia (continuous 21% O2) for 7 days beginning 8 weeks post-injury. Treatments were then reduced to 4 daily treatments per week, and continued for 11 weeks. Performance on 2 pre-conditioned motor tasks, single pellet reaching and horizontal ladder walking, was recorded each week for up to 12 weeks after initiating treatment; performance on spontaneous adhesive removal was also tested. SCI significantly impaired reach-to-grasp task performance 8 weeks post-injury (pre-treatment). Daily AIH improved reaching success by the first week of treatment versus sham controls, and this difference was maintained at 12 weeks (p < 0.0001). Daily AIH did not affect step asymmetry or stride length during ladder walking or adhesive removal time. Thus, prolonged AIH combined with task-specific training improved forelimb reach-to-grasp function in rats with a chronic cervical hemisection, but not off-target motor tasks. This study further supports the idea that daily AIH improves limb function when combined with task-specific training.
Collapse
Affiliation(s)
- Breanna M Arnold
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| | - Behzad M Toosi
- Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| | - Sally Caine
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, College of Public Health & Health Professions, University of Florida, 1225 Center Drive, PO Box 100154, Gainesville, FL, United States of America.
| | - Gillian D Muir
- Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada.
| |
Collapse
|
49
|
Perim RR, El-Chami M, Gonzalez-Rothi EJ, Mitchell GS. Baseline Arterial CO 2 Pressure Regulates Acute Intermittent Hypoxia-Induced Phrenic Long-Term Facilitation in Rats. Front Physiol 2021; 12:573385. [PMID: 33716760 PMCID: PMC7943620 DOI: 10.3389/fphys.2021.573385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 02/02/2021] [Indexed: 01/25/2023] Open
Abstract
Moderate acute intermittent hypoxia (mAIH) elicits a progressive increase in phrenic motor output lasting hours post-mAIH, a form of respiratory motor plasticity known as phrenic long-term facilitation (pLTF). mAIH-induced pLTF is initiated by activation of spinally-projecting raphe serotonergic neurons during hypoxia and subsequent serotonin release near phrenic motor neurons. Since raphe serotonergic neurons are also sensitive to pH and CO2, the prevailing arterial CO2 pressure (PaCO2) may modulate their activity (and serotonin release) during hypoxic episodes. Thus, we hypothesized that changes in background PaCO2 directly influence the magnitude of mAIH-induced pLTF. mAIH-induced pLTF was evaluated in anesthetized, vagotomized, paralyzed and ventilated rats, with end-tidal CO2 (i.e., a PaCO2 surrogate) maintained at: (1) ≤39 mmHg (hypocapnia); (2) ∼41 mmHg (normocapnia); or (3) ≥48 mmHg (hypercapnia) throughout experimental protocols. Although baseline phrenic nerve activity tended to be lower in hypocapnia, short-term hypoxic phrenic response, i.e., burst amplitude (Δ = 5.1 ± 1.1 μV) and frequency responses (Δ = 21 ± 4 bpm), was greater than in normocapnic (Δ = 3.6 ± 0.6 μV and 8 ± 4, respectively) or hypercapnic rats (Δ = 2.0 ± 0.6 μV and −2 ± 2, respectively), followed by a progressive increase in phrenic burst amplitude (i.e., pLTF) for at least 60 min post mAIH. pLTF in the hypocapnic group (Δ = 4.9 ± 0.6 μV) was significantly greater than in normocapnic (Δ = 2.8 ± 0.7 μV) or hypercapnic rats (Δ = 1.7 ± 0.4 μV). In contrast, although hypercapnic rats also exhibited significant pLTF, it was attenuated versus hypocapnic rats. When pLTF was expressed as percent change from maximal chemoreflex stimulation, all pairwise comparisons were found to be statistically significant (p < 0.05). We conclude that elevated PaCO2 undermines mAIH-induced pLTF in anesthetized rats. These findings contrast with well-documented effects of PaCO2 on ventilatory LTF in awake humans.
Collapse
Affiliation(s)
- Raphael R Perim
- Department of Physical Therapy, McKnight Brain Institute, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, United States
| | - Mohamed El-Chami
- Department of Physical Therapy, McKnight Brain Institute, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, United States
| | - Elisa J Gonzalez-Rothi
- Department of Physical Therapy, McKnight Brain Institute, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, United States
| | - Gordon S Mitchell
- Department of Physical Therapy, McKnight Brain Institute, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, United States
| |
Collapse
|
50
|
Nichols NL, Mitchell GS. Mechanisms of severe acute intermittent hypoxia-induced phrenic long-term facilitation. J Neurophysiol 2021; 125:1146-1156. [PMID: 33566744 DOI: 10.1152/jn.00691.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Moderate acute intermittent hypoxia (mAIH; 35-55 mmHg PaO2) elicits phrenic long-term facilitation (pLTF) by a mechanism that requires activation of Gq protein-coupled serotonin type 2 receptors, MEK/ERK MAP kinase, and NADPH oxidase activity and is constrained by cAMP-PKA signaling. In contrast, severe AIH (sAIH; 25-35 mmHg PaO2) elicits Gs protein-coupled adenosine type 2 A receptor-dependent pLTF. Another Gs protein-coupled receptor, serotonin 7 receptors, elicits phrenic motor facilitation (pMF) by a mechanism that requires exchange protein activated by cyclic AMP (EPAC) and phosphatidylinositol 3-kinase/Akt (PI3K/Akt) activation and is constrained by NADPH oxidase activity. Here, we tested the hypothesis that the same downstream signaling mechanisms giving rise to serotonin 7 (vs. serotonin 2) receptor-induced pMF underlie sAIH-induced pLTF. In anesthetized rats, sAIH-induced pLTF was compared after pretreatment with intrathecal (C4) injections of inhibitors for: 1) EPAC (ESI-05); 2) MEK/ERK (UO126); 3) PKA (KT-5720); 4) PI3K/Akt (PI828); and 5) NADPH oxidase (apocynin). In partial agreement with our hypothesis, sAIH-induced pLTF was abolished by ESI-05 and PI828 and marginally enhanced by apocynin but, surprisingly, was abolished by UO126 and attenuated by KT-5720. Mechanisms of sAIH-induced pLTF reflect elements of both Gq and Gs pathways to pMF, likely as a consequence of the complex, cross-talk interactions between them.NEW & NOTEWORTHY Distinct mechanisms give rise to pLTF induced by moderate and severe AIH. We demonstrate that, unlike moderate AIH, severe AIH-induced pLTF requires EPAC and PI3K/Akt and is marginally constrained by NADPH oxidase activity. Surprisingly, sAIH-induced pLTF requires MEK/ERK activity similar to moderate AIH-induced pLTF and is reduced by PKA inhibition. We suggest sAIH-induced pLTF arises from complex interactions between dominant mechanisms characteristic of moderate versus severe AIH-induced pLTF.
Collapse
Affiliation(s)
- Nicole L Nichols
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Gordon S Mitchell
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|