Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Curr Dev Nutr ; 7(8): 101971, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37560461

RESUMEN

Background: There remains a need to identify low-cost interventions to improve coronavirus disease 2019 (COVID-19) outcomes. Vitamin D and zinc play a role in respiratory infections and could hold value as part of therapeutic regimens. Objectives: To determine the effect of vitamin D or zinc supplementation on recovery from COVID-19. Methods: We conducted a double-blind, randomly assigned 2 x 2 factorial placebo-controlled trial with 1:1:1:1 allocation ratio, enrolling nonpregnant adults with COVID-19 from hospitals in Mumbai and Pune, India (NCT04641195). Participants (N = 181) were randomly assigned to vitamin D3 (180,000 IU bolus, then 2000 IU daily), zinc (40 mg daily), vitamin D3 and zinc, or placebo, for 8 wk. Participants were followed until 8 wk. The primary outcome was time to resolution of fever, cough, and shortness of breath. Secondary outcomes were duration of individual symptoms; need for assisted ventilation; duration of hospital stay; all-cause mortality; and blood biomarkers, including nutritional, inflammatory, and immunological markers. Results: We observed no effect of vitamin D or zinc supplementation on time to resolution of all 3 symptoms [vitamin D hazard ratio (HR): 0.92; 95% confidence interval (95% CI): 0.66, 1.30; P = 0.650; zinc HR: 0.94; 95% CI: 0.67, 1.33; P = 0.745)]. Neither vitamin D nor zinc supplementation was associated with secondary outcomes, except for increased endline serum vitamin D with vitamin D supplementation [median (interquartile range) difference between endline and baseline for vitamin D: 5.3 ng/mL (-2.3 to 13.7); for no vitamin D: -1.4 ng/mL (-5.6 to 3.9); P = 0.003]. We observed nonsignificant increases in serum zinc at endline following zinc supplementation. There was no evidence of interaction between vitamin D and zinc supplementation, no effect of either on hypercalcemia, and no adverse events. Conclusions: Results suggest that neither vitamin D nor zinc supplementation improves COVID-19 treatment outcomes in this population. However, much larger-scale evidence, particularly from populations with vitamin D or zinc deficiency and severe infection, is required to corroborate our findings. This trial was registered at ClinicalTrials.gov and the Clinical Trials Registry of India as NCT04641195 and CTRI/2021/04/032593 respectively.

2.
BMJ Open ; 12(8): e061301, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-36038172

RESUMEN

INTRODUCTION: Presently, there are few population-level strategies to address SARS-CoV-2 infection except preventive measures such as vaccination. Micronutrient deficiency, particularly vitamin D and zinc deficiency, has been associated with dysregulated host responses, and may play an important role in COVID-19. METHODS AND ANALYSIS: We have designed a 2×2 factorial, randomised, double-blind, multi-centre placebo-controlled trial to evaluate the effect of vitamin D and zinc on COVID-19 outcomes in Maharashtra, India. COVID-19 positive individuals are recruited from hospitals in Mumbai and Pune. Participants are provided (1) vitamin D3 bolus (180 000 IU) maintained by daily dose of 2000 IU and/or (2) zinc gluconate (40 mg daily), versus placebo for 8 weeks. Participants undergo a detailed assessment at baseline and at 8 weeks, and are monitored daily in hospital or every 3 days after leaving the hospital to assess symptoms and other clinical measures. A final follow-up telephone call occurs 12 weeks post-enrolment to assess long-term outcomes. The primary outcome of the study is to time to recovery, defined as time to resolution of all of fever, cough and shortness of breath. Secondary outcomes include: duration of hospital stay, all-cause mortality, necessity of assisted ventilation, change in blood biomarker levels and individual symptoms duration. Participant recruitment commenced on April 2021. ETHICS AND DISSEMINATION: Ethical approval was obtained from institutional ethical committees of all participating institutions. The study findings will be presented in peer-reviewed medical journals. TRIAL REGISTRATION NUMBERS: NCT04641195, CTRI/2021/04/032593, HMSC (GOI)-2021-0060.


Asunto(s)
COVID-19 , Suplementos Dietéticos , Humanos , India/epidemiología , Estudios Multicéntricos como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Resultado del Tratamiento , Vitamina D/uso terapéutico , Zinc/uso terapéutico
3.
Adv Nutr ; 10(5): 765-777, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31075164

RESUMEN

The available data support the hypothesis that L-arginine or L-citrulline supplementation would be suitable for implementation in resource-constrained settings and will enhance placental vascular development and improve birth outcomes. In resource-constrained settings, the rates of adverse birth outcomes, including fetal growth restriction, preterm birth, and low birth weight, are disproportionately high. Complications resulting from preterm birth are now the leading cause of mortality in children <5 y of age worldwide. Despite the global health burden of adverse birth outcomes, few effective interventions are currently available and new strategies are urgently needed, especially for low-resource settings. L-arginine is a nutritionally essential amino acid in pregnancy and an immediate precursor of nitric oxide. During pregnancy, placental and embryonic growth increases the demand for L-arginine, which can exceed endogenous synthesis of L-arginine from L-citrulline, necessitating increased dietary intake. In many low-resource settings, dietary intake of L-arginine in pregnancy is inadequate owing to widespread protein malnutrition and depletion of endogenous L-arginine due to maternal infections, in particular malaria. Here we examine the role of the L-arginine-nitric oxide biosynthetic pathway in pregnancy including placental vascular development and fetal growth. We review the evidence for the relations between altered L-arginine bioavailability and pregnancy outcomes, and strategies for arginine supplementation in pregnancy. Existing studies of L-arginine supplementation in pregnancy in high-resource settings have shown improved maternal and fetal hemodynamics, prevention of pre-eclampsia, and improved birth outcomes including higher birth weight and longer gestation. Arginine supplementation studies now need to be extended to pregnant women in low-resource settings, especially those at risk of malaria.


Asunto(s)
Arginina/administración & dosificación , Citrulina/administración & dosificación , Suplementos Dietéticos , Fenómenos Fisiologicos Nutricionales Maternos , Atención Prenatal/métodos , Femenino , Desarrollo Fetal/efectos de los fármacos , Retardo del Crecimiento Fetal/prevención & control , Recursos en Salud/provisión & distribución , Humanos , Recién Nacido de Bajo Peso , Recién Nacido , Placenta/irrigación sanguínea , Preeclampsia/prevención & control , Embarazo , Resultado del Embarazo , Nacimiento Prematuro/prevención & control
4.
Parasite Immunol ; 41(4): e12614, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30703256

RESUMEN

AIMS: Malaria in pregnancy (MiP) alters the expression of ATP-binding cassette efflux transporters in maternal and foetal tissues, as well as the placenta. Malaria induces oxidative stress, and pregnancy is associated with arginine deficiency. We hypothesized that reducing oxidative stress during MiP by supplementation with L-arginine, a NO precursor, would attenuate transcriptional changes in a second superfamily of transporters, solute carrier (SLC) transporters, and improve pregnancy outcomes. METHODS AND RESULTS: Pregnant BALB/c mice receiving L-arginine (1.2%) in water, or water alone, were infected with Plasmodium berghei ANKA on gestational day 13 and sacrificed on gestational day 19. Compared to controls, the mRNA of numerous SLC transporters was downregulated in maternal and foetal tissues, as well as in the placentas of infected mice. While supplementation with L-arginine did improve foetal viability, it did not improve the mRNA expression of oxidative stress markers, transporters nor other indices of foetal and maternal health. Moreover, amino acid uptake transporters were downregulated upon infection, which could potentially contribute to decreased foetal birthweight. CONCLUSIONS: Malaria in pregnancy significantly alters the expression of SLC transporters in maternal and foetal tissues as well as the placenta, regardless of L-arginine supplementation. Further studies to investigate methods of reducing oxidative stress in MiP are warranted.


Asunto(s)
Malaria/patología , Estrés Oxidativo/fisiología , Placenta/metabolismo , Plasmodium berghei , Proteínas Transportadoras de Solutos/biosíntesis , Animales , Arginina/farmacología , Transporte Biológico , Femenino , Viabilidad Fetal/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Embarazo , Proteínas Transportadoras de Solutos/genética
5.
Sci Transl Med ; 10(431)2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29514999

RESUMEN

Reducing adverse birth outcomes due to malaria in pregnancy (MIP) is a global health priority. However, there are few safe and effective interventions. l-Arginine is an essential amino acid in pregnancy and an immediate precursor in the biosynthesis of nitric oxide (NO), but there are limited data on the impact of MIP on NO biogenesis. We hypothesized that hypoarginemia contributes to the pathophysiology of MIP and that l-arginine supplementation would improve birth outcomes. In a prospective study of pregnant Malawian women, we show that MIP was associated with lower concentrations of l-arginine and higher concentrations of endogenous inhibitors of NO biosynthesis, asymmetric and symmetric dimethylarginine, which were associated with adverse birth outcomes. In a model of experimental MIP, l-arginine supplementation in dams improved birth outcomes (decreased stillbirth and increased birth weight) compared with controls. The mechanism of action was via normalized angiogenic pathways and enhanced placental vascular development, as visualized by placental microcomputerized tomography imaging. These data define a role for dysregulation of NO biosynthetic pathways in the pathogenesis of MIP and support the evaluation of interventions to enhance l-arginine bioavailability as strategies to improve birth outcomes.


Asunto(s)
Malaria/tratamiento farmacológico , Placenta/efectos de los fármacos , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Arginina/uso terapéutico , Femenino , Humanos , Malaria/sangre , Malaria/metabolismo , Ratones , Óxido Nítrico/metabolismo , Placenta/metabolismo , Embarazo , Resultado del Embarazo , Estudios Prospectivos
6.
Proc Natl Acad Sci U S A ; 115(10): E2366-E2375, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463745

RESUMEN

Cerebral malaria (CM) is a severe and rapidly progressing complication of infection by Plasmodium parasites that is associated with high rates of mortality and morbidity. Treatment options are currently few, and intervention with artemisinin (Art) has limited efficacy, a problem that is compounded by the emergence of resistance to Art in Plasmodium parasites. Rocaglates are a class of natural products derived from plants of the Aglaia genus that have been shown to interfere with eukaryotic initiation factor 4A (eIF4A), ultimately blocking initiation of protein synthesis. Here, we show that the rocaglate CR-1-31B perturbs association of Plasmodium falciparum eIF4A (PfeIF4A) with RNA. CR-1-31B shows potent prophylactic and therapeutic antiplasmodial activity in vivo in mouse models of infection with Plasmodium berghei (CM) and Plasmodium chabaudi (blood-stage malaria), and can also block replication of different clinical isolates of P. falciparum in human erythrocytes infected ex vivo, including drug-resistant P. falciparum isolates. In vivo, a single dosing of CR-1-31B in P. berghei-infected animals is sufficient to provide protection against lethality. CR-1-31B is shown to dampen expression of the early proinflammatory response in myeloid cells in vitro and dampens the inflammatory response in vivo in P. berghei-infected mice. The dual activity of CR-1-31B as an antiplasmodial and as an inhibitor of the inflammatory response in myeloid cells should prove extremely valuable for therapeutic intervention in human cases of CM.


Asunto(s)
Aglaia/química , Antimaláricos/administración & dosificación , Malaria Cerebral/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Animales , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Factor 4F Eucariótico de Iniciación/genética , Factor 4F Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
7.
Am J Epidemiol ; 186(5): 603-611, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911008

RESUMEN

The amino acid arginine is a physiological precursor to nitric oxide, which is a key mediator of embryonic survival, fetal growth, and pregnancy maintenance. We evaluated the association between consumption of the amino acid arginine and the rate of adverse birth outcomes using data from a double-blind, randomized, placebo-controlled micronutrient supplementation trial among pregnant women in Dar es Salaam, Tanzania (2001-2004). Dietary intakes of arginine were assessed using repeated 24-hour recalls that were administered throughout pregnancy. Participants (n = 7,591) were monitored by research midwives throughout follow-up to assess pregnancy outcomes. Cubic-restricted splines and multivariable log-Poisson regression with empirical standard errors were used to estimate the continuous and categorical associations between arginine intake and adverse birth outcomes. Compared with women within the lowest quintile of arginine intake, those within the highest quintile had 0.79 times the risk of preterm birth before 37 weeks (95% confidence interval: 0.63, 1.00; P = 0.03). The continuous associations of arginine intake with preterm birth before 37 weeks and with preterm birth before 34 weeks were characterized by an initial rapid decrease in risk with increasing intake (P for nonlinearity < 0.01). Arginine intake was not associated with fetal loss or giving birth to infants who were born small for their gestational ages. This data suggest that the association between dietary arginine intake and preterm birth warrants further investigation.


Asunto(s)
Arginina/fisiología , Dieta , Resultado del Embarazo/epidemiología , Nacimiento Prematuro/epidemiología , Arginina/administración & dosificación , Suplementos Dietéticos/estadística & datos numéricos , Femenino , Humanos , Recién Nacido , Micronutrientes/administración & dosificación , Micronutrientes/fisiología , Distribución de Poisson , Embarazo , Nacimiento Prematuro/prevención & control , Estudios Prospectivos , Ensayos Clínicos Controlados Aleatorios como Asunto , Tanzanía/epidemiología
8.
Am J Trop Med Hyg ; 96(4): 826-834, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28115667

RESUMEN

AbstractVitamin A and zinc are important for immune function and may improve host defense against malaria and reduce the risk of adverse pregnancy outcomes. Our objective was to determine whether daily oral supplementation with either or both nutrients starting in the first trimester reduces the risk of placental malaria and adverse pregnancy outcomes. We undertook a randomized, double-blind placebo-controlled trial with a factorial design among 2,500 human immunodeficiency virus-negative primigravid or secundigravid pregnant women in their first trimester of pregnancy in Dar es Salaam, Tanzania. We randomly allocated equal numbers of participants to 2,500 IU of vitamin A, 25 mg of zinc, both 2,500 IU of vitamin A and 25 mg of zinc, or a placebo until delivery. A total of 625 participants were allocated to each treatment group. Our primary outcome, placental malaria infection (past or current), was assessed in all randomized participants for whom placental samples were obtained at delivery (N = 1,404), which represents 56% of total participants and 62% of all pregnancies lasting 28 weeks or longer (N = 2,266). Birth outcomes were obtained for 2,434 of the 2,500 randomized participants. Secondary outcomes included small for gestational age (SGA) births and prematurity. All analyses were intent to treat. Those who received zinc had a lower risk of histopathology-positive placental malaria compared with those who did not receive zinc (risk ratio = 0.64, 95% confidence interval = 0.44, 0.91), but neither nutrient had an effect on polymerase chain reaction-positive malaria, SGA, or prematurity. No safety concerns were identified. We recommend additional studies in other geographic locations to confirm these findings.


Asunto(s)
Malaria Falciparum/prevención & control , Placenta/parasitología , Complicaciones Parasitarias del Embarazo/prevención & control , Vitamina A/administración & dosificación , Zinc/administración & dosificación , Suplementos Dietéticos , Método Doble Ciego , Femenino , Humanos , Malaria Falciparum/diagnóstico , Malaria Falciparum/epidemiología , Placenta/patología , Reacción en Cadena de la Polimerasa , Embarazo , Resultado del Embarazo , Sensibilidad y Especificidad , Tanzanía/epidemiología
9.
Int J Med Sci ; 9(2): 157-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253563

RESUMEN

In vitro, nitric oxide (NO) has been shown to have antimicrobial activity against a wide range of viruses, including influenza A virus. Therefore, we hypothesized that inhaled nitric oxide (iNO) would increase survival in vivo by reducing the viral load in C57Bl/6 mice infected with a lethal dose of influenza A/WSN/33 (H1N1; WSN/33) virus. NO was delivered to influenza-infected mice either continuously or intermittently at 80 or 160 ppm, respectively, using both prophylactic and post-infection treatment strategies. Murine survival and weight loss were assessed, and lung viral load was quantified via plaque assay. Here, we report that iNO administered prophylactically or post-influenza infection failed to improve survival of infected mice. No difference in lung viral load was observed between experimental groups. Although NO has antiviral activity against influenza A virus in vitro, iNO therapy provided no apparent benefit when used for treatment of influenza A virus infection in vivo.


Asunto(s)
Óxido Nítrico/administración & dosificación , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Administración por Inhalación , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Evaluación Preclínica de Medicamentos , Femenino , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/fisiología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/efectos adversos , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Índice de Severidad de la Enfermedad , Insuficiencia del Tratamiento , Carga Viral/efectos de los fármacos
10.
Med Hypotheses ; 77(3): 437-44, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21745716

RESUMEN

We hypothesize that supplemental inhaled nitric oxide (iNO) will improve outcomes in children with severe malaria receiving standard antimalarial therapy. The rationale for the hypothesized efficacy of iNO rests on: (1) biological plausibility, based on known actions of NO in modulating endothelial activation; (2) pre-clinical efficacy data from animal models of experimental cerebral malaria; and (3) a human trial of the NO precursor l-arginine, which improved endothelial function in adults with severe malaria. iNO is an attractive new candidate for the adjunctive treatment of severe malaria, given its proven therapeutic efficacy in animal studies, track record of safety in clinical practice and numerous clinical trials, inexpensive manufacturing costs, and ease of administration in settings with limited healthcare infrastructure. We plan to test this hypothesis in a randomized controlled trial (ClinicalTrials.gov Identifier: NCT01255215).


Asunto(s)
Células Endoteliales/efectos de los fármacos , Malaria/tratamiento farmacológico , Óxido Nítrico Sintasa/genética , Óxido Nítrico/farmacología , Administración por Inhalación , Disponibilidad Biológica , Niño , Ensayos Clínicos como Asunto , Humanos , Pulmón/irrigación sanguínea , Óxido Nítrico/administración & dosificación , Óxido Nítrico/uso terapéutico
11.
Am J Trop Med Hyg ; 68(1): 107-10, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12557835

RESUMEN

We assessed whether mutations in the Plasmodium falciparum multidrug-resistance gene 1 (pfmdr1) (C1034S, D1042N, and Y1246D) would predict treatment outcome during a 28-day in vivo treatment trial in the Peruvian Amazon. Mefloquine (MQ) was compared with mefloquine-artesunate (MQ-AS) in a randomized, multi-clinic protocol for the first time in the Americas. Of 115 patients enrolled in the in vivo arm, 97 patients were eligible for molecular analysis. All 97 patients remained parasite-free during 28 days of follow-up (MQ, n = 46; MQ-AS, n = 51), indicating 100% clinical efficacy of the MQ and MQ-AS treatment regimens. The reported MQ-sensitive alleles (C1034, D1042, and Y1246) were present in 48.5% (n = 47) of the cases, whereas 49 isolates (50.5%) contained the D1246 mutation reported to confer MQ resistance in vitro. However, neither this mutation nor a double mutation (S1034, D1246; n = 16) was predictive of MQ treatment outcome.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Antimaláricos/uso terapéutico , Artemisininas/uso terapéutico , Malaria Falciparum/tratamiento farmacológico , Mefloquina/uso terapéutico , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Sesquiterpenos/uso terapéutico , Adulto , Animales , Antimaláricos/farmacología , Artemisininas/farmacología , Artesunato , Secuencia de Bases , Estudios de Cohortes , ADN Protozoario/química , Resistencia a Medicamentos/genética , Femenino , Humanos , Malaria Falciparum/parasitología , Masculino , Mefloquina/farmacología , Datos de Secuencia Molecular , Mutación , Perú , Plasmodium falciparum/efectos de los fármacos , Valor Predictivo de las Pruebas , Alineación de Secuencia , Sesquiterpenos/farmacología
12.
Lancet ; 359(9315): 1404-6, 2002 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11978340

RESUMEN

Supplementation with vitamin A potentiates host resistance to malaria, however, the underlying mechanism is unknown. We tested the effects of 9-cis-retinoic acid, a metabolite of vitamin A, on CD36 expression, non-opsonic phagocytic clearance of parasitised erythrocytes, and TNFalpha production in human monocytes and macrophages. We found reduced secretion of TNFalpha, upregulated CD36 expression, and increased phagocytosis of Plasmodium falciparum-parasitised erythrocytes. Increased parasite clearance and reduced proinflammatory cytokine responses to infection might partly explain the beneficial effects of supplementation with vitamin A in malaria.


Asunto(s)
Antiprotozoarios/farmacología , Malaria Falciparum/sangre , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Tretinoina/farmacología , Vitamina A/farmacología , Alitretinoína , Animales , Antiprotozoarios/administración & dosificación , Antígenos CD36/efectos de los fármacos , Niño , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Fagocitosis/efectos de los fármacos , Tretinoina/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Vitamina A/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA