Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.198
Filtrar
1.
Nutr Res ; 118: 104-115, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37634306

RESUMEN

The impact of diets high in saturated fatty acids in individuals who have undergone maternal protein restriction is not clear. Here, we tested the hypothesis that a saturated fatty acid-enriched hyperlipidic diet (HL) affects liver expression of genes of the redox balance and inflammatory pathway in postweaning rat offspring subjected to maternal protein restriction. Pregnant Wistar rats received either a control (C; 19% protein) or low protein (LP; 8% protein) diet during gestation and lactation. At weaning, pups received either C or HL diets up to 90 days of life. The LP+HL group showed an upregulation of transcription of peroxisome proliferator-activated receptor γ (+48%) and peroxisome proliferator-activated receptor γ coactivator α (+96%) compared with the LP+C group (P < .05), respectively. Similarly, gene expression of the markers of inflammation, nuclear factor-kappa B1 (+194%) and tumor necrosis factor-α (+192%), was enhanced (P < .05). Although other antioxidant enzymes were not modified in gene expression, catalase (CAT) was 66% higher in LP+HL compared with LP+C. In contrast, CAT protein content in the liver was 50% lower in LP groups compared with C, and superoxide dismutase 2 (SOD2) was twice as high in LP groups compared with C. Postweaning HL after maternal protein restriction induces hepatic metabolic adaptation characterized by enhanced oxidative stress, unbalanced expression in the antioxidant enzymes SOD1, SOD2 and CAT, and activation of inflammatory pathways but does not impact circulating markers of lipid metabolism and liver function.


Asunto(s)
Ácidos Grasos , Deficiencia de Proteína , Embarazo , Femenino , Ratas , Animales , Ácidos Grasos/metabolismo , Ratas Wistar , Antioxidantes/metabolismo , PPAR gamma/metabolismo , Hígado/metabolismo , Estrés Oxidativo , Dieta con Restricción de Proteínas/efectos adversos , Deficiencia de Proteína/metabolismo
2.
Glia ; 70(7): 1337-1358, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35373853

RESUMEN

Survival motor neuron (SMN) protein deficiency results in loss of alpha motor neurons and subsequent muscle atrophy in patients with spinal muscular atrophy (SMA). Reactive microglia have been reported in SMA mice and depleting microglia rescues the number of proprioceptive synapses, suggesting a role in SMA pathology. Here, we explore the contribution of lymphocytes on microglia reactivity in SMA mice and investigate how SMN deficiency alters the reactive profile of human induced pluripotent stem cell (iPSC)-derived microglia. We show that microglia adopt a reactive morphology in spinal cords of SMA mice. Ablating lymphocytes did not alter the reactive morphology of SMA microglia and did not improve the survival or motor function of SMA mice, indicating limited impact of peripheral immune cells on the SMA phenotype. We found iPSC-derived SMA microglia adopted an amoeboid morphology and displayed a reactive transcriptome profile, increased cell migration, and enhanced phagocytic activity. Importantly, cell morphology and electrophysiological properties of motor neurons were altered when they were incubated with conditioned media from SMA microglia. Together, these data reveal that SMN-deficient microglia adopt a reactive profile and exhibit an exaggerated inflammatory response with potential impact on SMA neuropathology.


Asunto(s)
Células Madre Pluripotentes Inducidas , Atrofia Muscular Espinal , Deficiencia de Proteína , Animales , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Microglía/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Deficiencia de Proteína/metabolismo , Deficiencia de Proteína/patología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
3.
Exp Neurol ; 347: 113911, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767796

RESUMEN

Nutritional inadequacy before birth and during postnatal life can seriously interfere with brain development and lead to persistent deficits in learning and behavior. In this work, we asked if protein malnutrition affects domains of social cognition and if these phenotypes can be transmitted to the next generation. Female mice were fed with a normal or hypoproteic diet during pregnancy and lactation. After weaning, offspring were fed with a standard chow. Social interaction, social recognition memory, and dominance were evaluated in both sexes of F1 offspring and in the subsequent F2 generation. Glucose metabolism in the whole brain was analyzed through preclinical positron emission tomography. Genome-wide transcriptional analysis was performed in the medial prefrontal cortex followed by gene-ontology enrichment analysis. Compared with control animals, malnourished mice exhibited a deficit in social motivation and recognition memory and displayed a dominant phenotype. These altered behaviors, except for dominance, were transmitted to the next generation. Positron emission tomography analysis revealed lower glucose metabolism in the medial prefrontal cortex of F1 malnourished offspring. This brain region showed genome-wide transcriptional dysregulation, including 21 transcripts that overlapped with autism-associated genes. Our study cannot exclude that the lower maternal care provided by mothers exposed to a low-protein diet caused an additional impact on social cognition. Our results showed that maternal protein malnutrition dysregulates gene expression in the medial prefrontal cortex, promoting altered offspring behavior that was intergenerationally transmitted. These results support the hypothesis that early nutritional deficiency represents a risk factor for the emergence of symptoms associated with neurodevelopmental disorders.


