Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Respir Physiol Neurobiol ; 302: 103913, 2022 08.
Article in English | MEDLINE | ID: mdl-35436602

ABSTRACT

Continuous positive airway pressure (CPAP) is a primary non-invasive mode of respiratory support for preterm infants. However, emerging evidence suggests CPAP could be an underlying contributor to the unintended pathophysiology of wheezing and associated airway hyperreactivity (AHR) in former preterm infants. The therapeutic benefits of mesenchymal stem cells (MSCs) have been demonstrated in a variety of animal models and several clinical trials are currently underway to assess their safety profiles in the setting of prematurity and bronchopulmonary dysplasia (BPD). In the present study, using a mouse model of neonatal CPAP, we investigated whether conditioned medium harvested from cultures of human bone-marrow derived mesenchymal stem cells (hMSC) could rescue the CPAP-induced AHR, based upon previous observations of their anti-AHR properties. Newborn mice (male and female) were fitted with a custom-made mask for delivery of daily CPAP 3 h/day for the first 7 postnatal days. At postnatal day 21 (two weeks after CPAP ended), lungs were removed, precision-cut lung slices were sectioned and incubated for 48 h in vitro in conditioned medium collected from cultures of three different hMSC donors. As expected, CPAP resulted in AHR to methacholine compared to untreated control mice. hMSC conditioned medium from the cultures of all three donors completely reversed AHR. These data reveal potential therapeutic benefits of hMSC therapy, which may be capable of rescuing the long-term adverse effects of neonatal CPAP on human airway function.


Subject(s)
Bronchopulmonary Dysplasia , Mesenchymal Stem Cells , Respiratory Distress Syndrome, Newborn , Animals , Bone Marrow , Bronchopulmonary Dysplasia/etiology , Continuous Positive Airway Pressure/methods , Culture Media, Conditioned/pharmacology , Female , Humans , Infant, Newborn , Infant, Premature , Male
2.
Am J Physiol Regul Integr Comp Physiol ; 319(2): R233-R242, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32579854

ABSTRACT

Continuous infusion of prostaglandin E1 (PGE1) is used to maintain ductus arteriosus patency in infants with critical congenital heart disease, but it can also cause central apnea suggesting an effect on respiratory neural control. In this study, we investigated whether 1) PGE1 inhibits the various phases of the acute hypoxic ventilatory response (HVR; an index of respiratory control dysfunction) and increases apnea incidence in neonatal rats; and 2) whether these changes would be reversible with caffeine pretreatment. Whole body plethysmography was used to assess the HVR and apnea incidence in neonatal rats 2 h following a single bolus intraperitoneal injection of PGE1 with and without prior caffeine treatment. Untreated rats exhibited a biphasic HVR characterized by an initial increase in minute ventilation followed by a ventilatory decline of the late phase (~5th minute) of the HVR. PGE1 had a dose-dependent effect on the HVR. Contrary to our hypothesis, the lowest dose (1 µg/kg) of PGE1 prevented the ventilatory decline of the late phase of the HVR. However, PGE1 tended to increase postsigh apnea incidence and the coefficient of variability (CV) of breathing frequency, suggesting increased respiratory instability. PGE1 also decreased brainstem microglia mRNA and increased neuronal nitric oxide synthase (nNOS) and platelet-derived growth factor-ß (PDGF-ß) gene expression. Caffeine pretreatment prevented these effects of PGE1, and the adenosine A2A receptor inhibitor MSX-3 had similar preventative effects. Prostaglandin appears to have deleterious effects on brainstem respiratory control regions, possibly involving a microglial-dependent mechanism. The compensatory effects of caffeine or MSX-3 treatment raises the question of whether prostaglandin may also operate on an adenosine-dependent pathway.


Subject(s)
Alprostadil/pharmacology , Brain Stem/drug effects , Caffeine/pharmacology , Pulmonary Ventilation/drug effects , Respiration/drug effects , Animals , Brain Stem/metabolism , Microglia/drug effects , Microglia/metabolism , Nitric Oxide Synthase Type I/metabolism , Plethysmography, Whole Body , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Purinergic P1 Receptor Antagonists/pharmacology , Rats , Rats, Sprague-Dawley
3.
Respir Physiol Neurobiol ; 279: 103465, 2020 08.
Article in English | MEDLINE | ID: mdl-32450147

ABSTRACT

Former preterm infants, many of whom required supplemental O2 support, exhibit sleep disordered breathing and attenuated ventilatory responses to acute hypoxia (HVR) beyond their NICU stay. There is an increasing awareness that early detection of biomarkers in biological fluids may be useful predictors/identifiers of short- and long-term morbidities. In the present study, we identified serotonin (5-HT), dopamine (DA) and hyaluronan (HA) as three potential biomarkers that may be increased by neonatal hyperoxia and tested whether they would be associated with an impaired HVR in a rat model of supplemental O2 exposure. Neonatal rats (postnatal age (P) 6 days, P6) exposed to hyperoxia (40% FIO2, 24 h/day between P1-P5 days of age) exhibited an attenuated early (1 min), but not the late (4-5 min) phase of the HVR compared to normoxia control rats; the attenuated early phase HVR was associated with increased levels of DA (urine and serum), 5-HT (platelet poor plasma only, PPP), and HA (serum only). At P21, both the early and late phases of the HVR were attenuated, but serum and urine levels of all 3 biomarkers were similar to age-matched control rats. These data indicate that changes in several serum and/or urine biomarkers (5-HT, DA, and HA) following short-term (days) neonatal hyperoxia can signify long-term (weeks) respiratory control dysfunction. Further studies are needed to determine whether early detection of similar biomarkers could be convenient predictors of increased risk of abnormalities in respiratory control including sleep disordered breathing in former preterm infants who had received prior supplemental O2 and who might also be at increased risk of SIDS.


Subject(s)
Adaptation, Physiological/physiology , Brain Stem/metabolism , Dopamine/metabolism , Hyaluronic Acid/metabolism , Hyperoxia/metabolism , Hypoxia/metabolism , Oxygen Inhalation Therapy/adverse effects , Serotonin/metabolism , Animals , Animals, Newborn , Dopamine/blood , Dopamine/urine , Gene Expression , Humans , Hyaluronan Synthases/genetics , Hyaluronic Acid/blood , Hyaluronic Acid/urine , Hyperoxia/chemically induced , Hyperoxia/physiopathology , Hypoxia/physiopathology , Infant, Newborn , Infant, Premature , Plethysmography, Whole Body , Pulmonary Ventilation , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Rats , Receptor, Serotonin, 5-HT1A/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics , Respiratory Mechanics/physiology , Serotonin/blood , Serotonin/urine , Sleep Apnea Syndromes/metabolism , Sleep Apnea Syndromes/physiopathology , Sudden Infant Death
4.
Am J Physiol Regul Integr Comp Physiol ; 314(2): R216-R227, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29046314

ABSTRACT

The extracellular matrix (ECM) modulates brain maturation and plays a major role in regulating neuronal plasticity during critical periods of development. We examined 1) whether there is a critical postnatal period of ECM expression in brain stem cardiorespiratory control regions and 2) whether the attenuated hypoxic ventilatory response (HVR) following neonatal sustained (5 days) hypoxia [SH (11% O2, 24 h/day)] exposure is associated with altered ECM formation. The nucleus tractus solitarius (nTS), dorsal motor nucleus of the vagus, hypoglossal motor nucleus, cuneate nucleus, and area postrema were immunofluorescently processed for aggrecan and Wisteria floribunda agglutinin (WFA), a key proteoglycan of the ECM and the perineuronal net. From postnatal day ( P) 5 ( P5), aggrecan and WFA expression increased postnatally in all regions. We observed an abrupt increase in aggrecan expression in the nTS, a region that integrates and receives afferent inputs from the carotid body, between P10 and P15 followed by a distinct and transient plateau between P15 and P20. WFA expression in the nTS exhibited an analogous transient plateau, but it occurred earlier (between P10 and P15). SH between P11 and P15 attenuated the HVR (assessed at P16) and increased aggrecan (but not WFA) expression in the nTS, dorsal motor nucleus of the vagus, and area postrema. An intracisternal microinjection of chondroitinase ABC, an enzyme that digests chondroitin sulfate proteoglycans, rescued the HVR and the increased aggrecan expression. These data indicate that important stages of ECM formation take place in key brain stem respiratory neural control regions and appear to be associated with a heightened vulnerability to hypoxia.


Subject(s)
Brain Stem/metabolism , Extracellular Matrix/metabolism , Hypoxia/complications , Lung/innervation , Respiration , Respiratory Insufficiency/etiology , Age Factors , Aggrecans/metabolism , Animals , Animals, Newborn , Brain Stem/drug effects , Brain Stem/growth & development , Chondroitin ABC Lyase/administration & dosage , Disease Models, Animal , Extracellular Matrix/drug effects , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Morphogenesis , Plant Lectins/metabolism , Rats, Inbred Lew , Receptors, N-Acetylglucosamine/metabolism , Respiration/drug effects , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/physiopathology , Respiratory Insufficiency/prevention & control , Risk Factors
5.
Acta Neuropathol Commun ; 5(1): 42, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28578681

ABSTRACT

Bioactive lipids contribute to the pathophysiology of multiple sclerosis. Here, we show that lysophosphatidic acids (LPAs) are dysregulated in multiple sclerosis (MS) and are functionally relevant in this disease. LPAs and autotaxin, the major enzyme producing extracellular LPAs, were analyzed in serum and cerebrospinal fluid in a cross-sectional population of MS patients and were compared with respective data from mice in the experimental autoimmune encephalomyelitis (EAE) model, spontaneous EAE in TCR1640 mice, and EAE in Lpar2 -/- mice. Serum LPAs were reduced in MS and EAE whereas spinal cord LPAs in TCR1640 mice increased during the 'symptom-free' intervals, i.e. on resolution of inflammation during recovery hence possibly pointing to positive effects of brain LPAs during remyelination as suggested in previous studies. Peripheral LPAs mildly re-raised during relapses but further dropped in refractory relapses. The peripheral loss led to a redistribution of immune cells from the spleen to the spinal cord, suggesting defects of lymphocyte homing. In support, LPAR2 positive T-cells were reduced in EAE and the disease was intensified in Lpar2 deficient mice. Further, treatment with an LPAR2 agonist reduced clinical signs of relapsing-remitting EAE suggesting that the LPAR2 agonist partially compensated the endogenous loss of LPAs and implicating LPA signaling as a novel treatment approach. Graphical summary of lysophosphatidic signaling in multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Lysophospholipids/metabolism , Multiple Sclerosis/metabolism , Adolescent , Adult , Animals , Biomarkers/metabolism , Cohort Studies , Cross-Sectional Studies , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunologic Factors/pharmacology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Young Adult
6.
Respir Physiol Neurobiol ; 242: 45-51, 2017 08.
Article in English | MEDLINE | ID: mdl-28330778

