Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 71(2): 195-206, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38597725

RESUMEN

Extreme heat caused by climate change is increasing the transmission of infectious diseases, resulting in a sharp rise in heat-related illness and mortality. Understanding the mechanistic link between heat, inflammation, and disease is thus important for public health. Thermal hyperpnea, and consequent respiratory alkalosis, is crucial in febrile seizures and convulsions induced by heat stress in humans. Here, we address what causes thermal hyperpnea in neonates and how it is affected by inflammation. Transient receptor potential cation channel subfamily V member 1 (TRPV1), a heat-activated channel, is sensitized by inflammation and modulates breathing and thus may play a key role. To investigate whether inflammatory sensitization of TRPV1 modifies neonatal ventilatory responses to heat stress, leading to respiratory alkalosis and an increased susceptibility to hyperthermic seizures, we treated neonatal rats with bacterial LPS, and breathing, arterial pH, in vitro vagus nerve activity, and seizure susceptibility were assessed during heat stress in the presence or absence of a TRPV1 antagonist (AMG-9810) or shRNA-mediated TRPV1 suppression. LPS-induced inflammatory preconditioning lowered the threshold temperature and latency of hyperthermic seizures. This was accompanied by increased tidal volume, minute ventilation, expired CO2, and arterial pH (alkalosis). LPS exposure also elevated vagal spiking and intracellular calcium concentrations in response to hyperthermia. TRPV1 inhibition with AMG-9810 or shRNA reduced the LPS-induced susceptibility to hyperthermic seizures and altered the breathing pattern to fast shallow breaths (tachypnea), making each breath less efficient and restoring arterial pH. These results indicate that inflammation exacerbates thermal hyperpnea-induced respiratory alkalosis associated with increased susceptibility to hyperthermic seizures, primarily mediated by TRPV1 localized to vagus neurons.


Asunto(s)
Inflamación , Convulsiones Febriles , Canales Catiónicos TRPV , Convulsiones Febriles/fisiopatología , Convulsiones Febriles/metabolismo , Animales , Canales Catiónicos TRPV/metabolismo , Inflamación/metabolismo , Ratas , Respuesta al Choque Térmico , Animales Recién Nacidos , Lipopolisacáridos/farmacología , Nervio Vago/fisiopatología , Ratas Sprague-Dawley , Alcalosis Respiratoria/metabolismo , Alcalosis Respiratoria/fisiopatología , Hipertermia/metabolismo , Hipertermia/fisiopatología
2.
BMC Anesthesiol ; 24(1): 323, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261779

RESUMEN

BACKGROUND: The perioperative management of patients with chronic cough or cough hypersensitivity syndrome and its sometimes severe effects is currently under-researched and under-reported. CASE PRESENTATION: A 46-year-old female patient with a history of chronic cough and Cough Hypersensitivity Syndrome. After laparoscopic hiatoplasty and anterior fundoplication under general anesthesia, experienced a pronounced exacerbation of coughing symptoms. Despite prompt and extensive treatment involving antitussives, inhalants, anxiolytics, and sedatives, the symptoms remained uncontrollable. Within a few hours, the patient developed a respiratory alkalosis with severe and life-threatening electrolyte shift (pH 7.705, pCO2 1.72 kPa, K+ 2.1 mmol/l). Lactatemia lasted for more than 12 hours with values up to 6.6 mmol/l. Acute bleeding, pneumothorax, and an acute cardiac event were ruled out. Deep analgosedation and inhalation of high-percentage local anesthetics were necessary to manage the clinical symptoms. CONCLUSIONS: This case highlights the challenging nature of chronic cough and hypersensitivity syndrome perioperatively. A tailored anesthesiologic approach, exclusion of other provoking medical problems, and knowledge of possible management and treatment options are key.


Asunto(s)
Alcalosis Respiratoria , Tos , Complicaciones Posoperatorias , Humanos , Femenino , Persona de Mediana Edad , Desequilibrio Hidroelectrolítico , Anestesia General/métodos , Anestesia General/efectos adversos , Hiperlactatemia , Fundoplicación/efectos adversos , Síndrome , Enfermedades Respiratorias
3.
J Physiol ; 601(24): 5601-5616, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37975212