Asunto(s)
Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/psicología , Efectos Tardíos de la Exposición Prenatal/psicología , Deficiencia de Proteína/complicaciones , Deficiencia de Proteína/psicología , Cognición Social , Animales , Femenino , Humanos , Masculino , Desnutrición/complicaciones , Desnutrición/metabolismo , Desnutrición/psicología , Ratones , Trastornos del Neurodesarrollo/metabolismo , Tomografía de Emisión de Positrones/métodos , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Deficiencia de Proteína/metabolismo
4.
Nutrients ; 13(9)2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34579160

RESUMEN

Protein imbalance during pregnancy affects women in underdeveloped and developing countries and is associated with compromised offspring growth and an increased risk of metabolic diseases in later life. We studied in a porcine model the glucose and urea metabolism, and circulatory hormone and metabolite profile of offspring exposed during gestation, to maternal isoenergetic low-high (LP-HC), high-low (HP-LC) or adequate (AP) protein-carbohydrate ratio diets. At birth, LP-HC were lighter and the plasma acetylcarnitine to free carnitine ratios at 1 day of life was lower compared to AP offspring. Plasma urea concentrations were lower in 1 day old LP-HC offspring than HP-LC. In the juvenile period, increased insulin concentrations were observed in LP-HC and HP-LC offspring compared to AP, as was body weight from HP-LC compared to LP-HC. Plasma triglyceride concentrations were lower in 80 than 1 day old HP-LC offspring, and glucagon concentrations lower in 80 than 1 day old AP and HP-LC offspring. Plasma urea and the ratio of glucagon to insulin were lower in all 80 than 1 day old offspring. Aminoacyl-tRNA, arginine and phenylalanine, tyrosine and tryptophan metabolism, histidine and beta-alanine metabolism differed between 1 and 80 day old AP and HP-LC offspring. Maternal protein imbalance throughout pregnancy did not result in significant consequences in offspring metabolism compared to AP, indicating enormous plasticity by the placenta and developing offspring.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Proteínas en la Dieta/administración & dosificación , Fenómenos Fisiologicos Nutricionales Maternos , Metaboloma , Efectos Tardíos de la Exposición Prenatal/metabolismo , Acetilcarnitina/sangre , Animales , Animales Recién Nacidos/metabolismo , Carnitina/sangre , Carbohidratos de la Dieta/administración & dosificación , Femenino , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Masculino , Embarazo , Deficiencia de Proteína/metabolismo , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo , Triglicéridos/sangre , Urea/sangre , Urea/metabolismo
5.
Sci Rep ; 11(1): 12436, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34127689

RESUMEN

To study, in young growing rats, the consequences of different levels of dietary protein deficiency on food intake, body weight, body composition, and energy balance and to assess the role of FGF21 in the adaptation to a low protein diet. Thirty-six weanling rats were fed diets containing 3%, 5%, 8%, 12%, 15% and 20% protein for three weeks. Body weight, food intake, energy expenditure and metabolic parameters were followed throughout this period. The very low-protein diets (3% and 5%) induced a large decrease in body weight gain and an increase in energy intake relative to body mass. No gain in fat mass was observed because energy expenditure increased in proportion to energy intake. As expected, Fgf21 expression in the liver and plasma FGF21 increased with low-protein diets, but Fgf21 expression in the hypothalamus decreased. Under low protein diets (3% and 5%), the increase in liver Fgf21 and the decrease of Fgf21 in the hypothalamus induced an increase in energy expenditure and the decrease in the satiety signal responsible for hyperphagia. Our results highlight that when dietary protein decreases below 8%, the liver detects the low protein diet and responds by activating synthesis and secretion of FGF21 in order to activate an endocrine signal that induces metabolic adaptation. The hypothalamus, in comparison, responds to protein deficiency when dietary protein decreases below 5%.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Factores de Crecimiento de Fibroblastos/metabolismo , Hipotálamo/metabolismo , Hígado/metabolismo , Deficiencia de Proteína/metabolismo , Animales , Modelos Animales de Enfermedad , Ingestión de Energía , Metabolismo Energético , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Masculino , Deficiencia de Proteína/sangre , Ratas , Respuesta de Saciedad
6.
Aging Cell ; 20(7): e13382, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34128315

RESUMEN

Hematopoietic stem cells (HSCs) reside in a quiescent niche to reserve their capacity of self-renewal. Upon hematopoietic injuries, HSCs enter the cell cycle and encounter protein homeostasis problems caused by accumulation of misfolded proteins. However, the mechanism by which protein homeostasis influences HSC function and maintenance remains poorly understood. Here, we show that C/EBP homologous protein (CHOP), demonstrated previously to induces cell death upon unfolded protein response (UPR), plays an important role in HSCs regeneration. CHOP-/- mice showed normal hematopoietic stem and progenitor cell frequencies in steady state. However, when treated with 5-FU, CHOP deficiency resulted in higher survival rates, associated with an increased number of HSCs and reduced level of apoptosis. In serial competitive transplantation experiments, CHOP-/- HSCs showed a dramatic enhancement of repopulation ability and a reduction of protein aggresomes. Mechanistically, CHOP deletion causes reduced ATF3 expression and further leads to decreased protein aggregation and ROS. In addition, CHOP-/- HSCs exhibited an increased resistance to IR-induced DNA damage and improved HSCs homeostasis and function in telomere dysfunctional (G3Terc-/- ) mice. In summary, these findings disclose a new role of CHOP in the regulation of the HSCs function and homeostasis through reducing ATF3 and ROS signaling.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/deficiencia , Células Madre Hematopoyéticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/fisiología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Ratones , Deficiencia de Proteína/metabolismo
7.
Neurotoxicology ; 85: 54-67, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33964344

RESUMEN

Konzo is a toxico-nutritional upper motor neuron disease causing a spastic paraparesis in schoolchildren and childbearing women in some African countries. Almost a century since the first description of konzo, its underlying etiopathogenic mechanisms and causative agent remain unknown. This paper aims at refreshing the current knowledge of konzo determinants and pathogenesis in order to enlighten potential new research and management perspectives. Literature research was performed in PubMed and Web of Science databases according to the PRISMA methodology. Available data show that cassava-derived cyanide poisoning and protein malnutrition constitute two well-documented risk factors of konzo. However, observational studies have failed to demonstrate the causal relationship between konzo and cyanide poisoning. Thiocyanate, the current marker of choice of cyanide exposure, may underestimate the actual level of cyanide poisoning in konzo patients as a larger amount of cyanide is detoxified via other unusual pathways in the context of protein malnutrition characterizing these patients. Furthermore, the appearance of konzo may be the consequence of the interplay of several factors including cyanide metabolites, nutritional deficiencies, psycho-emotional and geo-environmental factors, resulting in pathophysiologic phenomena such as excitotoxicity or oxidative stress, responsible for neuronal damage that takes place at sparse cellular and/or subcellular levels.


Asunto(s)
Cianuros/envenenamiento , Desnutrición/epidemiología , Manihot/efectos adversos , Enfermedad de la Neurona Motora/inducido químicamente , Enfermedad de la Neurona Motora/epidemiología , Deficiencia de Proteína/epidemiología , África/epidemiología , Proteínas en la Dieta , Humanos , Desnutrición/metabolismo , Enfermedad de la Neurona Motora/metabolismo , Deficiencia de Proteína/metabolismo , Factores de Riesgo , Tiocianatos/metabolismo
8.
Sci Rep ; 11(1): 8003, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33850206

RESUMEN

Very low-density lipoprotein receptor (VLDLR) is a member of the LDL receptor family that is involved in the uptake of VLDL into cells. Increased hepatic VLDLR under endoplasmic reticulum (ER) stress has been shown to cause fatty liver. In this study, the effect of dietary protein restriction on hepatic VLDLR and the role of VLDLR in fatty liver were investigated using Vldlr knockout (KO) mice. Growing wild-type (WT) and KO mice were fed a control diet containing 20% ​​protein or a low protein diet containing 3% protein for 11 days. In WT mice, the amount of hepatic Vldlr mRNA and VLDLR protein increased by approximately 8- and 7-fold, respectively, due to protein restriction. Vldlr mRNA and protein levels increased in both type 1 and type 2 VLDLR. However, neither Vldlr mRNA nor protein levels were significantly increased in heart, muscle, and adipose tissue, demonstrating that VLDLR increase due to protein restriction occurred in a liver-specific manner. Increased liver triglyceride levels during protein restriction occurred in KO mice to the same extent as in WT mice, indicating that increased VLDLR during protein restriction was not the main cause of fatty liver, which was different from the case of ER stress.


Asunto(s)
Hígado Graso/complicaciones , Hígado Graso/metabolismo , Hígado/metabolismo , Deficiencia de Proteína/complicaciones , Deficiencia de Proteína/metabolismo , Receptores de LDL/metabolismo , Animales , Apolipoproteínas E/deficiencia , Aspartatoamoníaco Ligasa/genética , Aspartatoamoníaco Ligasa/metabolismo , Dieta con Restricción de Proteínas , Hígado Graso/sangre , Factores de Crecimiento de Fibroblastos/deficiencia , Regulación de la Expresión Génica , Inflamación/sangre , Inflamación/complicaciones , Lípidos/sangre , Hígado/lesiones , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Deficiencia de Proteína/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/genética
9.
J Nutr Biochem ; 93: 108626, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33705953

RESUMEN

Protein malnutrition causes anemia and leukopenia as it reduces hematopoietic precursors and impairs the production of mediators that regulate hematopoiesis. Hematopoiesis occurs in distinct bone marrow niches that modulate the processes of differentiation, proliferation and self-renewal of hematopoietic stem cells (HSCs). Mesenchymal stem cells (MSCs) contribute to the biochemical composition of bone marrow niches by the secretion of several growth factors and cytokines, and they play an important role in the regulation of HSCs and hematopoietic progenitors. In this study, we investigated the effect of protein malnutrition on the hematopoietic regulatory function of MSCs. C57BL/6NTaq mice were divided into control and protein malnutrition groups, which received, respectively, a normal protein diet (12% casein) and a low protein diet (2% casein). The results showed that protein malnutrition altered the synthesis of SCF, TFG-ß, Angpt-1, CXCL-12, and G-CSF by MSCs. Additionally, MSCs from the protein malnutrition group were not able to maintain the lymphoid, granulocytic and megakaryocytic-erythroid differentiation capacity compared to the MSCs of the control group. In this way, the comprehension of the role of MSCs on the regulation of the hematopoietic cells, in protein malnutrition states, is for the first time showed. Therefore, we infer that hematopoietic alterations caused by protein malnutrition are due to multifactorial alterations and, at least in part, the MSCs' contribution to hematological impairment.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Proteínas en la Dieta/administración & dosificación , Hematopoyesis/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Deficiencia de Proteína/metabolismo , Animales , Células de la Médula Ósea/fisiología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Hematopoyesis/fisiología , Leucocitos Mononucleares/fisiología , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN/efectos de los fármacos , ARN/genética , ARN/metabolismo
10.
Int J Exp Pathol ; 102(2): 93-104, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33729619

RESUMEN

In a large part of the population inefficient ingestion of proteins, whether for cultural, aesthetic or economic reasons, is a global concern. Low-protein diets can cause severe functional complications, mainly during the development and maturation of organs and systems, including the female reproductive system. The present study investigated the effect of nutritional protein restriction during puberty on the oestrous cycle and expression of sex steroid receptors (AR, ERα e ERß) in ovarian and uterine tissues of adult rats. Protein restriction promoted lower body weight gain, feed efficiency and higher caloric intake. There was an increase in the oestrus phase arrest without changing the total length of the oestrous cycle. The consumption of low-protein diet also reduced the thickness of the uterine endometrium (uterine epithelium and endometrial stroma) in addition to increasing the number of primary and atretic follicles in the ovaries. Furthermore, the low-protein diet reduced the levels of androgen receptor (AR) and increased the oestrogen receptor ß (ERß) in the ovary, while no significant changes were observed in the uterus. Our study reinforces the importance of adequate protein intake during puberty, since physiological changes in this developmental period interfere with the histomorphometry of the ovaries and uteri, possibly resulting in impaired folliculogenesis and fertility in the reproductive period.


Asunto(s)
Ciclo Estral/fisiología , Ovario/patología , Deficiencia de Proteína/fisiopatología , Maduración Sexual/fisiología , Útero/patología , Animales , Femenino , Ovario/metabolismo , Deficiencia de Proteína/metabolismo , Deficiencia de Proteína/patología , Ratas , Ratas Endogámicas F344 , Útero/metabolismo
11.
Br J Nutr ; 125(6): 633-643, 2021 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-32814607

RESUMEN

Protein undernutrition contributes to the development of various diseases in broad generations. Urinary metabolites may serve as non-invasive biomarkers of protein undernutrition; however, this requires further investigation. We aimed to identify novel urinary metabolites as biomarker candidates responsive to protein undernutrition. Adult rats were fed control (CT; 14 % casein) or isoenergetic low-protein (LP; 5 % casein) diets for 4 weeks. 1H NMR metabolomics was applied to urine, plasma and liver samples to identify metabolites responsive to protein undernutrition. Liver samples were subjected to mRNA microarray and quantitative PCR analyses to elucidate the mechanisms causing fluctuations in identified metabolites. Urinary taurine levels were significantly lower in the LP group than in the CT group at week 1 and remained constant until week 4. Hepatic taurine level and gene expression level of cysteine dioxygenase type 1 were also significantly lower in the LP group than in the CT group. Urinary trimethylamine N-oxide (TMAO) levels were significantly higher in the LP group than in the CT group at week 2 and remained constant until week 4. Hepatic TMAO level and gene expression levels of flavin-containing mono-oxygenase 1 and 5 were also significantly higher in the LP group than in the CT group. In conclusion, urinary taurine and TMAO levels substantially responded to protein undernutrition. Furthermore, changes in hepatic levels of these metabolites and gene expressions associated with their metabolic pathways were also reflected in their fluctuating urinary levels. Thus, taurine and TMAO could act as non-invasive urinary biomarker candidates to detect protein undernutrition.


Asunto(s)
Metilaminas/orina , Deficiencia de Proteína/orina , Taurina/orina , Animales , Biomarcadores/orina , Cisteína-Dioxigenasa/genética , Cisteína-Dioxigenasa/metabolismo , Dieta con Restricción de Proteínas , Perfilación de la Expresión Génica , Ontología de Genes , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Metaboloma , Deficiencia de Proteína/sangre , Deficiencia de Proteína/diagnóstico , Deficiencia de Proteína/metabolismo , Ratas , Ratas Wistar , Transcriptoma
12.
Neurochem Res ; 45(1): 68-82, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31664654

RESUMEN

Hapln4 is a link protein which stabilizes the binding between lecticans and hyaluronan in perineuronal nets (PNNs) in specific brain regions, including the medial nucleus of the trapezoid body (MNTB). The aim of this study was: (1) to reveal possible age-related alterations in the extracellular matrix composition in the MNTB and inferior colliculus, which was devoid of Hapln4 and served as a negative control, (2) to determine the impact of the Hapln4 deletion on the values of the ECS diffusion parameters in young and aged animals and (3) to verify that PNNs moderate age-related changes in the ECS diffusion, and that Hapln4-brevican complex is indispensable for the correct protective function of the PNNs. To achieve this, we evaluated the ECS diffusion parameters using the real-time iontophoretic method in the selected region in young adult (3 to 6-months-old) and aged (12 to 18-months-old) wild type and Hapln4 knock-out (KO) mice. The results were correlated with an immunohistochemical analysis of the ECM composition and astrocyte morphology. We report that the ECM composition is altered in the aged MNTB and aging is a critical point, revealing the effect of Hapln4 deficiency on the ECS diffusion. All of our findings support the hypothesis that the ECM changes in the MNTB of aged KO animals affect the ECS parameters indirectly, via morphological changes of astrocytes, which are in direct contact with synapses and can be influenced by the ongoing synaptic transmission altered by shifts in the ECM composition.


Asunto(s)
Envejecimiento/metabolismo , Vías Auditivas/metabolismo , Difusión , Proteínas de la Matriz Extracelular/deficiencia , Espacio Extracelular/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Cuerpo Trapezoide/metabolismo , Envejecimiento/patología , Animales , Vías Auditivas/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Deficiencia de Proteína/metabolismo , Deficiencia de Proteína/patología , Cuerpo Trapezoide/patología
13.
J Pediatr Hematol Oncol ; 42(8): e768-e771, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31876783

RESUMEN

In recent years, monogenic causes of immune dysregulation syndromes, with variable phenotypes, have been documented. Mutations in the lipopolysaccharide-responsive beige-like anchor (LRBA) protein are associated with common variable immunodeficiency, autoimmunity, chronic enteropathy, and immune dysregulation disorders. The LRBA protein prevents degradation of cytotoxic T-lymphocyte antigen 4 (CTLA4) protein, thus inhibiting immune responses. Both LRBA and CTLA4 deficiencies usually present with immune dysregulation, mostly characterized by autoimmunity and lymphoproliferation. In this report, we describe a patient with an atypical clinical onset of LRBA deficiency and the patient's response to abatacept, a fusion protein-drug that mimics the action of CTLA4.


Asunto(s)
Abatacept/uso terapéutico , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Antígeno CTLA-4/agonistas , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Deficiencia de Proteína/tratamiento farmacológico , Enteropatías Perdedoras de Proteínas/tratamiento farmacológico , Edad de Inicio , Antígeno CTLA-4/deficiencia , Preescolar , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Inmunosupresores/uso terapéutico , Masculino , Pronóstico , Deficiencia de Proteína/complicaciones , Deficiencia de Proteína/metabolismo , Deficiencia de Proteína/patología , Enteropatías Perdedoras de Proteínas/complicaciones , Enteropatías Perdedoras de Proteínas/metabolismo , Enteropatías Perdedoras de Proteínas/patología
14.
J Cell Physiol ; 234(5): 6313-6323, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30317568

RESUMEN

Nutrient malnutrition, during the early stages of development, may facilitate the onset of metabolic diseases later in life. However, the consequences of nutritional insults, such as a high-fat diet (HFD) after protein restriction, are still controversial. We assessed overall glucose homeostasis and molecular markers of mitochondrial function in the gastrocnemius muscle of protein-restricted mice fed an HFD until early adulthood. Male C57BL/6 mice were fed a control (14% protein-control diet) or a protein-restricted (6% protein-restricted diet) diet for 6 weeks. Afterward, mice received an HFD or not for 8 weeks (mice fed a control diet and HFD [CH] and mice fed a protein-restricted diet and HFD [RH]). RH mice showed lower weight gain and fat accumulation and did not show an increase in fasting plasma glucose and insulin levels compared with CH mice. RH mice showed higher energy expenditure, increased citrate synthase, peroxisome-proliferator-activated receptor gamma coactivator 1-alpha protein content, and higher levels of malate and α-ketoglutarate compared with CH mice. Moreover, RH mice showed increased AMPc-dependent kinase and acetyl coenzyme-A (CoA) carboxylase phosphorylation, lower intramuscular triacylglycerol content, and similar malonyl-CoA levels. In conclusion, protein undernourishment after weaning does not potentiate fat accumulation and insulin resistance in adult young mice fed an HFD. This outcome seems to be associated with increased skeletal muscle mitochondrial oxidative capacity and reduced lipids accumulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Homeostasis/fisiología , Músculo Esquelético/metabolismo , Deficiencia de Proteína/metabolismo , Animales , Metabolismo Energético/fisiología , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo
15.
Biomed Pharmacother ; 109: 610-620, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30399598

RESUMEN

The occurrence of inflammation and protein malnutrition is an aggravating risk factor for morbidity and mortality in the clinical setting. The green propolis, a natural product made by Apis mellifera bees from Baccharis dracunculifolia resin, has therapeutic potential to modulate chronic inflammation. However, its effect on inflammation in an impaired nutritional status is not known. The aim of this study was to characterize the effects of the administration of the hydroalcoholic extract of the green propolis in the chronic inflammatory process of mice submitted to a low-protein diet. For this, we used the subcutaneous implantation of sponge disks as an inflammatory model and the animals were distributed in the following groups: standard protein diet (12% protein content), control treatment; standard protein diet, propolis treatment; low-protein diet (3% protein content), control treatment; low-protein diet, propolis treatment. Propolis was given daily at a dose of 500 mg/kg (p.o.) during a period of 7 or 15 days. Our main findings show that animals fed with standard protein diet and treated with propolis had low levels of red blood cells, hemoglobin, and hematocrit, with the subsequent reestablishment of these levels, in addition to monocyte count elevation and higher TNF levels after one week of treatment. In the low-protein diet group, the propolis treatment provided a significant recovery in weight and maintenance of total serum protein levels at the end of two weeks of treatment. Histological analysis showed propolis reduced the inflammatory infiltrate in the sponges of both standard and low-protein diet groups. In addition, the propolis extract presented antiangiogenic effect in both groups. Therefore, our data suggests that the hydroalcoholic extract of the green propolis promotes weight recovery and avoid the reduction of protein levels, in addition to inhibit inflammation and angiogenesis in animals fed with a low-protein diet.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Mediadores de Inflamación/metabolismo , Própolis/administración & dosificación , Deficiencia de Proteína/tratamiento farmacológico , Deficiencia de Proteína/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Etanol/administración & dosificación , Femenino , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Ratones , Deficiencia de Proteína/inducido químicamente , Distribución Aleatoria , Agua/administración & dosificación
17.
Physiol Rep ; 6(23): e13932, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30516001

RESUMEN

Malnutrition remains a major health problem in low- and middle-income countries. During low protein intake, <0.67 g/kg/day, there is a loss of nitrogen (N2 ) balance, due to the unavailability of amino acid for metabolism and unbalanced protein catabolism results. However, there are individuals, who consume the same low protein intake, and preserve N2 balance for unknown reasons. A novel factor, the gut microbiota, may account for these N2 balance differences. To investigate this, we correlated gut microbial profiles with the growth of four murine strains (C57Bl6/J, CD-1, FVB, and NIH-Swiss) on protein deficient (PD) diet. Results show that a PD diet exerts a strain-dependent impact on growth and N2 balance as determined through analysis of urinary urea, ammonia and creatinine excretion. Bacterial alpha diversity was significantly (P < 0.05, FDR) lower across all strains on a PD diet compared to normal chow (NC). Multi-group analyses of the composition of microbiomes (ANCOM) revealed significantly differential microbial signatures between the four strains independent of diet. However, mice on a PD diet demonstrated differential enrichment of bacterial genera including, Allobaculum (C57Bl6/J), Parabacteroides (CD-1), Turicibacter (FVB), and Mucispirillum (NIH-Swiss) relative to NC. For instance, selective comparison of the CD-1 (gained weight) and C57Bl6/J (did not gain weight) strains on PD diet also demonstrated significant pathway enrichment of dihydroorodate dehydrogenase, rRNA methyltransferases, and RNA splicing ligase in the CD-1 strains compared to C57Bl6/J strains; which might account in their ability to retain growth despite a protein deficient diet. Taken together, these results suggest a potential relationship between the specific gut microbiota, N2 balance and animal response to malnutrition.


Asunto(s)
Microbioma Gastrointestinal , Deficiencia de Proteína/microbiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Nitrógeno/metabolismo , Fenotipo , Deficiencia de Proteína/metabolismo
18.
Vaccine ; 36(42): 6270-6281, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30219368

RESUMEN

BACKGROUND: Low efficacy of rotavirus (RV) vaccines in developing African and Asian countries, where malnutrition is prevalent, remains a major concern and a challenge for global health. METHODS: To understand the effects of protein malnutrition on RV vaccine efficacy, we elucidated the innate, T cell and cytokine immune responses to attenuated human RV (AttHRV) vaccine and virulent human RV (VirHRV) challenge in germ-free (GF) pigs or human infant fecal microbiota (HIFM) transplanted gnotobiotic (Gn) pigs fed protein-deficient or -sufficient bovine milk diets. We also analyzed serum levels of tryptophan (TRP), a predictor of malnutrition, and kynurenine (KYN). RESULTS: Protein-deficient pigs vaccinated with oral AttHRV vaccine had lower protection rates against diarrhea post-VirHRV challenge and significantly increased fecal virus shedding titers (HIFM transplanted but not GF pigs) compared with their protein-sufficient counterparts. Reduced vaccine efficacy in protein-deficient pigs coincided with altered serum IFN-α, TNF-α, IL-12 and IFN-γ responses to oral AttHRV vaccine and the suppression of multiple innate immune parameters and HRV-specific IFN-γ producing T cells post-challenge. In protein-deficient HIFM transplanted pigs, decreased serum KYN, but not TRP levels were observed throughout the experiment, suggesting an association between the altered TRP metabolism and immune responses. CONCLUSION: Collectively, our findings confirm the negative effects of protein deficiency, which were exacerbated in the HIFM transplanted pigs, on innate, T cell and cytokine immune responses to HRV and on vaccine efficacy, as well as on TRP-KYN metabolism.


Asunto(s)
Heces/microbiología , Vida Libre de Gérmenes , Deficiencia de Proteína/complicaciones , Vacunas Atenuadas/uso terapéutico , Animales , Humanos , Lactante , Microbiota/inmunología , Deficiencia de Proteína/inmunología , Deficiencia de Proteína/metabolismo , Rotavirus/inmunología , Rotavirus/patogenicidad , Vacunas contra Rotavirus/uso terapéutico , Porcinos , Triptófano/metabolismo
19.
Lakartidningen ; 1152018 05 21.
Artículo en Sueco | MEDLINE | ID: mdl-29786804

RESUMEN

There is a widespread myth that we have to be careful about what we eat so that we do not cause protein deficiency. We know today that it is virtually impossible to design a calorie-sufficient diet, whether it is based on meat, fish, eggs, various vegetarian diets or even unprocessed whole natural plant foods, which is lacking in protein and any of the amino acids. The body is capable of taking incomplete proteins and making them complete by utilizing the amino acid recycling mechanism. The majority of amino acids absorbed from the intestinal tract are derived from recycled body protein. Research shows that high levels of animal protein intake may significantly increase the risk of premature mortality from all causes, among them cardiovascular diseases, cancer and type 2 diabetes.


Asunto(s)
Dieta Rica en Proteínas/efectos adversos , Deficiencia de Proteína , Animales , Dieta , Dieta Vegana , Peces , Humanos , Política Nutricional , Deficiencia de Proteína/etiología , Deficiencia de Proteína/metabolismo
20.
Int J Parasitol ; 48(1): 51-58, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28903026

RESUMEN

Maternal dietary protein deficiency and gastrointestinal nematode infection during early pregnancy have negative impacts on both maternal placental gene expression and fetal growth in the mouse. Here we used next-generation RNA sequencing to test our hypothesis that maternal protein deficiency and/or nematode infection also alter the expression of genes in the developing fetal brain. Outbred pregnant CD1 mice were used in a 2×2 design with two levels of dietary protein (24% versus 6%) and two levels of infection (repeated sham versus Heligmosomoides bakeri beginning at gestation day 5). Pregnant dams were euthanized on gestation day 18 to harvest the whole fetal brain. Four fetal brains from each treatment group were analyzed using RNA Hi-Seq sequencing and the differential expression of genes was determined by the edgeR package using NetworkAnalyst. In response to maternal H. bakeri infection, 96 genes (88 up-regulated and eight down-regulated) were differentially expressed in the fetal brain. Differentially expressed genes were involved in metabolic processes, developmental processes and the immune system according to the PANTHER classification system. Among the important biological functions identified, several up-regulated genes have known neurological functions including neuro-development (Gdf15, Ing4), neural differentiation (miRNA let-7), synaptic plasticity (via suppression of NF-κß), neuro-inflammation (S100A8, S100A9) and glucose metabolism (Tnnt1, Atf3). However, in response to maternal protein deficiency, brain-specific serine protease (Prss22) was the only up-regulated gene and only one gene (Dynlt1a) responded to the interaction of maternal nematode infection and protein deficiency. In conclusion, maternal exposure to GI nematode infection from day 5 to 18 of pregnancy may influence developmental programming of the fetal brain.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Fetales/genética , Herencia Materna , Complicaciones del Embarazo/genética , Deficiencia de Proteína/embriología , Trichostrongyloidea/fisiología , Tricostrongiloidiasis/parasitología , Animales , Encéfalo/embriología , Encéfalo/parasitología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Femenino , Desarrollo Fetal , Enfermedades Fetales/metabolismo , Enfermedades Fetales/parasitología , Enfermedades Fetales/fisiopatología , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/parasitología , Deficiencia de Proteína/genética , Deficiencia de Proteína/metabolismo , Deficiencia de Proteína/parasitología , Trichostrongyloidea/genética , Trichostrongyloidea/aislamiento & purificación , Tricostrongiloidiasis/embriología , Tricostrongiloidiasis/genética , Tricostrongiloidiasis/metabolismo , Troponina T/genética , Troponina T/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...