ABSTRACT

Perinatal inflammation and infection are commonly associated with various respiratory morbidities in preterm infants including apnea of prematurity. In this study, we investigated whether pulmonary inflammation via intra-tracheal micro-injection of lipopolysaccharide (LPS) into neonatal rats modifies respiratory neural control via an IL-1ß receptor-dependent mechanism. Prior to an intra-tracheal micro-injection of LPS (1mg/kg), 10day old (Postnatal age, P10) rats received an intraperitoneal (i.p.) or intracisternal (i.c.) micro-injection of the IL-1ß receptor antagonist AF12198. Whole-body plethysmography was performed two hours later to assess the magnitude of the acute hypoxic (HVR) and hypercapnic (HCVR) ventilatory responses. Intra-tracheal LPS dose-dependently attenuated the acute HVR compared to saline (control) treated rats, whereas the HCVR was not affected. Pre-treatment with an i.c. (but not i.p.) micro-injection of AF12198 15min prior to LPS prevented the attenuated HVR. These data indicate that intrapulmonary inflammation affects brainstem respiratory neural pathways mediating the ventilatory response to acute hypoxia via an IL-1ß-dependent pathway. These findings are relevant to our understanding of the way that pulmonary inflammation may affect central neural mechanisms of respiratory insufficiency commonly seen in preterm infants.


Subject(s)
Brain Stem/immunology , Hypoxia/immunology , Interleukin-1beta/metabolism , Pneumonia/immunology , Respiration , Animals , Animals, Newborn , Brain Stem/drug effects , Central Nervous System Agents/pharmacology , Escherichia coli , Hypercapnia/immunology , Injections, Intraperitoneal , Lipopolysaccharides , Male , Microinjections , Neural Pathways/drug effects , Neural Pathways/immunology , Plethysmography , Proteins/pharmacology , Random Allocation , Rats, Sprague-Dawley , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Trachea
7.
J Physiol ; 594(11): 3079-94, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26659585

ABSTRACT

KEY POINTS: Neonatal sustained hypoxia exposure modifies brainstem microglia and serotonin expression. The altered brainstem neurochemistry is associated with impaired ventilatory responses to acute hypoxia and mortality. The deleterious effects of sustained hypoxia exposure can be prevented by an inhibitor of activated microglia. These observations demonstrate a potential cause of the brainstem serotonin abnormalities thought to be involved in sudden infant death syndrome. ABSTRACT: We showed previously that the end of the second postnatal week (days P11-15) represents a period of development during which the respiratory neural control system exhibits a heightened vulnerability to sustained hypoxia (SH, 11% O2 , 5 days) exposure. In the current study, we investigated whether the vulnerability to SH during the same developmental time period is associated with changes in brainstem serotonin (5-HT) expression and whether it can be prevented by the microglia inhibitor minocycline. Using whole-body plethysmography, SH attenuated the acute (5 min) hypoxic ventilatory response (HVR) and caused a high incidence of mortality compared to normoxia rats. SH also increased microglia cell numbers and decreased 5-HT immunoreactivity in the nucleus of the solitary tract (nTS) and dorsal motor nucleus of the vagus (DMNV). The attenuated HVR, mortality, and changes in nTS and DMNV immunoreactivity was prevented by minocycline (25 mg kg(-1) /2 days during SH). These data demonstrate that the 5-HT abnormalities in distinct respiratory neural control regions can be initiated by prolonged hypoxia exposure and may be modulated by microglia activity. These observations share several commonalities with the risk factors thought to underlie the aetiology of sudden infant death syndrome, including: (1) a vulnerable neonate; (2) a critical period of development; (3) evidence of hypoxia; (4) brainstem gliosis (particularly the nTS and DMNV); and (5) 5-HT abnormalities.


Subject(s)
Brain Stem/metabolism , Hypoxia/metabolism , Microglia/metabolism , Serotonin/biosynthesis , Age Factors , Animals , Animals, Newborn , Brain Stem/drug effects , Female , Gene Expression , Hypoxia/drug therapy , Hypoxia/genetics , Microglia/drug effects , Minocycline/pharmacology , Minocycline/therapeutic use , Pregnancy , Rats , Rats, Inbred Lew , Serotonin/genetics
8.
Respir Physiol Neurobiol ; 205: 28-36, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25266393

ABSTRACT

We investigated whether pre-treatment with neonatal sustained hypoxia (SH) prior to chronic intermittent hypoxia (SH+CIH) would modify in vitro carotid body (CB) chemoreceptor activity and the excitability of neurons in the caudal nucleus of the solitary tract (nTS). Sustained hypoxia followed by CIH exposure simulates an oxygen paradigm experienced by extremely premature infants who developed persistent apnea. Rat pups were treated with 5 days of SH (11% O2) from postnatal age 1 (P1) followed by 10 days of subsequent chronic intermittent hypoxia (CIH, 5% O2/5 min, 8 h/day, between P6 and P15) as described previously (Mayer et al., Respir. Physiol. Neurobiol. 187(2): 167-75, 2013). At the end of SH+CIH exposure (P16), basal firing frequency was enhanced, and the hypoxic sensory response of single unit CB chemoafferents was attenuated. Further, basal firing frequency and the amplitude of evoked excitatory post-synaptic currents (ESPC's) of nTS neurons was augmented compared to age-matched rats raised in normoxia. These effects were unique to SH+CIH exposure as neither SH or CIH alone elicited any comparable effect on chemoafferent activity or nTS function. These data indicated that pre-treatment with neonatal SH prior to CIH exposure uniquely modified mechanisms of peripheral (CB) and central (nTS) neural function in a way that would be expected to disturb the ventilatory response to acute hypoxia.


Subject(s)
Carotid Body/physiopathology , Hypoxia/physiopathology , Solitary Nucleus/physiopathology , Animals , Animals, Newborn , Disease Models, Animal , Male , Rats , Rats, Inbred Lew
9.
J Appl Physiol (1985) ; 116(5): 514-21, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24371020

ABSTRACT

The first postnatal weeks represent a period of development in the rat during which the respiratory neural control system may be vulnerable to aberrant environmental stressors. In the present study, we investigated whether sustained hypoxia (SH; 11% O2) exposure starting at different postnatal ages differentially modifies the acute hypoxic (HVR) and hypercapnic ventilatory response (HCVR). Three different groups of rat pups were exposed to 5 days of SH, starting at either postnatal age 1 (SH1-5), 11 (SH11-15), or 21 (SH21-25) days. Whole body plethysmography was used to assess the HVR and HCVR the day after SH exposure ended. The primary results indicated that 1) the HVR and HCVR of SH11-15 rats were absent or attenuated (respectively) compared with age-matched rats raised in normoxia; 2) there was a profoundly high (∼84% of pups) incidence of unexplained mortality in the SH11-15 rats; and 3) these phenomena were unique to the SH11-15 group with no comparable effect of the SH exposure on the HVR, HCVR, or mortality in the younger (SH1-5) or older (SH21-25) rats. These results share several commonalities with the risk factors thought to underlie the etiology of sudden infant death syndrome, including 1) a vulnerable neonate; 2) a critical period of development; and 3) an environmental stressor.


Subject(s)
Asphyxia Neonatorum/physiopathology , Respiratory Mechanics/physiology , Aging/physiology , Animals , Animals, Newborn , Asphyxia Neonatorum/mortality , Body Weight/physiology , Carbon Dioxide/metabolism , Humans , Hypercapnia/physiopathology , Infant , Male , Metabolism/physiology , Oxygen Consumption , Plethysmography , Rats , Rats, Inbred Lew , Sudden Infant Death , Tidal Volume/physiology
10.
Respir Physiol Neurobiol ; 187(2): 167-75, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23562917

ABSTRACT

Neonatal chronic intermittent hypoxia (CIH) enhances the ventilatory sensitivity to acute hypoxia (acute hypoxic ventilatory response, HVR), whereas sustained hypoxia (SH) can have the opposite effect. Therefore, we investigated whether neonatal rats pre-treated with SH prior to CIH exhibit a modified HVR. Rat pups were exposed to CIH (5% O2/5min, 8h/day) between 6 and 15 days of postnatal age (P6-15) after pre-treatment with either normoxia or SH (11% O2; P1-5). Using whole-body plethysmography, the acute (5min, 10% O2) HVR at P16 (1 day post-CIH) was unchanged following CIH (67.9±6.7% above baseline) and also SH (58.8±10.5%) compared to age-matched normoxic rats (54.7±6.3%). In contrast, the HVR was attenuated (16.5±6.0%) in CIH exposed rats pre-treated with SH. These data suggest that while neonatal SH and CIH alone have little effect on the magnitude of the acute HVR, their combined effects impose a synergistic disturbance to postnatal development of the HVR. These data could provide important insight into the consequences of not maintaining adequate levels of oxygen saturation during the early neonatal period, especially in vulnerable preterm infants susceptible to frequent bouts of hypoxemic events (CIH) that are commonly associated with apnea of prematurity.


Subject(s)
Hypoxia/complications , Lung Diseases/etiology , Pulmonary Ventilation/physiology , Age Factors , Animals , Animals, Newborn , Female , Hypercapnia/physiopathology , Male , Oxygen Consumption , Plethysmography, Whole Body , Pregnancy , Pulmonary Gas Exchange , Rats , Rats, Inbred Lew
12.
Psychiatry ; 49(4): 350-8, 1986 Nov.
Article in English | MEDLINE | ID: mdl-3101089

ABSTRACT

In a previous report, the authors identified four dimensions of patient pathology associated with treatment difficulty: withdrawn psychoticism, character pathology, violence-agitation and suicidal-depressed behavior. In a subsequent study, they linked these dimensions to patterns of countertransference. The present research extends the two prior reports by examining the relations of the patient pathology dimensions to staff members' dissatisfaction with four areas of treatment: interpersonal approaches, structure and control, quality of teamwork, and medication. The major findings are: withdrawn psychoticism primarily relates to dissatisfaction with interpersonal treatment approaches; character pathology entails dissatisfaction with the level of structure and control; violence-agitation poses particular problems for teamwork; and suicidal-depressed behavior is unrelated to dissatisfaction with any dimension of treatment. The authors propose that these various problems in treatment are, in part, mediated by patterns of countertransference which they described in the prior paper. These findings should help staff members to focus their attention on areas of treatment in which problems are bound to arise in work with different types of difficult patients.


Subject(s)
Hospitals, Psychiatric , Mental Disorders/therapy , Adolescent , Adult , Countertransference , Depressive Disorder/therapy , Female , Hospitalization , Humans , Long-Term Care , Male , Mental Disorders/psychology , Middle Aged , Personality Disorders/therapy , Psychotic Disorders/therapy , Suicide Prevention
13.
Hosp Community Psychiatry ; 37(9): 923-8, 1986 Sep.
Article in English | MEDLINE | ID: mdl-3758976

ABSTRACT

Countertransference among hospital staff was investigated as part of ongoing research on difficult-to-treat psychiatric hospital patients. Staff's ratings of their emotional reactions to 127 patients on long-term units were analyzed by factor analysis, and the resulting factors were correlated by discipline with patient problem behaviors. Among the conclusions were that different forms of psychopathology elicit characteristic patterns of emotional reaction from staff; that some dimensions of psychopathology, particularly suicidal-depressed behavior and violence-agitation, elicit different emotional reactions among different disciplines, thus laying the groundwork for division among staff; and that the more difficult the process of hospital treatment, the more likely staff will experience a variety of emotions.


Subject(s)
Countertransference , Hospitals, Psychiatric , Adolescent , Adult , Character , Emotions , Female , Humans , Male , Mental Disorders/psychology , Mental Disorders/therapy , Middle Aged , Nurse-Patient Relations , Occupational Therapy , Professional-Patient Relations , Psychiatry , Social Work , Suicide, Attempted/psychology , Violence
14.
N M Dent J ; 19(1): 17-8, 1968 May.
Article in English | MEDLINE | ID: mdl-5242621
SELECTION OF CITATIONS
SEARCH DETAIL
...