RESUMEN

Passive hyperthermia causes cerebral hypoperfusion primarily from heat-induced respiratory alkalosis. However, despite the cerebral hypoperfusion, it is possible that the mild alkalosis might help to attenuate cerebral inflammation. In this study, the cerebral exchange of extracellular vesicles (microvesicles), which are known to elicit pro-inflammatory responses when released in conditions of stress, were examined in hyperthermia with and without respiratory alkalosis. Ten healthy male adults were heated passively, using a warm water-perfused suit, up to core temperature + 2°C. Blood samples were taken from the radial artery and internal jugular bulb. Microvesicle concentrations were determined in platelet-poor plasma via cells expressing CD62E (activated endothelial cells), CD31+ /CD42b- (apoptotic endothelial cells), CD14 (monocytes) and CD45 (pan-leucocytes). Cerebral blood flow was measured via duplex ultrasound of the internal carotid and vertebral arteries to determine cerebral exchange kinetics. From baseline to poikilocapnic (alkalotic) hyperthermia, there was no change in microvesicle concentration from any cell origin measured (P-values all >0.05). However, when blood CO2 tension was normalized to baseline levels in hyperthermia, there was a marked increase in cerebral uptake of microvesicles expressing CD62E (P = 0.028), CD31+ /CD42b- (P = 0.003) and CD14 (P = 0.031) compared with baseline, corresponding to large increases in arterial but not jugular venous concentrations. In a subset of seven participants who underwent hypercapnia and hypocapnia in the absence of heating, there was no change in microvesicle concentrations or cerebral exchange, suggesting that hyperthermia potentiated the CO2 /pH-mediated cerebral uptake of microvesicles. These data provide insight into a potential beneficial role of respiratory alkalosis in heat stress. KEY POINTS: The hyperthermia-induced hyperventilatory response is observed in most humans, despite causing potentially harmful reductions in cerebral blood flow. We tested the hypothesis that the respiratory-induced alkalosis is associated with lower circulating microvesicle concentrations, specifically in the brain, despite the reductions in blood flow. At core temperature + 2°C with respiratory alkalosis, microvesicles derived from endothelial cells, monocytes and leucocytes were at concentrations similar to baseline in the arterial and cerebral venous circulation, with no changes in cross-brain microvesicle kinetics. However, when core temperature was increased by 2°C with CO2 /pH normalized to resting levels, there was a marked cerebral uptake of microvesicles derived from endothelial cells and monocytes. The CO2 /pH-mediated alteration in cerebral microvesicle uptake occurred only in hyperthermia. These new findings suggest that the heat-induced hyperventilatory response might serve a beneficial role by preventing potentially inflammatory microvesicle uptake in the brain.


Asunto(s)
Alcalosis Respiratoria , Hipertermia Inducida , Adulto , Humanos , Masculino , Hipocapnia , Células Endoteliales/fisiología , Dióxido de Carbono , Hiperventilación , Circulación Cerebrovascular/fisiología
4.
Am J Kidney Dis ; 82(3): 347-359, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37341662

RESUMEN

The respiratory system plays an integral part in maintaining acid-base homeostasis. Normal ventilation participates in the maintenance of an open buffer system, allowing for excretion of CO2 produced from the interaction of nonvolatile acids and bicarbonate. Quantitatively of much greater importance is the excretion of CO2 derived from volatile acids produced from the complete oxidation of fat and carbohydrate. A primary increase in CO2 tension of body fluids is the cause of respiratory acidosis and develops most commonly from one or more of the following: (1) disorders affecting gas exchange across the pulmonary capillary, (2) disorders of the chest wall and the respiratory muscles, and/or (3) inhibition of the medullary respiratory center. Respiratory alkalosis or primary hypocapnia is most commonly caused by disorders that increase alveolar ventilation and is defined by an arterial partial pressure of CO2 <35 mm Hg with subsequent alkalization of body fluids. Both disorders can lead to life-threatening complications, making it of paramount importance for the clinician to have a thorough understanding of the cause and treatment of these acid-base disturbances.


Asunto(s)
Acidosis Respiratoria , Alcalosis Respiratoria , Alcalosis , Humanos , Alcalosis Respiratoria/diagnóstico , Alcalosis Respiratoria/etiología , Dióxido de Carbono , Hipocapnia , Bicarbonatos , Alcalosis/etiología , Alcalosis/complicaciones , Concentración de Iones de Hidrógeno , Equilibrio Ácido-Base
5.
J Clin Monit Comput ; 37(4): 1115-1118, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37138040

RESUMEN

The neuromuscular blocking potency of rocuronium varies with respiratory pH changes, increasing at lower pH and decreasing at higher pH; thus, hyperventilation-induced respiratory alkalosis is expected to decrease the potency of rocuronium. We report a case of anesthetic management of modified electroconvulsive therapy (m-ECT) for a patient monitored with electromyography-based neuromuscular monitoring during two patterns of ventilation to elucidate their relationship and propose the possible mechanisms underlying the effects by computational simulations. Case presentation: The patient was a 25-year-old man with schizophrenia. In m-ECT, hyperventilation may be used to produce longer seizures. We compared the neuromuscular monitoring data recorded during hyperventilation and during normal ventilation while receiving the same dose of rocuronium. Despite receiving the same dose of rocuronium, the time required for the first twitch to decrease to 80% of the control value was delayed in hyperventilation compared to normal ventilation. Conclusions: This case report and computational simulation suggest that respiratory alkalosis might delay the action of rocuronium. It is necessary to consider the delayed action of rocuronium when hyperventilation is performed.


Asunto(s)
Alcalosis Respiratoria , Bloqueo Neuromuscular , Fármacos Neuromusculares no Despolarizantes , Masculino , Humanos , Adulto , Rocuronio , Androstanoles , Hiperventilación
6.
Br J Anaesth ; 129(5): 726-733, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36096944

RESUMEN

BACKGROUND: Acid-base status in full-term pregnant women is characterised by hypocapnic alkalosis. Whether this respiratory alkalosis is primary or consequent to changes in CSF electrolytes is not clear. METHODS: We enrolled third-trimester pregnant women (pregnant group) and healthy, non-pregnant women of childbearing age (controls) undergoing spinal anaesthesia for Caesarean delivery and elective surgery, respectively. Electrolytes, strong ion difference (SID), partial pressure of carbon dioxide ( [Formula: see text] ), and pH were measured in simultaneously collected CSF and arterial blood samples. RESULTS: All pregnant women (20) were hypocapnic, whilst only four (30%) of the controls (13) had an arterial [Formula: see text] <4.7 kPa (P<0.001). The incidence of hypocapnic alkalosis was higher in the pregnant group (65% vs 8%; P=0.001). The CSF-to-plasma Pco2 difference was significantly higher in pregnant women (1.5 [0.3] vs 1.0 [0.4] kPa; P<0.001), mainly because of a decrease in arterial Pco2 (3.9 [0.3] vs 4.9 [0.5] kPa; P<0.001). Similarly, the CSF-to-plasma difference in SID was less negative in pregnant women (-7.8 [1.4] vs -11.4 [2.3] mM; P<0.001), mainly because of a decreased arterial SID (31.5 [1.2] vs 36.1 [1.9] mM; P<0.001). The major determinant of the reduced plasma SID of pregnant women was a relative increase in plasma chloride compared with sodium. CONCLUSIONS: Primary hypocapnic alkalosis characterises third-trimester pregnant women leading to chronic acid-base adaptations of CSF and plasma. The compensatory SID reduction, mainly sustained by an increase in chloride concentration, is more pronounced in plasma than in CSF, as the decrease in Pco2 is more marked in this compartment. CLINICAL TRIAL REGISTRATION: NCT03496311.


Asunto(s)
Alcalosis , Femenino , Humanos , Embarazo , Equilibrio Ácido-Base , Bicarbonatos , Dióxido de Carbono , Cloruros , Electrólitos , Concentración de Iones de Hidrógeno , Tercer Trimestre del Embarazo , Sodio
7.
Medicina (Kaunas) ; 58(3)2022 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-35334595

RESUMEN

Dyspnea, shortness of breath, and chest pain are frequent symptoms of post-COVID syndrome (PCS). These symptoms are unrelated to organ damage in most patients after mild acute COVID infection. Hyperventilation has been identified as a cause of exercise-induced dyspnea in PCS. Since there is a broad overlap in symptomatology with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), causes for dyspnea and potential consequences can be deduced by a stringent application of assumptions made for ME/CFS in our recent review papers. One of the first stimuli of respiration in exercise is caused by metabolic feedback via skeletal muscle afferents. Hyperventilation in PCS, which occurs early on during exercise, can arise from a combined disturbance of a poor skeletal muscle energetic situation and autonomic dysfunction (overshooting respiratory response), both found in ME/CFS. The exaggerated respiratory response aggravating dyspnea does not only limit the ability to exercise but further impairs the muscular energetic situation: one of the buffering mechanisms to respiratory alkalosis is a proton shift from intracellular to extracellular space via the sodium-proton-exchanger subtype 1 (NHE1), thereby loading cells with sodium. This adds to two other sodium loading mechanisms already operative, namely glycolytic metabolism (intracellular acidosis) and impaired Na+/K+ATPase activity. High intracellular sodium has unfavorable effects on mitochondrial calcium and metabolism via sodium-calcium-exchangers (NCX). Mitochondrial calcium overload by high intracellular sodium reversing the transport mode of NCX to import calcium is a key driver for fatigue and chronification. Prevention of hyperventilation has a therapeutic potential by keeping intracellular sodium below the threshold where calcium overload occurs.


Asunto(s)
COVID-19 , Síndrome de Fatiga Crónica , COVID-19/complicaciones , Disnea/etiología , Ejercicio Físico , Síndrome de Fatiga Crónica/etiología , Síndrome de Fatiga Crónica/terapia , Humanos , Sodio
8.
Indian J Crit Care Med ; 26(5): 643-645, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35719437

RESUMEN

Ventilator auto triggering is an avoidable complication in ventilators, if left unnoticed can lead to deleterious effects. There are various causes for ventilator auto triggering. Though rare, there are some cardiac causes for inadvertent ventilator triggering. We report a case of 44-years-old male paced with atrial epicardial wires postcoronary artery bypass. The wires were close to the right phrenic nerve, causing the right diaphragm to contract in synchronization with the heartbeat. This caused ventilator auto triggering and ended up delivering inadvertent breaths. The pacemaker output was immediately reduced to the required minimum to continue pacing the heart and decrease phrenic nerve stimulation. This caused immediate changes in ventilator waveform and auto triggering was completely stopped and the patient could be successfully weaned off the ventilator. This case report emphasizes he need for timely recognition of alteration in ventilator waveforms and early intervention to avert any untoward events. How to cite this article: Mukunthan MN, Bhardwaj V. Cardiac Pacing a Rare Cause of Ventilator Auto Triggering. Indian J Crit Care Med 2022;26(5):643-645.

9.
Exp Physiol ; 106(1): 175-190, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347666

RESUMEN

NEW FINDINGS: What is the central question of this study? The pathophysiology of acute mountain sickness (AMS), involving the respiratory, renal and cerebrovascular systems, remains poorly understood. How do the early adaptations in these systems during a simulated altitude of 5000 m relate to AMS risk? What is the main finding and its importance? The rate of blood alkalosis and cerebral artery dilatation predict AMS severity during the first 10 h of exposure to a simulated altitude of 5000 m. Slow metabolic compensation by the kidneys of respiratory alkalosis attributable to a brisk breathing response together with excessive brain blood vessel dilatation might be involved in early development of AMS. ABSTRACT: The complex pathophysiology of acute mountain sickness (AMS) remains poorly understood and is likely to involve maladaptive responses of the respiratory, renal and cerebrovascular systems to hypoxia. Using stepwise linear regression, we tested the hypothesis that exacerbated respiratory alkalosis, as a result of a brisk ventilatory response, sluggish renal compensation in acute hypoxia and dysregulation of cerebral perfusion predict AMS severity. We assessed the Lake Louise score (LLS, an index of AMS severity), fluid balance, ventilation, venous pH, bicarbonate, sodium and creatinine concentrations, body weight, urinary pH and cerebral blood flow [internal carotid artery (ICA) and vertebral artery (VA) blood flow and diameter], in 27 healthy individuals (13 women) throughout 10 h exposures to normobaric normoxia (fraction of inspired O2 = 0.21) and normobaric hypoxia (fraction of inspired O2 = 0.117, simulated 5000 m) in a randomized, single-blinded manner. In comparison to normoxia, hypoxia increased the LLS, ventilation, venous and urinary pH, and blood flow and diameter in the ICA and VA, while venous concentrations of both bicarbonate and creatinine were decreased (P < 0.001 for all). There were significant correlations between AMS severity and the rates of change in blood pH, sodium concentration and VA diameter and more positive fluid balance (P < 0.05). Stepwise regression found increased blood pH [beta coefficient (ß) = 0.589, P < 0.001] and VA diameter (ß = 0.418, P = 0.008) to be significant predictors of AMS severity in our cohort [F(2, 20) = 16.1, R2  = 0.617, P < 0.001, n = 24], accounting for 62% of the variance in peak LLS. Using classic regression variable selection, our data implicate the degree of respiratory alkalosis and cerebrovascular dilatation in the early stages of AMS development.


Asunto(s)
Aclimatación/fisiología , Mal de Altura/fisiopatología , Altitud , Hipoxia/fisiopatología , Arteria Cerebral Posterior/fisiopatología , Enfermedad Aguda , Adolescente , Adulto , Encéfalo/metabolismo , Femenino , Hemodinámica/fisiología , Humanos , Masculino , Oxígeno/metabolismo , Arteria Cerebral Posterior/metabolismo , Adulto Joven
10.
Pacing Clin Electrophysiol ; 44(12): 2115-2118, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34390012

RESUMEN

Bidirectional ventricular tachycardia (BVT) is a rare arrhythmia that is generally observed in patients with catecholaminergic ventricular tachycardia or digoxin overdose. Herein, we present a case of BVT and electrical storm (ES) in an acute ischemic heart failure patient that is typically induced by hypokalemia. The patient was in invasive mechanical ventilator (MV) support and hypokalemia was related to acute respiratory alkalosis and that caused refractory hypokalemia despite intravenous (IV) potassium replacement. We also discuss our approach to solve refractory hypokalemia caused by respiratory alkalosis.


Asunto(s)
Alcalosis Respiratoria/complicaciones , Insuficiencia Cardíaca/complicaciones , Hipopotasemia/complicaciones , Taquicardia/etiología , Anciano , Alcalosis Respiratoria/terapia , Electrocardiografía , Femenino , Insuficiencia Cardíaca/terapia , Humanos , Hipopotasemia/terapia , Taquicardia/terapia
11.
Int Arch Occup Environ Health ; 94(7): 1627-1636, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33646335

RESUMEN

OBJECTIVES: During the COVID-19 pandemic, healthcare professionals are recommended to use PPE to prevent the transmission of disease. Healthcare workers who use N95 FFR, which has an important place, experience complaints such as headache and dizziness. In this study, we plan to find the cause of these complaints and aim to clarify whether they are associated with the use of N95 mask. METHOD: Healthcare workers first put on a surgical mask for at least 1 h and a maximum of 4 h, this process was then repeated on another day with the same workers wearing N95 masks. After removing the mask, capillary blood gases were taken and a questionnaire was given. RESULTS: Thirty-four participants over the age of 18 were included in the study; 19 participants were female (56%) and 15 male (44%). The results of the capillary blood gas analysis after the use of surgical mask and N95 mask, respectively: pH: 7.43 ± 0.03; 7.48 ± 0.04 (p < 0.001); pCO2: 37.33 ± 8.81; 28.46 ± 7.77 mmHg (p < 0.001); HCO3: 24.92 ± 2.86; 23.73 ± 3.29 mmol/L (p = 0.131); Base excess (BE): 1.40 (- 3.90-3.10); - 2.68 (- 4.50-1.20) [median (Q1-Q3)] (p = 0.039); lactate: 1.74 ± 0.68; 1.91 ± 0.61 (p = 0314). Headache, attention deficit and difficulty in concentrating were significantly higher after using N95 mask. CONCLUSION: Respiratory alkalosis and hypocarbia were detected after the use of N95. Acute respiratory alkalosis can cause headache, anxiety, tremor, muscle cramps. In this study, it was quantitatively shown that the participants' symptoms were due to respiratory alkalosis and hypocarbia.


Asunto(s)
COVID-19/epidemiología , Mareo/etiología , Cefalea/etiología , Respiradores N95/efectos adversos , Adulto , Factores de Edad , Análisis de los Gases de la Sangre , COVID-19/prevención & control , Femenino , Personal de Salud , Hospitales Universitarios , Humanos , Concentración de Iones de Hidrógeno , Masculino , Máscaras/efectos adversos , Pandemias , SARS-CoV-2 , Factores Sexuales , Factores Socioeconómicos
12.
Neurocrit Care ; 30(2): 405-413, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30386962

RESUMEN

BACKGROUND: Hypocapnia induces cerebral vasoconstriction leading to a decrease in cerebral blood flow, which might precipitate cerebral ischemia. Hypocapnia can be intentional to treat intracranial hypertension or unintentional due to a spontaneous hyperventilation (SHV). SHV is frequent after subarachnoid hemorrhage. However, it is understudied in patients with severe traumatic brain injury (TBI). The objective of this study was to describe the incidence and consequences on outcome of SHV after severe TBI. METHODS: We conducted a retrospective, observational study including all intubated TBI patients admitted in the trauma center and still comatose 24 h after the withdrawal of sedation. SHV was defined by the presence of at least one arterial blood gas (ABG) with both PaCO2 < 35 mmHg and pH > 7.45. Patient characteristics and outcome were extracted from a prospective registry of all intubated TBI admitted in the intensive care unit. ABG results were retrieved from patient files. A multivariable logistic regression model was developed to determine factors independently associated with unfavorable outcome (defined as a Glasgow Outcome Scale between 1 and 3) at 6-month follow-up. RESULTS: During 7 years, 110 patients fully respecting inclusion criteria were included. The overall incidence of SHV was 69.1% (95% CI [59.9-77]). Patients with SHV were more severely injured (median head AIS score (5 [4-5] vs. 4 [4-5]; p = 0.016)) and exhibited an elevated morbidity during their stay. The proportion of patients with an unfavorable functional neurologic outcome was significantly higher in patients with SHV: 40 (52.6%) versus 6 (17.6%), p = 0.0006. After adjusting for confounders, SHV remains an independent factor associated with unfavorable outcome at the 6-month follow-up (OR 4.1; 95% CI [1.2-14.4]). CONCLUSIONS: SHV is common in patients with a persistent coma after a severe TBI (overall rate: 69%) and was independently associated with unfavorable outcome at 6-month follow-up.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Coma/etiología , Hiperventilación/etiología , Hipocapnia/etiología , Sistema de Registros , Adulto , Alcalosis Respiratoria/epidemiología , Alcalosis Respiratoria/etiología , Lesiones Traumáticas del Encéfalo/epidemiología , Coma/epidemiología , Femenino , Estudios de Seguimiento , Escala de Consecuencias de Glasgow , Humanos , Hiperventilación/epidemiología , Hipocapnia/epidemiología , Puntaje de Gravedad del Traumatismo , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Estudios Retrospectivos , Adulto Joven
13.
J Physiol ; 596(24): 6191-6203, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30267579

RESUMEN

KEY POINTS: Ascent to high altitude imposes an acid-base challenge in which renal compensation is integral for maintaining pH homeostasis, facilitating acclimatization and helping prevent mountain sicknesses. The time-course and extent of plasticity of this important renal response during incremental ascent to altitude is unclear. We created a novel index that accurately quantifies renal acid-base compensation, which may have laboratory, fieldwork and clinical applications. Using this index, we found that renal compensation increased and plateaued after 5 days of incremental altitude exposure, suggesting plasticity in renal acid-base compensation mechanisms. The time-course and extent of plasticity in renal responsiveness may predict severity of altitude illness or acclimatization at higher or more prolonged stays at altitude. ABSTRACT: Ascent to high altitude, and the associated hypoxic ventilatory response, imposes an acid-base challenge, namely chronic hypocapnia and respiratory alkalosis. The kidneys impart a relative compensatory metabolic acidosis through the elimination of bicarbonate (HCO3- ) in urine. The time-course and extent of plasticity of the renal response during incremental ascent is unclear. We developed an index of renal reactivity (RR), indexing the relative change in arterial bicarbonate concentration ([HCO3- ]a ) (i.e. renal response) against the relative change in arterial pressure of CO2 ( PaCO2 ) (i.e. renal stimulus) during incremental ascent to altitude ( Δ[HCO3-]a/ΔPaCO2 ). We aimed to assess whether: (i) RR magnitude was inversely correlated with relative changes in arterial pH (ΔpHa ) with ascent and (ii) RR increased over time and altitude exposure (i.e. plasticity). During ascent to 5160 m over 10 days in the Nepal Himalaya, arterial blood was drawn from the radial artery for measurement of blood gas/acid-base variables in lowlanders at 1045/1400 m and after 1 night of sleep at 3440 m (day 3), 3820 m (day 5), 4240 m (day 7) and 5160 m (day 10) during ascent. At 3820 m and higher, RR significantly increased and plateaued compared to 3440 m (P < 0.04), suggesting plasticity in renal acid-base compensations. At all altitudes, we observed a strong negative correlation (r ≤ -0.71; P < 0.001) between RR and ΔpHa from baseline. Renal compensation plateaued after 5 days of altitude exposure, despite subsequent exposure to higher altitudes. The time-course, extent of plasticity and plateau in renal responsiveness may predict severity of altitude illness or acclimatization at higher or more prolonged stays at altitude.


Asunto(s)
Aclimatación/fisiología , Equilibrio Ácido-Base , Altitud , Bicarbonatos/metabolismo , Hipocapnia/metabolismo , Hipoxia/metabolismo , Adulto , Humanos , Masculino
14.
Metab Brain Dis ; 33(1): 191-199, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29116603

RESUMEN

Leigh syndrome (LS), subacute necrotizing encephalomyelopathy is caused by various genetic defects, including m.9185T>C MTATP6 variant. Mechanism of LS development remains unknown. We report on the acid-base status of three patients with m.9185T>C related LS. At the onset, it showed respiratory alkalosis, reflecting excessive respiration effort (hyperventilation with low pCO2). In patient 1, the deterioration occurred in temporal relation to passive oxygen therapy. To the contrary, on the recovery, she demonstrated a relatively low respiratory drive, suggesting that a "hypoventilation" might be beneficial for m.9185T>C carriers. As long as circumstances of the development of LS have not been fully explained, we recommend to counteract hyperventilation and carefully dose oxygen in patients with m.9185T>C related LS.


Asunto(s)
Hiperventilación/genética , Enfermedad de Leigh/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Mutación/genética , Adulto , Alcalosis Respiratoria/genética , Niño , Preescolar , Humanos , Hiperventilación/diagnóstico , Enfermedad de Leigh/diagnóstico
15.
Asian-Australas J Anim Sci ; 31(2): 263-269, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28823132

RESUMEN

OBJECTIVE: Heat stress (HS) triggers oxidative stress and respiratory alkalosis in pigs. The objective of this experiment was to study whether a short-term supranutritional amount of dietary vitamin E (VE) can mitigate oxidative stress and respiratory alkalosis in heat-stressed pigs. METHODS: A total of 24 pigs were given either a control diet (17 IU/kg VE) or a high VE (200 IU/kg VE; HiVE) diet for 14 d, then exposed to thermoneutral (TN; 20°C, 45% humidity) or HS (35°C, 35% to 45% humidity, 8 h daily) conditions for 7 d. Respiration rate and rectal temperature were measured three times daily during the thermal exposure. Blood gas variables and oxidative stress markers were studied in blood samples collected on d 7. RESULTS: Although HiVE diet did not affect the elevated rectal temperature or respiration rate observed during HS, it alleviated (all p<0.05 for diet×temperature) the loss of blood CO2 partial pressure and bicarbonate, as well as the increase in blood pH in the heat-stressed pigs. The HS reduced (p = 0.003) plasma biological antioxidant potential (BAP) and tended to increase (p = 0.067) advanced oxidized protein products (AOPP) in the heat-stressed pigs, suggesting HS triggers oxidative stress. The HiVE diet did not affect plasma BAP or AOPP. Only under TN conditions the HiVE diet reduced the plasma reactive oxygen metabolites (p<0.05 for diet× temperature). CONCLUSION: A short-term supplementation with 200 IU/kg VE partially alleviated respiratory alkalosis but did not reduce oxidative stress in heat-stressed pigs.

16.
Am J Kidney Dis ; 70(3): 440-444, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28599903

RESUMEN

Hypobicarbonatemia, or a reduced bicarbonate concentration in plasma, is a finding seen in 3 acid-base disorders: metabolic acidosis, chronic respiratory alkalosis and mixed metabolic acidosis and chronic respiratory alkalosis. Hypobicarbonatemia due to chronic respiratory alkalosis is often misdiagnosed as a metabolic acidosis and mistreated with the administration of alkali therapy. Proper diagnosis of the cause of hypobicarbonatemia requires integration of the laboratory values, arterial blood gas, and clinical history. The information derived from the urinary response to the prevailing acid-base disorder is useful to arrive at the correct diagnosis. We discuss the use of urine anion gap, as a surrogate marker of urine ammonium excretion, in the evaluation of a patient with low plasma bicarbonate concentration to differentiate between metabolic acidosis and chronic respiratory alkalosis. The interpretation and limitations of urine acid-base indexes at bedside (urine pH, urine bicarbonate, and urine anion gap) to evaluate urine acidification are discussed.


Asunto(s)
Acidosis/diagnóstico , Alcalosis Respiratoria , Hiperventilación , Accidente Cerebrovascular/complicaciones , Desequilibrio Hidroelectrolítico , Anciano de 80 o más Años , Alcalosis Respiratoria/sangre , Alcalosis Respiratoria/diagnóstico , Alcalosis Respiratoria/etiología , Diagnóstico Diferencial , Manejo de la Enfermedad , Femenino , Humanos , Concentración de Iones de Hidrógeno , Hiperventilación/sangre , Hiperventilación/etiología , Desequilibrio Hidroelectrolítico/sangre , Desequilibrio Hidroelectrolítico/diagnóstico , Desequilibrio Hidroelectrolítico/etiología
17.
Clin Exp Nephrol ; 21(5): 866-876, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28289910

RESUMEN

BACKGROUND: The Stewart model for analyzing acid-base disturbances emphasizes serum albumin levels, which are ignored in the traditional Boston model. We compared data derived using the Stewart model to those using the Boston model in patients with nephrotic syndrome. METHODS: Twenty-nine patients with nephrotic syndrome and six patients without urinary protein or acid-base disturbances provided blood and urine samples for analysis that included routine biochemical and arterial blood gas tests, plasma renin activity, and aldosterone. The total concentration of non-volatile weak acids (ATOT), apparent strong ion difference (SIDa), effective strong ion difference (SIDe), and strong ion gap (SIG) were calculated according to the formulas of Agrafiotis in the Stewart model. RESULTS: According to the Boston model, 25 of 29 patients (90%) had alkalemia. Eighteen patients had respiratory alkalosis, 11 had metabolic alkalosis, and 4 had both conditions. Only three patients had hyperreninemic hyperaldosteronism. The Stewart model demonstrated respiratory alkalosis based on decreased PaCO2, metabolic alkalosis based on decreased ATOT, and metabolic acidosis based on decreased SIDa. We could diagnose metabolic alkalosis or acidosis with a normal anion gap after comparing delta ATOT [(14.09 - measured ATOT) or (11.77 - 2.64 × Alb (g/dL))] and delta SIDa [(42.7 - measured SIDa) or (42.7 - (Na + K - Cl)]). We could also identify metabolic acidosis with an increased anion gap using SIG > 7.0 (SIG = 0.9463 × corrected anion gap-8.1956). CONCLUSIONS: Patients with nephrotic syndrome had primary respiratory alkalosis, decreased ATOT due to hypoalbuminemia (power to metabolic alkalosis), and decreased levels of SIDa (power to metabolic acidosis). We could detect metabolic acidosis with an increased anion gap by calculating SIG. The Stewart model in combination with the Boston model facilitates the analysis of complex acid-base disturbances in nephrotic syndrome.


Asunto(s)
Equilibrio Ácido-Base , Desequilibrio Ácido-Base/sangre , Bicarbonatos/sangre , Dióxido de Carbono/sangre , Modelos Biológicos , Síndrome Nefrótico/sangre , Desequilibrio Ácido-Base/diagnóstico , Desequilibrio Ácido-Base/fisiopatología , Desequilibrio Ácido-Base/orina , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/orina , Estudios de Casos y Controles , Femenino , Tasa de Filtración Glomerular , Humanos , Hipoalbuminemia/sangre , Hipoalbuminemia/fisiopatología , Hipoalbuminemia/orina , Riñón/fisiopatología , Masculino , Persona de Mediana Edad , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/fisiopatología , Síndrome Nefrótico/orina , Proteinuria/sangre , Proteinuria/fisiopatología , Proteinuria/orina , Sistema Renina-Angiotensina , Albúmina Sérica Humana/metabolismo
18.
Eur J Appl Physiol ; 117(11): 2251-2261, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28914359

RESUMEN

PURPOSE: Both exercise and hypoxia cause complex changes in acid-base homeostasis. The aim of the present study was to investigate whether during intense physical exercise in normoxia and hypoxia, the modified physicochemical approach offers a better understanding of the changes in acid-base homeostasis than the traditional Henderson-Hasselbalch approach. METHODS: In this prospective, randomized, crossover trial, 19 healthy males completed an exercise test until voluntary fatigue on a bicycle ergometer on two different study days, once during normoxia and once during normobaric hypoxia (12% oxygen, equivalent to an altitude of 4500 m). Arterial blood gases were sampled during and after the exercise test and analysed according to the modified physicochemical and Henderson-Hasselbalch approach, respectively. RESULTS: Peak power output decreased from 287 ± 9 Watts in normoxia to 213 ± 6 Watts in hypoxia (-26%, P < 0.001). Exercise decreased arterial pH to 7.21 ± 0.01 and 7.27 ± 0.02 (P < 0.001) during normoxia and hypoxia, respectively, and increased plasma lactate to 16.8 ± 0.8 and 17.5 ± 0.9 mmol/l (P < 0.001). While the Henderson-Hasselbalch approach identified lactate as main factor responsible for the non-respiratory acidosis, the modified physicochemical approach additionally identified strong ions (i.e. plasma electrolytes, organic acid ions) and non-volatile weak acids (i.e. albumin, phosphate ion species) as important contributors. CONCLUSIONS: The Henderson-Hasselbalch approach might serve as basis for screening acid-base disturbances, but the modified physicochemical approach offers more detailed insights into the complex changes in acid-base status during exercise in normoxia and hypoxia, respectively.


Asunto(s)
Equilibrio Ácido-Base , Ejercicio Físico , Hipoxia/sangre , Adulto , Humanos , Hipoxia/fisiopatología , Ácido Láctico/sangre , Masculino , Oxígeno/sangre
19.
Eur J Appl Physiol ; 117(8): 1573-1583, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28527012

RESUMEN

PURPOSE: To investigate the effect of voluntary hypocapnic hyperventilation or moderate hypoxia on metabolic and heart rate responses during high-intensity intermittent exercise. METHODS: Ten males performed three 30-s bouts of high-intensity cycling [Ex1 and Ex2: constant-workload at 80% of the power output in the Wingate anaerobic test (WAnT), Ex3: WAnT] interspaced with 4-min recovery periods under normoxic (Control), hypocapnic or hypoxic (2500 m) conditions. Hypocapnia was developed through voluntary hyperventilation for 20 min prior to Ex1 and during each recovery period. RESULTS: End-tidal CO2 pressure was lower before each exercise in the hypocapnia than control trials. Oxygen uptake ([Formula: see text]) was lower in the hypocapnia than control trials (822 ± 235 vs. 1645 ± 245 mL min-1; mean ± SD) during Ex1, but not Ex2 or Ex3, without a between-trial difference in the power output during the exercises. Heart rates (HRs) during Ex1 (127 ± 8 vs. 142 ± 10 beats min-1) and subsequent post-exercise recovery periods were lower in the hypocapnia than control trials, without differences during or after Ex2, except at 4 min into the second recovery period. [Formula: see text] did not differ between the control and hypoxia trials throughout. CONCLUSIONS: These results suggest that during three 30-s bouts of high-intensity intermittent cycling, (1) hypocapnia reduces the aerobic metabolic rate with a compensatory increase in the anaerobic metabolic rate during the first but not subsequent exercises; (2) HRs during the exercise and post-exercise recovery periods are lowered by hypocapnia, but this effect is diminished with repeated exercise bouts, and (3) moderate hypoxia (2500 m) does not affect the metabolic response during exercise.


Asunto(s)
Ciclismo/fisiología , Frecuencia Cardíaca/fisiología , Entrenamiento de Intervalos de Alta Intensidad , Hiperventilación/fisiopatología , Hipocapnia/fisiopatología , Hipoxia/fisiopatología , Humanos , Masculino , Músculo Esquelético/fisiología , Consumo de Oxígeno/fisiología , Adulto Joven
20.
Adv Exp Med Biol ; 903: 457-63, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27343114

RESUMEN

When Edmund Hillary and Tenzing Norgay reached the summit of Mt. Everest in 1953, it was the culmination of many attempts beginning in 1921. Alexander Kellas had actually predicted as early as 1920 that the mountain could be climbed, but the extreme altitude of 8848 m with the consequent oxygen deprivation had foiled previous attempts. One reason for the success of the 1953 expedition was the work done by the British physiologist Griffith Pugh in 1952 when he studied many of the physiological factors at high altitude including the oxygen requirements. Seven years later, Pugh and Hillary teamed up again for the Silver Hut Expedition in 1960-1961 that elucidated many of the problems of very high altitude. A group of physiologists spent several months at an altitude of 5800 m in a prefabricated hut and studied many aspects of exercise, pulmonary gas exchange, control of ventilation, and blood changes. Maximal exercise was measured as high as 7440 m and raised anew the question of whether Everest could ever be climbed without supplementary oxygen. The answer was shown to be yes in 1978 by Messner and Habeler, and 3 years later the American Medical Research Expedition to Everest clarified the physiological adaptations that allow humans to reach the highest point on earth. Five people reached the summit, the barometric pressure there was measured for the first time, and alveolar gas samples from the summit showed the critical importance of the extreme hyperventilation. However, the maximal oxygen consumption for the summit inspired PO2 of 43 mmHg was shown to be only about 1 l min(-1). In other words, the highest point on earth is very close to the limit of human tolerance to oxygen deprivation. As we celebrate the anniversary of Charles Darwin, it would be nice to have an evolutionary explanation for this, but in fact it is a cosmic coincidence.


Asunto(s)
Altitud , Fenómenos Fisiológicos , Presión Atmosférica , Expediciones , Humanos , Oxígeno/metabolismo , Consumo de Oxígeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA