Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 12: 669980, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149618

RESUMEN

Anorexia nervosa (AN) is an eating disorder leading to malnutrition and, ultimately, to energy wasting and cachexia. Rodents develop activity-based anorexia (ABA) when simultaneously exposed to a restricted feeding schedule and allowed free access to running wheels. These conditions lead to a life-threatening reduction in body weight, resembling AN in human patients. Here, we investigate the effect of ABA on whole body energy homeostasis at different housing temperatures. Our data show that ABA rats develop hyperactivity and hypophagia, which account for a massive body weight loss and muscle cachexia, as well as reduced uncoupling protein 1 (UCP1) expression in brown adipose tissue (BAT), but increased browning of white adipose tissue (WAT). Increased housing temperature reverses not only the hyperactivity and weight loss of animals exposed to the ABA model, but also hypothermia and loss of body and muscle mass. Notably, despite the major metabolic impact of ABA, none of the changes observed are associated to changes in key hypothalamic pathways modulating energy metabolism, such as AMP-activated protein kinase (AMPK) or endoplasmic reticulum (ER) stress. Overall, this evidence indicates that although temperature control may account for an improvement of AN, key hypothalamic pathways regulating thermogenesis, such as AMPK and ER stress, are unlikely involved in later stages of the pathophysiology of this devastating disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/patología , Anorexia/fisiopatología , Hipotálamo/patología , Termogénesis , Proteína Desacopladora 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Peso Corporal , Metabolismo Energético , Homeostasis , Hipotálamo/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Desacopladora 1/genética
2.
Nutrients ; 13(3)2021 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33802580

RESUMEN

BACKGROUND: Nutrition plays an important role in maintaining the overall health of older people. Inadequate intake may lead to impaired body function, higher morbidity, and mortality. Oral nutritional supplements (ONS) showed positive effect on the nutritional status of the elderly; however, systematic evidence is currently lacking on the effect of ONS on the elderly with anorexia. AIMS: The current systematic review and meta-analysis included randomized controlled trial (RCT) articles to investigate the effectiveness of ONS on the main aspects of anorexia of aging (AA). METHODS: By using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method, researchers independently searched PubMed/MEDLINE, EMBASE, CINAHL, PsycINFO, the Cochrane Library, China National Knowledge Infrastructure (CNKI) and other gray literature resources for publications that met the inclusion criteria by October 2020. The Cochrane Risk of Bias Tools were used for quality assessment. The inverse-variance method was used for the fixed model (FM) while the DerSimonian-Laird method was used for the random model (RM). Respective 95% confidence intervals (95% CIs), mean difference (MD) or standardized mean difference (SMD) was used for indices in terms of effect size (ES). RESULTS: 2497 records were found through the systematic search, while 17 RCTs (n = 1204) were included, with a mean age of 81.9 years (range: 74-87 years). Supplementation occurred in the morning, mid-day, and evening, while the times varied from one to three times a day. The results of meta-analysis showed that, generally, ONS had a positive effect on the overall appetite, MD = 0.18, 95% CI (0.03, 0.33), p = 0.02, and consumption, MD = 1.43, 95% CI (0.01, 2.86), p = 0.05; but not significant in terms of other aspects of appetite: hunger, p = 0.73; fullness, p = 0.60; desire to eat, p = 0.80; preoccupation, p = 0.15. Additionally, it showed an increase in the overall energy intake, SMD = 0.46, 95% CI (0.29, 0.63), p < 0.001, in protein intake, SMD = 0.59, 95% CI (0.16, 1.02), p = 0.007, and in fat intake, MD = 3.47, 95% CI (1.98, 4.97), p < 0.001, while no positive effect was found on carbohydrates intake, p = 0.06. Significance differences were also found in the body weight, SMD = 0.53, 95% CI (0.41, 0.65), p < 0.001, and body mass index (BMI), MD = 0.53, 95% CI (0.12, 0.95), p = 0.01. Moreover, subgroup analyses were conducted according to the nutrient density with no positive results showed except for the low-density ONS on overall energy intake. CONCLUSIONS: The results of the present study indicated that ONS had beneficial effects on overall appetite, energy intake, body weight and BMI.


Asunto(s)
Anorexia/terapia , Suplementos Dietéticos , Anciano , Anciano de 80 o más Años , Anorexia/fisiopatología , Apetito , Índice de Masa Corporal , Peso Corporal , Ingestión de Energía , Femenino , Humanos , Masculino , Estado Nutricional , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
3.
ACS Chem Neurosci ; 11(11): 1520-1522, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32427468

RESUMEN

Accumulating data have now shown strong evidence that COVID-19 infection leads to the occurrence of neurological signs with different injury severity. Anosmia and agueusia are now well documented and included in the criteria list for diagnosis, and specialists have stressed that doctors screen COVID-19 patients for these two signs. The eventual brainstem dysregulation, due to the invasion of SARS CoV-2, as a cause of respiratory problems linked to COVID-19, has also been extensively discussed. All these findings lead to an implication of the central nervous system in the pathophysiology of COVID-19. Here we provide additional elements that could explain other described signs like appetite loss, vomiting, and nausea. For this, we investigated the role of brainstem structures located in the medulla oblongata involved in food intake and vomiting control. We also discussed the possible pathways the virus uses to reach the brainstem, i.e., neurotropic and hematogenous (with its two variants) routes.


Asunto(s)
Anorexia/fisiopatología , Regulación del Apetito/fisiología , Sistema Nervioso Autónomo/fisiopatología , Infecciones por Coronavirus/fisiopatología , Ingestión de Alimentos/fisiología , Náusea/fisiopatología , Neumonía Viral/fisiopatología , Núcleo Solitario/fisiopatología , Vómitos/fisiopatología , Ageusia/etiología , Anorexia/etiología , Área Postrema/fisiopatología , Barrera Hematoencefálica , COVID-19 , Infecciones por Coronavirus/complicaciones , Humanos , Hipotálamo/fisiopatología , Bulbo Raquídeo/fisiopatología , Náusea/etiología , Vías Nerviosas/fisiopatología , Trastornos del Olfato/etiología , Nervio Olfatorio , Pandemias , Neumonía Viral/complicaciones , Nervio Vago , Vómitos/etiología
4.
Nutrients ; 11(6)2019 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-31208031

RESUMEN

BACKGROUND: During activity-based anorexia (ABA) in mice, enhanced paracellular permeability and reduced protein synthesis have been shown in the colon while the gut-brain axis has received increasing attention in the regulation of intestinal and mood disorders that frequently occur during anorexia nervosa, a severe eating disorder for which there is no specific treatment. In the present study, we assessed the effects of oral glutamine (Gln) or branched-chain amino acids (BCAA) supplementation during ABA to target intestinal functions, body composition and feeding behavior. METHODS: C57BL/6 male mice were randomized in Control (CTRL) and ABA groups. After ABA induction, mice received, or not, either 1% Gln or 2.5% BCAA (Leu, Ile, Val) for one week in drinking water. RESULTS: Neither Gln nor BCAA supplementation affected body weight and body composition, while only Gln supplementation slightly increased food intake. ABA mice exhibited increased paracellular permeability and reduced protein synthesis in the colonic mucosa. Oral Gln restored colonic paracellular permeability and protein synthesis and increased the mucin-2 mRNA level, whereas BCAA did not affect colonic parameters. CONCLUSION: In conclusion, oral Gln specifically improves colonic response during ABA. These data should be further confirmed in AN patients.


Asunto(s)
Aminoácidos de Cadena Ramificada/farmacología , Anorexia/tratamiento farmacológico , Suplementos Dietéticos , Glutamina/farmacología , Mucosa Intestinal/efectos de los fármacos , Animales , Anorexia/fisiopatología , Composición Corporal/efectos de los fármacos , Colon/efectos de los fármacos , Colon/fisiopatología , Conducta Alimentaria/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos
5.
Addict Biol ; 24(4): 707-716, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-29659126

RESUMEN

The α2a-adrenergic agonist, lofexidine, reduced cannabis withdrawal-related sleep disruption in the laboratory, but side effects (e.g. fatigue, hypotension) limit its utility as a treatment for cannabis use disorder. This study tested the potential efficacy and tolerability of a daily bedtime administration of the FDA-approved α2a-adrenergic agonist, guanfacine, in a human laboratory model of cannabis use disorder. Daily, nontreatment-seeking cannabis smokers (13M, 2F) completed a within-subject study comprising two 9-day inpatient study phases. Each phase tested the effects of daily placebo or immediate-release guanfacine (2 mg) on cannabis intoxication (5.6 percent THC; 2 days), withdrawal (4 days of abstinence) and subsequent 'relapse' (3 days of cannabis self-administration). Ratings of mood, sleep, cardiovascular effects, food intake, psychomotor performance and cannabis self-administration were assessed. An outpatient phase preceded each inpatient phase for medication clearance or dose induction. Under placebo medication conditions, cannabis abstinence produced significant withdrawal, including irritability, sleep disruption and anorexia. Guanfacine reduced ratings of irritability and improved objective measures of sleep during cannabis withdrawal relative to placebo but did not reduce cannabis self-administration. Guanfacine was well tolerated with little evidence of fatigue and only small decreases in blood pressure: no dose was held due to hypotension. Thus, a single daily administration of guanfacine at bedtime improved sleep and mood during cannabis withdrawal relative to placebo. This positive signal supports further studies varying the guanfacine dose, formulation or frequency of administration, or combining it with other medications to increase the likelihood of having an impact on cannabis use.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Guanfacina/uso terapéutico , Abuso de Marihuana , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Adulto , Afecto , Anorexia/etiología , Anorexia/fisiopatología , Presión Sanguínea , Cannabis/efectos adversos , Conducta Alimentaria , Femenino , Humanos , Genio Irritable , Masculino , Desempeño Psicomotor , Autoadministración , Sueño , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Síndrome de Abstinencia a Sustancias/psicología , Adulto Joven
6.
J Hum Nutr Diet ; 32(1): 128-138, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30159922

RESUMEN

BACKGROUND: Anorexia of ageing (AA) may be considered as a risk factor for frailty and has an important impact on quality of life, morbidity and mortality. METHODS: A systematic review and a meta-analysis were performed to summarise the results from several trials on the effectiveness of treatments in AA, as associated with depression, sensory impairment of taste and smell, decreased appetite or early satiety, and disability. Eligible studies were required to report baseline and follow-up values, the mean change (∆-change) from baseline, and/or the mean difference among intervention groups versus control group, concerning food intake (kcal/daily) and/or nutritional outcomes, such as body weight, body mass index, albumin and Mini Nutritional Assessment. RESULTS: The systematic review included 20 papers based on different therapeutic approaches concerning food intake and/or nutritional outcomes. The results of the meta-analysis indicate that the interventions for AA have an important impact on body weight [+1.59 kg; 95% confidence interval (CI) = 1.48-+1.71 kg; P < 0.001) and on energy intake (+56.09 kcal; 95% CI = -54.05 to +166.25 kcal; P = 0.32). Regarding secondary outcomes, it was not possible to meta-analyse the limited amount of data availab le. CONCLUSIONS: The different variants of AA need to be defined because diverse therapeutic approaches are available. A more precise definition of the functional impairments associated with AA may allow a more correct decision about the most appropriate therapy to be prescribed. Moreover, this may allow for a more effective performance of the different therapeutic approaches once they are better targeted to the different scenarios of AA.


Asunto(s)
Envejecimiento/patología , Anorexia/terapia , Terapia Nutricional/métodos , Estado Nutricional , Anciano , Anciano de 80 o más Años , Anorexia/fisiopatología , Índice de Masa Corporal , Peso Corporal , Ingestión de Energía , Femenino , Evaluación Geriátrica , Humanos , Masculino , Persona de Mediana Edad , Evaluación Nutricional , Calidad de Vida , Resultado del Tratamiento
7.
Trials ; 18(1): 485, 2017 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-29047408

RESUMEN

BACKGROUND: Cisplatin is a key drug in lung cancer therapy. However, cisplatin is also well known to induce gastrointestinal disorders, such as chemotherapy-induced nausea and vomiting, anorexia, and weight loss. These symptoms sometimes affect patients' quality of life and make continuation of chemotherapy difficult. Anorexia is a cause of concern for patients with cancer because a persistent loss of appetite progresses to cancer cachexia. Although evidence-based management for chemotherapy has recently been established, there is room for improvement. METHODS/DESIGN: This placebo-controlled, double-blind, randomized trial will aim to determine the efficacy of the traditional Japanese Kampo medicine rikkunshito (TJ-43) for preventing anorexia caused by cisplatin-including chemotherapy in patients with lung cancer. Patients with lung cancer who plan to receive cisplatin-including chemotherapy will be recruited. Patients who provide written consent will be randomly allocated to receive either TJ-43 (arm A) or placebo (arm B) for one course of chemotherapy (21 or 28 consecutive days). Investigators and patients will be masked to the treatment assignment throughout the trial. The primary endpoint will be evaluated as the change in dietary intake from day 0 (the day before the start of chemotherapy) to day 7 of cisplatin-including chemotherapy. The two arms of the trial will comprise 30 patients each. From November 2014, a total of 60 patients will be recruited, and recruitment for the study is planned to be complete by October 2017. DISCUSSION: This trial is designed to examine the efficacy of rikkunshito (TJ-43) for reducing anorexia and maintaining food intake caused by cisplatin-including chemotherapy in patients with lung cancer. TRIAL REGISTRATION: Japan Pharmaceutical Information Center Clinical Trials Information (JAPIC CTI), trial registration: JAPIC CTI-142747 . Registered on 15 December 2014; the RICH trial.


Asunto(s)
Anorexia/prevención & control , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Cisplatino/efectos adversos , Medicamentos Herbarios Chinos/uso terapéutico , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Medicina Kampo/métodos , Anorexia/inducido químicamente , Anorexia/fisiopatología , Anorexia/psicología , Protocolos Clínicos , Método Doble Ciego , Medicamentos Herbarios Chinos/efectos adversos , Humanos , Japón , Proyectos de Investigación , Factores de Tiempo , Resultado del Tratamiento
8.
J Neurosci ; 37(36): 8678-8687, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28821663

RESUMEN

To maintain energy homeostasis, orexigenic (appetite-inducing) and anorexigenic (appetite suppressing) brain systems functionally interact to regulate food intake. Within the hypothalamus, neurons that express agouti-related protein (AgRP) sense orexigenic factors and orchestrate an increase in food-seeking behavior. In contrast, calcitonin gene-related peptide (CGRP)-expressing neurons in the parabrachial nucleus (PBN) suppress feeding. PBN CGRP neurons become active in response to anorexigenic hormones released following a meal, including amylin, secreted by the pancreas, and cholecystokinin (CCK), secreted by the small intestine. Additionally, exogenous compounds, such as lithium chloride (LiCl), a salt that creates gastric discomfort, and lipopolysaccharide (LPS), a bacterial cell wall component that induces inflammation, exert appetite-suppressing effects and activate PBN CGRP neurons. The effects of increasing the homeostatic drive to eat on feeding behavior during appetite suppressing conditions are unknown. Here, we show in mice that food deprivation or optogenetic activation of AgRP neurons induces feeding to overcome the appetite suppressing effects of amylin, CCK, and LiCl, but not LPS. AgRP neuron photostimulation can also increase feeding during chemogenetic-mediated stimulation of PBN CGRP neurons. AgRP neuron stimulation reduces Fos expression in PBN CGRP neurons across all conditions. Finally, stimulation of projections from AgRP neurons to the PBN increases feeding following administration of amylin, CCK, and LiCl, but not LPS. These results demonstrate that AgRP neurons are sufficient to increase feeding during noninflammatory-based appetite suppression and to decrease activity in anorexigenic PBN CGRP neurons, thereby increasing food intake during homeostatic need.SIGNIFICANCE STATEMENT The motivation to eat depends on the relative balance of activity in distinct brain regions that induce or suppress appetite. An abnormal amount of activity in neurons that induce appetite can cause obesity, whereas an abnormal amount of activity in neurons that suppress appetite can cause malnutrition and a severe reduction in body weight. The purpose of this study was to determine whether a population of neurons known to induce appetite ("AgRP neurons") could induce food intake to overcome appetite-suppression following administration of various appetite-suppressing compounds. We found that stimulating AgRP neurons could overcome various forms of appetite suppression and decrease neural activity in a separate population of appetite-suppressing neurons, providing new insights into how the brain regulates food intake.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Anorexia/fisiopatología , Regulación del Apetito , Ingestión de Alimentos , Inhibición Neural , Neuronas/metabolismo , Núcleos Parabraquiales/fisiopatología , Proteína Relacionada con Agouti/genética , Animales , Anorexia/patología , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Núcleos Parabraquiales/patología
9.
Curr Opin Clin Nutr Metab Care ; 20(5): 396-401, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28708669

RESUMEN

PURPOSE OF REVIEW: In cancer patients, the development of cachexia (muscle wasting) is frequently aggravated by anorexia (loss of appetite). Their concurrence is often referred to as anorexia-cachexia syndrome. This review focusses on the recent evidence underlining hypothalamic inflammation as key driver of these processes. Special attention is given to the involvement of hypothalamic serotonin. RECENT FINDINGS: The anorexia-cachexia syndrome is directly associated with higher mortality in cancer patients. Recent reports confirm its severe impact on the quality of life of patients and their families.Hypothalamic inflammation has been shown to contribute to muscle and adipose tissue loss in cancer via central hypothalamic interleukine (IL)1ß-induced activation of the hypothalamic-pituitary-adrenal axis. The resulting release of glucocorticoids directly stimulates catabolic processes in these tissues via activation of the ubiquitin-proteosome pathway. Next to this, hypothalamic inflammation has been shown to reduce food intake in cancer by triggering changes in orexigenic and anorexigenic responses via upregulation of serotonin availability and stimulation of its signalling pathways in hypothalamic tissues. This combination of reduced food intake and stimulation of tissue catabolism represents a dual mechanism by which hypothalamic inflammation contributes to the development and maintenance of anorexia and cachexia in cancer. SUMMARY: Hypothalamic inflammation is a driving force in the development of the anorexia-cachexia syndrome via hypothalamic-pituitary-adrenal axis and serotonin pathway activation.


Asunto(s)
Anorexia/etiología , Caquexia/etiología , Enfermedades Hipotalámicas/etiología , Hipotálamo/inmunología , Modelos Neurológicos , Neoplasias/fisiopatología , Serotonina/metabolismo , Adiposidad , Animales , Anorexia/inmunología , Anorexia/metabolismo , Anorexia/fisiopatología , Caquexia/inmunología , Caquexia/metabolismo , Caquexia/fisiopatología , Humanos , Enfermedades Hipotalámicas/inmunología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/fisiopatología , Sistema Hipotálamo-Hipofisario/inmunología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/metabolismo , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Neoplasias/sangre , Neoplasias/inmunología , Neoplasias/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Sistema Hipófiso-Suprarrenal/inmunología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Serotonina/sangre
10.
J Neurosci ; 37(2): 362-370, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28077715

RESUMEN

Cisplatin chemotherapy is commonly used to treat cancer despite severe energy balance side effects. In rats, cisplatin activates nucleus tractus solitarius (NTS) projections to the lateral parabrachial nucleus (lPBN) and calcitonin-gene related peptide (CGRP) projections from the lPBN to the central nucleus of the amygdala (CeA). We demonstrated previously that CeA glutamate receptor signaling mediates cisplatin-induced anorexia and body weight loss. Here, we used neuroanatomical tracing, immunofluorescence, and confocal imaging to demonstrate that virtually all NTS→lPBN and lPBN→CeA CGRP projections coexpress vesicular glutamate transporter 2 (VGLUT2), providing evidence that excitatory projections mediate cisplatin-induced energy balance dysregulation. To test whether lPBN→CeA projection neurons are required for cisplatin-induced anorexia and weight loss, we inhibited these neurons chemogenetically using a retrograde Cre-recombinase-expressing canine adenovirus-2 in combination with Cre-dependent inhibitory Designer Receptors Exclusive Activated by Designer Drugs (DREADDs) before cisplatin treatment. Inhibition of lPBN→CeA neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Using a similar approach, we additionally demonstrated that inhibition of NTS→lPBN neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Together, our data support the view that excitatory hindbrain-forebrain projections are necessary for cisplatin's untoward effects on energy intake, elucidating a key neuroanatomical circuit driving pathological anorexia and weight loss that accompanies chemotherapy treatment. SIGNIFICANCE STATEMENT: Chemotherapy treatments are commonly used to treat cancers despite accompanying anorexia and weight loss that may limit treatment adherence and reduce patient quality of life. Strikingly, we lack a neural understanding of, and effective treatments for, chemotherapy-induced anorexia and weight loss. The current data characterize the excitatory nature of neural projections activated by cisplatin in rats and reveal the necessity of specific hindbrain-forebrain projections for cisplatin-induced anorexia and weight loss. Together, these findings help to characterize the neural mechanisms mediating cisplatin-induced anorexia, advancing opportunities to develop better-tolerated chemotherapies and adjuvant therapies to prevent anorexia and concurrent nutritional deficiencies during cancer treatment.


Asunto(s)
Amígdala del Cerebelo/fisiología , Anorexia/inducido químicamente , Cisplatino/toxicidad , Núcleos Parabraquiales/fisiología , Núcleo Solitario/fisiología , Pérdida de Peso/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anorexia/fisiopatología , Antineoplásicos/toxicidad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Núcleos Parabraquiales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
11.
Drugs Today (Barc) ; 52(9): 519-529, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27883118

RESUMEN

Cachexia is a multiorgan, multifactorial and often irreversible wasting syndrome associated with cancer and other serious, chronic illnesses including AIDS, chronic heart failure, chronic kidney disease and chronic obstructive pulmonary disease. Treatment of the patient with cachexia is currently targeted to correcting the two underlying features of the condition: anorexia and metabolic disturbances. Greater understanding of the mechanisms behind cachexia and muscle wasting have led to new therapeutic possibilities, however. Several classes of drugs are under active development for cachexia including drugs acting on hormone receptors or cytokine receptors, myostatin/activin pathway antagonists, beta-adrenoceptor agonists and cannabinoids. This review will cover the pathophysiology, epidemiology, diagnosis, treatment, drug candidates under active development and targets for therapeutic intervention of cachexia.


Asunto(s)
Anorexia/tratamiento farmacológico , Caquexia , Cannabinoides/farmacología , Suplementos Dietéticos , Glucocorticoides/farmacología , Hormonas/farmacología , Entrenamiento de Fuerza/métodos , Congéneres de la Testosterona/farmacología , Anorexia/fisiopatología , Caquexia/diagnóstico , Caquexia/etiología , Caquexia/metabolismo , Caquexia/fisiopatología , Caquexia/terapia , Terapia Combinada , Diagnóstico Diferencial , Humanos , Transducción de Señal/efectos de los fármacos
12.
Sci Rep ; 6: 35813, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27779218

RESUMEN

Anorexia nervosa (AN) is associated with low-grade systemic inflammation and altered gut microbiota. However, the molecular origin of the inflammation remains unknown. Toll-like receptors are key regulators of innate immune response and their activation seems also to be involved in the control of food intake. We used activity-based anorexia (ABA) model to investigate the role of TLR4 and its contribution in anorexia-associated low-grade inflammation. Here, we found that ABA affected early the intestinal inflammatory status and the hypothalamic response. Indeed, TLR4 was upregulated both on colonic epithelial cells and intestinal macrophages, leading to elevated downstream mucosal cytokine production. These mucosal changes occurred earlier than hypothalamic changes driving to increased levels of IL-1ß and IL-1R1 as well as increased levels of plasma corticosterone. Paradoxically, TLR4-deficient mice exhibited greater vulnerability to ABA with increased mortality rate, suggesting a major contribution of TLR4-mediated responses during ABA-induced weight loss.


Asunto(s)
Anorexia/fisiopatología , Gastroenteritis/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Anorexia/metabolismo , Colon/metabolismo , Colon/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Gastroenteritis/inmunología , Hipotálamo/inmunología , Hipotálamo/fisiopatología , Mucosa Intestinal/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Condicionamiento Físico Animal , Transducción de Señal , Receptor Toll-Like 4/genética , Pérdida de Peso/genética
13.
Obesity (Silver Spring) ; 24(11): 2361-2367, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27663886

RESUMEN

OBJECTIVE: High-fat diet (HFD) is known to induce low-grade hypothalamic inflammation. Whether inflammation occurs in other brain areas remains unknown. This study tested the effect of short-term HFD on cytokine gene expression and identified leukemia inhibitory factor (LIF) as a responsive cytokine in the brain stem. Thus, functional and cellular effects of LIF in the brain stem were investigated. METHODS: Male rats were fed chow or HFD for 3 days, and then gene expression was analyzed in different brain regions for IL-1ß, IL-6, TNF-α, and LIF. The effect of intracerebroventricular injection of LIF on chow intake and body weight was also tested. Patch clamp recording was performed in the nucleus tractus solitarius (NTS). RESULTS: HFD increased pontine TNF-α mRNA while downregulating LIF in all major parts of the brain stem, but not in the hypothalamus or hippocampus. LIF injection into the cerebral aqueduct suppressed food intake without conditioned taste aversion, suggesting that LIF can induce anorexia via lower brain regions without causing malaise. In the NTS, a key brain stem nucleus for food intake regulation, LIF induced acute changes in neuronal excitability. CONCLUSIONS: HFD-induced downregulation of anorexic LIF in the brain stem may provide a permissive condition for HFD overconsumption. This may be at least partially mediated by the NTS.


Asunto(s)
Anorexia/fisiopatología , Tronco Encefálico/metabolismo , Dieta Alta en Grasa/efectos adversos , Regulación hacia Abajo , Factor Inhibidor de Leucemia/fisiología , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia/administración & dosificación , Masculino , ARN Mensajero/metabolismo , Ratas , Núcleo Solitario/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
14.
PLoS One ; 11(7): e0159607, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27434128

RESUMEN

Acalabrutinib (ACP-196) is a second-generation inhibitor of Bruton agammaglobulinemia tyrosine kinase (BTK) with increased target selectivity and potency compared to ibrutinib. In this study, we evaluated acalabrutinib in spontaneously occurring canine lymphoma, a model of B-cell malignancy similar to human diffuse large B-cell lymphoma (DLBCL). First, we demonstrated that acalabrutinib potently inhibited BTK activity and downstream effectors in CLBL1, a canine B-cell lymphoma cell line, and primary canine lymphoma cells. Acalabrutinib also inhibited proliferation in CLBL1 cells. Twenty dogs were enrolled in the clinical trial and treated with acalabrutinib at dosages of 2.5 to 20mg/kg every 12 or 24 hours. Acalabrutinib was generally well tolerated, with adverse events consisting primarily of grade 1 or 2 anorexia, weight loss, vomiting, diarrhea and lethargy. Overall response rate (ORR) was 25% (5/20) with a median progression free survival (PFS) of 22.5 days. Clinical benefit was observed in 30% (6/20) of dogs. These findings suggest that acalabrutinib is safe and exhibits activity in canine B-cell lymphoma patients and support the use of canine lymphoma as a relevant model for human non-Hodgkin lymphoma (NHL).


Asunto(s)
Antineoplásicos/administración & dosificación , Benzamidas/administración & dosificación , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/veterinaria , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazinas/administración & dosificación , Agammaglobulinemia Tirosina Quinasa , Animales , Anorexia/inducido químicamente , Anorexia/fisiopatología , Antineoplásicos/efectos adversos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Benzamidas/efectos adversos , Línea Celular Tumoral , Diarrea/inducido químicamente , Diarrea/fisiopatología , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Perros , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Letargia/inducido químicamente , Letargia/fisiopatología , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/mortalidad , Masculino , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Pirazinas/efectos adversos , Vómitos/inducido químicamente , Vómitos/fisiopatología , Pérdida de Peso/efectos de los fármacos
15.
Mediators Inflamm ; 2016: 8071539, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27445441

RESUMEN

During an infection, expansion of immune cells, assembly of antibodies, and the induction of a febrile response collectively place continual metabolic strain on the host. These considerations also provide a rationale for nutritional support in critically ill patients. Yet, results from clinical and preclinical studies indicate that aggressive nutritional support does not always benefit patients and may occasionally be detrimental. Moreover, both vertebrates and invertebrates exhibit a decrease in appetite during an infection, indicating that such sickness-associated anorexia (SAA) is evolutionarily conserved. It also suggests that SAA performs a vital function during an infection. We review evidence signifying that SAA may present a mechanism by which autophagic flux is upregulated systemically. A decrease in serum amino acids during an infection promotes autophagy not only in immune cells, but also in nonimmune cells. Similarly, bile acids reabsorbed postprandially inhibit hepatic autophagy by binding to farnesoid X receptors, indicating that SAA may be an attempt to conserve autophagy. In addition, augmented autophagic responses may play a critical role in clearing pathogens (xenophagy), in the presentation of epitopes in nonprovisional antigen presenting cells and the removal of damaged proteins and organelles. Collectively, these observations suggest that some patients might benefit from permissive underfeeding.


Asunto(s)
Anorexia/fisiopatología , Apetito , Enfermedad Crítica/terapia , Terapia Nutricional/métodos , Aminoácidos/sangre , Aminoácidos/química , Animales , Ingestión de Energía , Epítopos/química , Ayuno , Humanos , Sistema Inmunológico , Necesidades Nutricionales , Estado Nutricional , Prevalencia , Receptores Citoplasmáticos y Nucleares/metabolismo , Inanición
16.
Clin Nutr ESPEN ; 11: e1-e11, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-28531420

RESUMEN

Anemia in elderly population have a great incidence and is related to increased mortality risk. The incidence of nutrition in anemia is about one third of the total. Caloric and protein restriction, iron, vitamin B12, folic deficiency are the causes of nutritional anemia. Protein and energy malnutrition stimulate an increased cytokines production with induction of inflammation, immunodeficiency and anemia. Anorexia and obesity can be associated with anemia due to increased cytokines and hepdicin serum level. Macrophages activity is inhibited and a decrease in red blood cells (RBC), hemoglobin (Hb) concentration due to ineffective erythropoiesis is observed. An adequate energy and protein diet is necessary to reduce inflammation and increase iron absorption. A minimum of 1700 kcal/day and 1.7 gr/kg/day of protein intake are necessary to maintain anabolism in chronic patients to prevent and treat anemia. Iron supplementation by intravenous injection is safe and effective to correct severe iron deficiency. The supplementation of vitamins and oligomineral are useful to reduce oxidative stress and improve RBC longevity. Anemia in elderly could be prevented by an adequate nutrition, a simple and not expensive intervention, and associated to physical exercise reduce the incidence of mortality rate.


Asunto(s)
Anemia/prevención & control , Dieta Saludable , Suplementos Dietéticos , Fenómenos Fisiológicos Nutricionales del Anciano , Envejecimiento Saludable , Estilo de Vida Saludable , Cooperación del Paciente , Anciano , Anciano de 80 o más Años , Anemia/epidemiología , Anemia/etiología , Anemia/inmunología , Anemia Ferropénica/epidemiología , Anemia Ferropénica/etiología , Anemia Ferropénica/inmunología , Anemia Ferropénica/prevención & control , Animales , Anorexia/epidemiología , Anorexia/mortalidad , Anorexia/fisiopatología , Anorexia/terapia , Terapia Combinada , Hogares para Ancianos , Humanos , Síndromes de Inmunodeficiencia/epidemiología , Síndromes de Inmunodeficiencia/etiología , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/prevención & control , Desnutrición/epidemiología , Desnutrición/mortalidad , Desnutrición/fisiopatología , Desnutrición/terapia , Casas de Salud , Apoyo Nutricional , Obesidad/epidemiología , Obesidad/mortalidad , Obesidad/fisiopatología , Obesidad/terapia
17.
J Physiol Sci ; 65(5): 385-95, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26123258

RESUMEN

Anorexia, a loss of appetite for food, can be caused by various physiological and pathophysiological conditions. In this review, firstly, clinical aspects of anorexia nervosa are summarized in brief. Secondly, hypothalamic neuropeptides responsible for feeding regulation in each hypothalamic nucleus are discussed. Finally, three different types of anorexigenic animal models; dehydration-induced anorexia, cisplatin-induced anorexia and cancer anorexia-cachexia, are introduced. In conclusion, hypothalamic neuropeptides may give us novel insight to understand and find effective therapeutics strategy essential for various kinds of anorexia.


Asunto(s)
Anorexia/metabolismo , Caquexia/metabolismo , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Animales , Anorexia/etiología , Anorexia/fisiopatología , Anorexia/psicología , Conducta Animal , Caquexia/etiología , Caquexia/fisiopatología , Caquexia/psicología , Cisplatino , Deshidratación/complicaciones , Modelos Animales de Enfermedad , Conducta Alimentaria , Humanos , Hipotálamo/fisiopatología , Neoplasias/complicaciones , Factores de Riesgo , Transducción de Señal , Especificidad de la Especie
18.
Int J Toxicol ; 34(3): 233-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25939350

RESUMEN

To examine 4-week toxicity of l-methionine (methionine), 5-week-old Fisher strain male rats were fed on diets containing 0, 0.1, 0.3, 0.9, 2.7 (w/w) of added methionine. Although no deaths were recorded, the highest dose of methionine (2.7% [w/w] of diet) reduced food intake and significantly suppressed growth rate. Growth suppression was characterized by an increase in hemolysis, splenic, and hepatic accumulation of hemosiderin, hemolytic anemia, and promotion of hematopoiesis. Other changes observed in the highest methionine intake group were a decrease in white blood cell count, thymus atrophy, and histological abnormalities in the adrenal gland and testis. Small, but significant, growth suppression, accompanied by some minor changes in plasma biochemical parameters, was also seen in rats fed on a test diet containing 0.9% (w/w) of additional methionine. Thus, no-observed-adverse-effect-level (NOAEL) and lowest-observed-adverse-effect level (LOAEL) of diet-added methionine were determined at 0.3% and 0.9% (w/w), corresponding to 236 and 705 mg/kg/d body weight, respectively. Since the basal diet contained protein-bound methionine at 0.5% (w/w), NOAEL and LOAEL of total dietary methionine were estimated at 0.8% and 1.4% (w/w) of diet.


Asunto(s)
Alimentación Animal/efectos adversos , Anorexia/etiología , Suplementos Dietéticos/efectos adversos , Trastornos del Crecimiento/etiología , Metionina/envenenamiento , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Anemia Hemolítica/etiología , Animales , Anorexia/metabolismo , Anorexia/patología , Anorexia/fisiopatología , Médula Ósea/metabolismo , Médula Ósea/patología , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/patología , Trastornos del Crecimiento/fisiopatología , Hemosiderosis/etiología , Hígado/metabolismo , Hígado/patología , Masculino , Nivel sin Efectos Adversos Observados , Páncreas/metabolismo , Páncreas/patología , Distribución Aleatoria , Ratas Endogámicas F344 , Bazo/metabolismo , Bazo/patología , Esternón , Testículo/metabolismo , Testículo/patología , Pruebas de Toxicidad Subaguda
19.
Age Ageing ; 44(2): 245-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25324332

RESUMEN

BACKGROUND AND OBJECTIVE: To evaluate the impact of a solid nutritional supplement on the weight gain of institutionalised older adults>70 years with protein-energy malnutrition. The innovation of these high-protein and high-energy cookies was the texture adapted to edentulous patients (Protibis®, Solidages, France). DESIGN: An open, multicentre, randomised controlled trial. SETTING: Seven nursing homes. PARTICIPANTS: One hundred and seventy-five malnourished older adults, aged 86±8 years. INTERVENTION: All participants received the standard institutional diet. In addition, Intervention group participants received eight cookies daily (11.5 g protein; 244 kcal) for 6 weeks (w0-w6). MEASUREMENTS: Five visits (w-4, w0, w6, w10 and w18). MAIN OUTCOME: Percentage of weight gain from w0 to w6 (body mass in kg). SECONDARY OUTCOMES: Appetite, rated using a numerical scale (0: no appetite to 10: extremely good appetite); current episodes of pressure ulcers and diarrhea. RESULTS: Average weight increased in Intervention group (n=88) compared with Control group (n=87) without cookies supplementation (+1.6 versus -0.7%, P=0.038). Weight gain persisted 1 month (+3.0 versus -0.2%, P=0.025) and 3 months after the end of cookies consumption (+3.9 versus -0.9%, P=0.003), with diarrhea reduction (P=0.027). There was a synergistic effect with liquid/creamy dietary supplements. Subgroup analysis confirmed the positive impact of cookies supplementation alone on weight increase (P=0.024), appetite increase (P=0.009) and pressure ulcers reduction (P=0.031). CONCLUSION: The trial suggested that, to fight against anorexia, the stimulation of touch (finger food; chewing, even on edentulous gums) and hearing (intra-oral sounds) could be valuable alternatives to sight, smell and taste alterations.


Asunto(s)
Anorexia/terapia , Nutrición Enteral/métodos , Alimentos Formulados , Hogares para Ancianos , Casas de Salud , Desnutrición Proteico-Calórica/terapia , Pérdida de Peso , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento , Anorexia/diagnóstico , Anorexia/fisiopatología , Anorexia/psicología , Apetito , Proteínas en la Dieta/administración & dosificación , Emociones , Ingestión de Energía , Femenino , Francia , Evaluación Geriátrica , Humanos , Masculino , Evaluación Nutricional , Estado Nutricional , Desnutrición Proteico-Calórica/diagnóstico , Desnutrición Proteico-Calórica/fisiopatología , Desnutrición Proteico-Calórica/psicología , Factores de Tiempo , Resultado del Tratamiento , Aumento de Peso
20.
Metabolism ; 63(11): 1455-61, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25200186

RESUMEN

OBJECTIVE: Estrogen plays an important role in the control of energy balance in the hypothalamus. Leptin-independent STAT3 activation (i.e., tyrosine(705)-phosphorylation of STAT3, pSTAT3) in the hypothalamus is hypothesized as the primary mechanism of the estrogen-induced anorexic response. However, the type of estrogen receptor that mediates this regulation is unknown. We investigated the role of the G protein-coupled receptor 30 (GPR30) in estradiol (E2)-induced STAT3 activation in the hypothalamus. MATERIALS/METHODS: Regulation of STAT3 activation by E2, G-1, a specific agonist of GPR30 and G-15, a specific antagonist of GPR30 was analyzed in vitro and in vivo. Effect of GPR30 activation on eating behavior was analyzed in vivo. RESULTS: E2 stimulated pSTAT3 in cells expressing GPR30, but not expressing estrogen receptor ERα and ERß. G-1 induced pSTAT3, and G-15 inhibited E2-induced pSTAT3 in primary cultures of hypothalamic neurons. A cerebroventricular injection of G-1 increased pSTAT3 in the arcuate nucleus of mice, which was associated with a decrease in food intake and body weight gain. CONCLUSIONS: These results suggest that GPR30 is the estrogen receptor that mediates the anorectic effect of estrogen through the STAT3 pathway in the hypothalamus.


Asunto(s)
Anorexia/fisiopatología , Estrógenos/fisiología , Hipotálamo/fisiopatología , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Células HeLa , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA