Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118.057
Filtrar
1.
J Coll Physicians Surg Pak ; 34(9): 1029-1034, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39262000

RESUMO

OBJECTIVE: To determine the long-term effects of early postnatal malnutrition and various degrees of catch-up growth on metabolic (serum glucose, leptin, triacylglycerides) and neurodevelopmental parameters (learning and memory) among male and female rodent models, mimicking human preterm infants. STUDY DESIGN: Randomized controlled trial. Place and Duration of the Study: CMH Multan Insitute of Medical Sciences, from September 2021 to December 2021. METHDOLOGY: This study included 142 neonatal Wister rats, stratified into subgroups to mimic the human preterm infant model of postnatal malnutrition and catch-up growth. Metabolic consequences were assessed via serum analysis of glucose, leptin, and triacylglycerides. The neurocognitive comparison was made among subgroups via a passive avoidance test. Gender-specific comparison of all quantitative parameters was made among subgroups. RESULTS: Malnourished rats with accelerated catch-up growth achieved similar weight gain as normally fed rats when provided with adlibitum feeding in both males (p = 0.92) and females (p >0.99). Rats undergoing accelerated catch-up growth exhibited higher fasting serum glucose levels compared to those undergoing no, or normal catch-up growth (p <0.001). Malnourished female rats undergoing accelerated (p = 0.007), or no catchup growth (p = 0.004) exhibited significant deficits in learning and memory as compared to normally fed rats. Female malnourished rats with normal catchup growth exhibited no neurocognitive deficit as compared to normally fed rats (p = 0.08). CONCLUSION: Accelerated catch-up growth effectively addresses somatic growth disparities, while normal catch-up growth offers more favourable metabolic and neurodevelopmental outcomes. Particularly, female malnourished rats exhibited poor neurodevelopment in response to both accelerated and no catch-up growth. Gender-specific variations in neurodevelopment underscore the need for personalised care approaches for preterm nutritional care. KEY WORDS: Growth retardation, Leptin, Extrauterine growth restriction, Malnutrition, Neurodevelopment.


Assuntos
Ratos Wistar , Animais , Ratos , Feminino , Masculino , Animais Recém-Nascidos , Glicemia/metabolismo , Fatores Sexuais , Modelos Animais de Doenças , Leptina/sangue , Desnutrição , Triglicerídeos/sangue
2.
Food Res Int ; 195: 114993, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39277254

RESUMO

Recent studies have shown that 1-oleo-2-palmito-3-linoleyl glycerol (OPL) is the most abundant triacylglycerol in human breast milk in China. Epidemiologic studies have shown that sn-2 palmitate improves the absorption of fatty acids and calcium in infants. However, there have been few studies of the specific mechanism by which OPL affects intestinal function. In the present study, we have characterized the effects of various levels of OPL supplementation on the development of the intestinal epithelium and the intestinal microbiota of neonatal mice. OPL supplementation increased the body masses and intestinal lengths of weaned mice and promoted defecation. These positive effects were related to the effect of OPL to promote the development of intestinal villi and crypts. OPL increased the expression of the intestinal stem cell markers Olfm4 and Sox9 in the jejunum and ileum, which promoted their differentiation into goblet cells and Paneth cells. It also promoted the integrity of the epithelial barrier by increasing the secretion of mucin 2 and lysozyme 1 and the expression of the tight junction proteins occludin, ZO1, claudin 2, and claudin 3. More importantly, we found that low dose-OPL promotes the transformation of the intestinal microbiota of neonatal mice to the mature state in 3-month-old mice, increases the proportion of Firmicutes, and reduces the proportion of Bacteroidota. The proportions of anaerobic genera of bacteria, such as Lachnospiraceae_NK4A136_group, Lachnoclostridium, Ligilactobacillus, and Bifidobacterium were higher, as were the key producers of short-chain fatty acids, such as Bacteroides and Blautia. OPL also increased the butyric acid content of the feces, which significantly correlated with the abundance of Lactobacillus. High-dose OPL tended to be more effective at promoting defecation and the development of the villi and crypts, but these effects did not significantly differ from those achieved using the lower dose. A low dose of OPL was more effective at increasing the butyric acid content and causing the maturation of microbes. In summary, the OPL supplementation of newborn mice promotes the establishment of the intestinal epithelial layer structure and barrier function, and also promotes the transformation of the intestinal microbiota to a mature state. This study lays a theoretical foundation for the inclusion of OPL in infant formula and provides a scientific basis for the development of intestinal health products.


Assuntos
Animais Recém-Nascidos , Suplementos Nutricionais , Microbioma Gastrointestinal , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Intestino Delgado/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/microbiologia , Mucosa Intestinal/metabolismo , Masculino , Glicerídeos/metabolismo , Camundongos Endogâmicos C57BL , Ácidos Oleicos
3.
Nat Commun ; 15(1): 8066, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39277581

RESUMO

High mitochondrial DNA (mtDNA) amount has been reported to be beneficial for resistance and recovery of metabolic stress, while increased mtDNA synthesis activity can drive aging signs. The intriguing contrast of these two mtDNA boosting outcomes prompted us to jointly elevate mtDNA amount and frequency of replication in mice. We report that high activity of mtDNA synthesis inhibits perinatal metabolic maturation of the heart. The offspring of the asymptomatic parental lines are born healthy but manifest dilated cardiomyopathy and cardiac collapse during the first days of life. The pathogenesis, further enhanced by mtDNA mutagenesis, involves prenatal upregulation of mitochondrial integrated stress response and the ferroptosis-inducer MESH1, leading to cardiac fibrosis and cardiomyocyte death after birth. Our evidence indicates that the tight control of mtDNA replication is critical for early cardiac homeostasis. Importantly, ferroptosis sensitivity is a potential targetable mechanism for infantile-onset cardiomyopathy, a common manifestation of mitochondrial diseases.


Assuntos
Replicação do DNA , DNA Mitocondrial , Miócitos Cardíacos , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Feminino , Masculino , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Ferroptose/genética , Miocárdio/metabolismo , Miocárdio/patologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/genética , Camundongos Endogâmicos C57BL , Animais Recém-Nascidos , Humanos , Coração/fisiopatologia , Fibrose
4.
BMC Vet Res ; 20(1): 404, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39256763

RESUMO

BACKGROUND: The peripartum period constitutes a delicate physiological moment in mares showing a transient state of oxidative stress. Diet supplementation with antioxidants during pregnancy in women appears to have a beneficial effect on mother and neonate health. The aim of this work was to evaluate the effects of diet supplementation with a commercial product containing a mix of antioxidants (Oxyliver®, Candioli) on the length of gestation, weight, and haemato-biochemical parameters in Italian Salernitano mares and their newborn foals. Eight late-term pregnant mares were randomly divided into two groups: Antiox group receiving 30 g/day of antioxidants, and Car group receiving the same amount of carrot powder, from 290 to 320 days of gestation. The following parameters were evaluated in mares: weight, colostrum composition, haemato-biochemical parameters, progesterone, and cortisol blood concentrations, along with blood oxidant/antioxidant status. Assessments were conducted at specific time points: immediately before the start of diet supplementation (T0), 15 days after (T1), at the end of diet supplementation (T2), within 8 h after parturition (T3), and 10 days post-partum (T4). Foal parameters such as weight, haemato-biochemical values, cortisol concentration, and blood oxidative stress variables were assessed within 8 h of birth (TF0) and at 10 days of age (TF1). RESULTS: Pregnancy was shorter in the Antiox group (P < 0.05) compared with the Car group; the foals' weight increase of group Antiox (40%) was higher (P < 0.05) compared to those of the Car group (28.6%). The colostrum of the Antiox group exhibited higher levels of Brix, total solids, protein, nonfat solids, casein, urea, density, free fatty acids, and glucose, while lower levels of fat and lactose were observed compared to the Car group (P < 0.05). Mares' serum albumin at T1 and T3, creatinine, glucose, total proteins, total bilirubin, AST, and ALT at T3 were lower in Antiox than in the Car group. No significant differences were found in foals. CONCLUSIONS: While the limited sample size and the potential variability of evaluated parameters, the observed outcomes suggest that Oxyliver® supplementation in mares might safely decrease gestation length and enhance liver function, thus potentially improving colostrum quality and offspring development.


Assuntos
Animais Recém-Nascidos , Antioxidantes , Dieta , Suplementos Nutricionais , Animais , Cavalos , Feminino , Antioxidantes/administração & dosagem , Gravidez , Dieta/veterinária , Ração Animal/análise , Estresse Oxidativo/efeitos dos fármacos , Colostro/química
5.
Stem Cell Res Ther ; 15(1): 295, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39256862

RESUMO

BACKGROUND: Premature infants requiring mechanical ventilation and supplemental oxygen for respiratory support are at increased risk for bronchopulmonary dysplasia (BPD), wherein inflammation have been proposed as a driver of hyperoxia-induced injuries, including persistent loss of endothelial progenitor cells (EPCs), impaired vascularization and eventual alveolar simplification in BPD lungs. However, the underlying mechanisms linking these phenomena remain poorly defined. METHODS: We used clodronate liposomes to deplete macrophages in a mouse model of neonatal hyperoxia-induced lung injury to evaluate if EPC loss in BPD lungs could be an effect of macrophage infiltration. We further generated in vitro culture systems initiated with cord blood (CB)-derived CD34+ EPCs and neonatal macrophages either polarized from CB-derived monocytes or isolated from tracheal aspirates of human preterm infants requiring mechanical ventilation and oxygen supplementation, to identify EV-transmitted molecular mechanism that is critical for inhibitory actions of hyperoxic macrophages on EPCs. RESULTS: Initial experiments using mouse model identified the crucial role of macrophage infiltration in eliciting significant reduction of c-Kit+ EPCs in BPD lungs. Further examination of this concept in human system, we found that hyperoxia-exposed neonatal macrophages hamper human CD34+ EPC maintenance and impair endothelial function in the differentiated progeny via the EV transmission of miR-23a-3p. Notably, treatment with antagomiR-23a-3p to silence miR-23a-3p in vivo enhances c-Kit+ EPC maintenance, and increases capillary density, and consequently mitigates simplified alveolarization in BPD lungs. CONCLUSION: Our findings highlight the importance of pulmonary intercellular communication in the pathophysiology of BPD, by identifying a linkage through vesicle transfer of miR-23a-3p from hyperoxic macrophages to EPCs, and thus demonstrating potential for novel therapeutic target in BPD.


Assuntos
Células Progenitoras Endoteliais , Vesículas Extracelulares , Hiperóxia , Lesão Pulmonar , Macrófagos , MicroRNAs , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Animais , Células Progenitoras Endoteliais/metabolismo , Hiperóxia/metabolismo , Vesículas Extracelulares/metabolismo , Camundongos , Macrófagos/metabolismo , Lesão Pulmonar/patologia , Lesão Pulmonar/metabolismo , Recém-Nascido , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/genética , Animais Recém-Nascidos , Modelos Animais de Doenças
6.
Nat Cardiovasc Res ; 3(9): 1083-1097, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39223390

RESUMO

The neonatal mammalian heart can regenerate following injury through cardiomyocyte proliferation but loses this potential by postnatal day 7. Stimulating adult cardiomyocytes to reenter the cell cycle remains unclear. Here we show that cardiomyocyte proliferation depends on its metabolic state. Given the connection between the tricarboxylic acid cycle and cell proliferation, we analyzed these metabolites in mouse hearts from postnatal day 0.5 to day 7 and found that α-ketoglutarate ranked highest among the decreased metabolites. Injection of α-ketoglutarate extended the window of cardiomyocyte proliferation during heart development and promoted heart regeneration after myocardial infarction by inducing adult cardiomyocyte proliferation. This was confirmed in Ogdh-siRNA-treated mice with increased α-ketoglutarate levels. Mechanistically, α-ketoglutarate decreases H3K27me3 deposition at the promoters of cell cycle genes in cardiomyocytes. Thus, α-ketoglutarate promotes cardiomyocyte proliferation through JMJD3-dependent demethylation, offering a potential approach for treating myocardial infarction.


Assuntos
Proliferação de Células , Histona Desmetilases com o Domínio Jumonji , Ácidos Cetoglutáricos , Infarto do Miocárdio , Miócitos Cardíacos , Regeneração , Animais , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Animais Recém-Nascidos , Células Cultivadas , Histonas/metabolismo , Camundongos , Complexo Cetoglutarato Desidrogenase/metabolismo , Complexo Cetoglutarato Desidrogenase/genética , Masculino
7.
Molecules ; 29(17)2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39275007

RESUMO

Agathisflavone is a flavonoid that exhibits anti-inflammatory and anti-oxidative properties. Here, we investigated the neuroprotective effects of agathisflavone on central nervous system (CNS) neurons and glia in the cerebellar slice ex vivo model of neonatal ischemia. Cerebellar slices from neonatal mice, in which glial fibrillary acidic protein (GFAP) and SOX10 drive expression of enhanced green fluorescent protein (EGFP), were used to identify astrocytes and oligodendrocytes, respectively. Agathisflavone (10 µM) was administered preventively for 60 min before inducing ischemia by oxygen and glucose deprivation (OGD) for 60 min and compared to controls maintained in normal oxygen and glucose (OGN). The density of SOX-10+ oligodendrocyte lineage cells and NG2 immunopositive oligodendrocyte progenitor cells (OPCs) were not altered in OGD, but it resulted in significant oligodendroglial cell atrophy marked by the retraction of their processes, and this was prevented by agathisflavone. OGD caused marked axonal demyelination, determined by myelin basic protein (MBP) and neurofilament (NF70) immunofluorescence, and this was blocked by agathisflavone preventative treatment. OGD also resulted in astrocyte reactivity, exhibited by increased GFAP-EGFP fluorescence and decreased expression of glutamate synthetase (GS), and this was prevented by agathisflavone pretreatment. In addition, agathisflavone protected Purkinje neurons from ischemic damage, assessed by calbindin (CB) immunofluorescence. The results demonstrate that agathisflavone protects neuronal and myelin integrity in ischemia, which is associated with the modulation of glial responses in the face of ischemic damage.


Assuntos
Animais Recém-Nascidos , Cerebelo , Flavonoides , Fármacos Neuroprotetores , Animais , Fármacos Neuroprotetores/farmacologia , Camundongos , Cerebelo/metabolismo , Cerebelo/efeitos dos fármacos , Flavonoides/farmacologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Glucose/metabolismo , Biflavonoides
8.
Commun Biol ; 7(1): 1121, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39261625

RESUMO

Neurodevelopmental disorders are currently one of the major complications faced by patients with congenital heart disease (CHD). Chronic hypoxia in the prenatal and postnatal preoperative brain may be associated with neurological damage and impaired long-term cognitive function, but the exact mechanisms are unknown. In this study, we find that delayed neuronal migration and impaired synaptic development are attributed to altered Atoh1 under chronic hypoxia. This is due to the fact that excessive Atoh1 facilitates expression of Kif21b, which causes excess in free-state α-tubulin, leading to disrupted microtubule dynamic stability. Furthermore, the delay in neonatal brain maturation induces cognitive disabilities in adult mice. Then, by down-regulating Atoh1 we alleviate the impairment of cell migration and synaptic development, improving the cognitive behavior of mice to some extent. Taken together, our work unveil that Atoh1 may be one of the targets to ameliorate hypoxia-induced neurodevelopmental disabilities and cognitive impairment in CHD.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Disfunção Cognitiva , Neurônios , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Camundongos , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Neurônios/metabolismo , Hipóxia/metabolismo , Feminino , Neurogênese , Animais Recém-Nascidos , Camundongos Endogâmicos C57BL , Masculino , Movimento Celular
9.
Front Endocrinol (Lausanne) ; 15: 1402905, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39268230

RESUMO

Background: Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and is closely associated with chronic low-grade inflammation and insulin resistance. To clarify the contribution of prepubertal weight gain to the development of insulin resistance in PCOS, we investigated the effects of early postnatal overfeeding on inflammatory and energy-sensing pathways as well as on markers of insulin signaling in the liver of the PCOS rat model. Methods: Obesity induced by overfeeding was achieved by reducing litter size, while the PCOS-like condition was developed by treatment with 5α-dihydrotestosterone (DHT). Western blot and qPCR were used to analyze the expression of pro-inflammatory transcription factors and cytokines, as well as markers of the energy sensing and insulin signaling pathways. Results: The results showed that hepatic insulin sensitivity was impaired only in DHT-treated rats raised in small litters, as evidenced by increased phosphorylation of IRS1 on Ser307 and decreased expression of total IRS1. Postnatal overfeeding stimulated JNK1 activation independent of hyperandrogenemia; nevertheless, the synergistic effect of both factors triggered NLRP3 activation and increased IL1ß expression in the small litter DHT-treated group. This pro-inflammatory state was accompanied by decreased activatory phosphorylation of AMPK and reduced levels of its protein targets. Conclusions: Overfeeding in the early postnatal period leads to a decrease in hepatic insulin sensitivity in the rat model of PCOS, which is associated with decreased activation of AMPK and stimulation of the hepatic NLRP3-IL1ß signaling pathway. Accordingly, the inhibition of NLRP3 activation could provide a basis for the development of new therapeutic strategies for the treatment of insulin resistance in women with PCOS.


Assuntos
Di-Hidrotestosterona , Modelos Animais de Doenças , Inflamação , Resistência à Insulina , Fígado , Hipernutrição , Síndrome do Ovário Policístico , Animais , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , Feminino , Ratos , Di-Hidrotestosterona/farmacologia , Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Inflamação/metabolismo , Inflamação/patologia , Hipernutrição/metabolismo , Hipernutrição/complicações , Ratos Wistar , Obesidade/metabolismo , Animais Recém-Nascidos , Transdução de Sinais/efeitos dos fármacos , Proteínas Substratos do Receptor de Insulina/metabolismo
10.
Nat Commun ; 15(1): 7754, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39237588

RESUMO

Cytomegalovirus (CMV) infection poses risks to newborns, necessitating effective therapies. Given that the damage includes both viral infection of brain cells and immune system-related damage, here we investigate the involvement of cellular prion protein (PrP), which plays vital roles in neuroprotection and immune regulation. Using a murine model, we show the role of PrP in tempering neonatal T cell immunity during CMV infection. PrP-null mice exhibit enhanced viral control through elevated virus-specific CD8 T cell responses, leading to reduced viral titers and pathology. We further unravel the molecular mechanisms by showing CMV-induced upregulation followed by release of PrP via the metalloproteinase ADAM10, impairing CD8 T cell response specifically in neonates. Additionally, we confirm PrP downregulation in human CMV (HCMV)-infected fibroblasts, underscoring the broader relevance of our observations beyond the murine model. Furthermore, our study highlights how PrP, under the stress of viral pathogenesis, reveals its impact on neonatal immune modulation.


Assuntos
Animais Recém-Nascidos , Linfócitos T CD8-Positivos , Infecções por Citomegalovirus , Citomegalovirus , Camundongos Knockout , Animais , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Citomegalovirus/imunologia , Humanos , Camundongos , Linfócitos T CD8-Positivos/imunologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/virologia , Proteínas Priônicas/metabolismo , Proteínas Priônicas/genética , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Proteína ADAM10/metabolismo , Proteína ADAM10/genética
11.
Brain Behav ; 14(9): e70009, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39236116

RESUMO

BACKGROUND: Preterm birth is associated with brain injury and long-term behavioral abnormalities, for which there are limited prevention options. When born preterm, infants prematurely lose placental neurosteroid (allopregnanolone) support. This increases the risk of excitotoxic damage to the brain, which increases the risk of injury, causing long-term deficits in behavior, myelination, and alterations to neurotransmitter pathways. We propose that postnatal restoration of neurosteroid action through zuranolone therapy will reduce neurological impairments following preterm birth. METHODS: Guinea pig dams underwent survival cesarean section surgery to deliver pups prematurely (GA64) or at term (GA69). Between birth and term equivalence age, preterm pups received vehicle (15% ß-cyclodextrin) or the allopregnanolone analogue zuranolone (1 mg/kg/day). Behavioral analysis was performed at postnatal day (PND) 7 and 40, before tissue collection at PND 42. Immunostaining for myelin basic protein (MBP), as well as real-time polymerase chain reaction to characterize oligodendrocyte lineage and neurotransmitter pathways, was performed in frontal cortex tissues. RESULTS: Zuranolone treatment prevented the hyperactive phenotype in preterm-born offspring, most markedly in males. Additionally, preterm-related reductions in MBP were ameliorated. Several preterm-related alterations in mRNA expression of dopaminergic, glutamatergic, and GABAergic pathways were also restored back to that of a term control level. CONCLUSION: This is the first study to assess zuranolone treatment as a neuroprotective therapy following preterm birth. Zuranolone treatment improved behavioral outcomes and structural changes in the preterm offspring, which continued long term until at least a late childhood timepoint. Clinical studies are warranted for further exploring the neuroprotective possibilities of this treatment following preterm birth.


Assuntos
Lobo Frontal , Pregnanolona , Nascimento Prematuro , Animais , Pregnanolona/farmacologia , Feminino , Cobaias , Masculino , Nascimento Prematuro/prevenção & controle , Nascimento Prematuro/tratamento farmacológico , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Animais Recém-Nascidos , Gravidez , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/administração & dosagem , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Proteína Básica da Mielina/metabolismo
12.
Respir Res ; 25(1): 328, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39223619

RESUMO

BACKGROUND: The interplay between intrauterine and early postnatal environments has been associated with an increased risk of cardiovascular diseases in adulthood, including pulmonary arterial hypertension (PAH). While emerging evidence highlights the crucial role of mitochondrial pathology in PAH, the specific mechanisms driving fetal-originated PAH remain elusive. METHODS AND RESULTS: To elucidate the role of mitochondrial dynamics in the pathogenesis of fetal-originated PAH, we established a rat model of postnatal catch-up growth following intrauterine growth restriction (IUGR) to induce pulmonary arterial hypertension (PAH). RNA-seq analysis of pulmonary artery samples from the rats revealed dysregulated mitochondrial metabolic genes and pathways associated with increased pulmonary arterial pressure and pulmonary arterial remodeling in the RC group (postnatal catch-up growth following IUGR). In vitro experiments using pulmonary arterial smooth muscle cells (PASMCs) from the RC group demonstrated elevated proliferation, migration, and impaired mitochondrial functions. Notably, reduced expression of Mitofusion 2 (Mfn2), a mitochondrial outer membrane protein involved in mitochondrial fusion, was observed in the RC group. Reconstitution of Mfn2 resulted in enhanced mitochondrial fusion and improved mitochondrial functions in PASMCs of RC group, effectively reversing the Warburg effect. Importantly, Mfn2 reconstitution alleviated the PAH phenotype in the RC group rats. CONCLUSIONS: Imbalanced mitochondrial dynamics, characterized by reduced Mfn2 expression, plays a critical role in the development of fetal-originated PAH following postnatal catch-up growth after IUGR. Mfn2 emerges as a promising therapeutic strategy for managing IUGR-catch-up growth induced PAH.


Assuntos
Retardo do Crescimento Fetal , GTP Fosfo-Hidrolases , Ratos Sprague-Dawley , Animais , Retardo do Crescimento Fetal/metabolismo , GTP Fosfo-Hidrolases/metabolismo , GTP Fosfo-Hidrolases/genética , Ratos , Feminino , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/patologia , Hipertensão Arterial Pulmonar/fisiopatologia , Dinâmica Mitocondrial/fisiologia , Masculino , Células Cultivadas , Gravidez , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Modelos Animais de Doenças , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Animais Recém-Nascidos , Proteínas Mitocondriais
13.
Prev Vet Med ; 232: 106316, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39180948

RESUMO

Piglet pre-weaning mortality (PWM) is a significant issue in the U.S. swine industry, causing economic losses and raising sustainability and animal welfare concerns. This study conducted a multivariable analysis to identify factors associated with PWM in a Midwestern U.S. swine production system. Weekly data from 47 sow farms (7207 weaning weeks) were captured from January 2020 to December 2022. Initially, 29 variables regarding farm infrastructure, productivity parameters, health status, and interventions were selected for univariate analysis to assess their association with PWM. The initial multivariable analysis included the variables with P < 0.20 in the univariate analyses. A backward stepwise model selection was conducted by excluding variables with P > 0.05, and the final multivariable model consisted of 19 significant risk factors and 6 interaction terms. The overall average PWM for the study population was 14.02 %. Yearly variations in PWM were observed, with the highest recorded in 2020 (16.61 %) and the lowest in 2021 (15.78 %). Cohorts with a pond water source, lower farrowing rate (71.9 %), higher farrowing parity (5.1), shorter gestation length (116.2 days), and using oxytocin during farrowing had increased PWM. The higher productivity parameters such as mummies rate, stillborn rate, and average total born, the higher the PWM was. Additionally, health status and intervention-related factors were associated with PWM, where higher PWM rates were observed in herds facing porcine reproductive and respiratory syndrome virus (PRRSV) outbreaks, porcine epidemic diarrhea virus (PEDV) positive, the weeks before and during feed medication, and weeks without using Rotavirus vaccine or Rotavirus feedback. Altogether, these results corroborate that PWM is a multifactorial problem, and a better understanding of the risk factors is essential in developing strategies to improve survival rates. Therefore, this study identified the major risk factors associated with PWM for groups of pigs raised under field conditions, and the results underscore the significance of data analysis in comprehending the unique challenges and opportunities inherent to each system.


Assuntos
Criação de Animais Domésticos , Doenças dos Suínos , Desmame , Animais , Fatores de Risco , Suínos , Criação de Animais Domésticos/métodos , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/virologia , Doenças dos Suínos/mortalidade , Feminino , Meio-Oeste dos Estados Unidos/epidemiologia , Sus scrofa , Animais Recém-Nascidos , Mortalidade
14.
Exp Neurol ; 381: 114929, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39168170

RESUMO

Neonatal hypoxia-ischemia (HI) is one of the main causes of mortality and long-term disabilities in newborns, and the only clinical approach to treat this condition is therapeutic hypothermia, which shows some limitations. Thus, putative neuroprotective agents have been tested in animal models of HI. Lactate is a preferential metabolic substrate of the neonatal brain and has already been shown to produce beneficial neuroprotective outcomes in neonatal animals exposed to HI. Here, we administered lactate as a treatment in neonatal rats previously exposed to HI and evaluated the impact of this treatment in adulthood. Seven-day-old (P7) male and female Wistar rats underwent permanent common right carotid occlusion combined with an exposition to a hypoxic atmosphere (8% oxygen) for 60 min. Animals were assigned to one of four experimental groups: HI, HI+LAC, SHAM, SHAM+LAC. Lactate was administered intraperitoneally 30 min and 2 h after hypoxia in HI+LAC and SHAM+LAC groups, whereas HI and SHAM groups received vehicle. Animals were tested in the behavioral tasks of negative geotaxis and righting reflex (P8), cylinder test (P24), and the modified neurological severity score was calculated (P25). Open field (OF), and novel object recognition (NOR) were evaluated in adulthood. Animals were killed at P60, and the brains were harvested and processed to evaluate the volume of brain injury. Our results showed that lactate administration reduced the volume of brain lesion and improved sensorimotor and cognitive behaviors in neonatal, juvenile, and adult life in HI animals from both sexes. Thus, lactate administration might be considered as a potential neuroprotective strategy for the treatment of neonatal HI, which is a prevalent disorder affecting newborns.


Assuntos
Animais Recém-Nascidos , Hipóxia-Isquemia Encefálica , Ácido Láctico , Fármacos Neuroprotetores , Ratos Wistar , Animais , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/metabolismo , Ratos , Feminino , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ácido Láctico/metabolismo , Modelos Animais de Doenças , Reconhecimento Psicológico/efeitos dos fármacos , Comportamento Exploratório/efeitos dos fármacos
15.
Neurotoxicol Teratol ; 105: 107385, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39182528

RESUMO

INTRODUCTION: Opioid use during pregnancy and subsequent neonatal opioid withdrawal syndrome (NOWS) have been associated with poor developmental outcomes including cognitive functioning. Less is known about the underlying molecular effects of prenatal opioid exposure and subsequent withdrawal; however, given the recent increase in NOWS cases, there is a pressing need to better understand these effects, which may partially explain cognitive deficits that have been observed in both preclinical NOWS models and patients with NOWS. This study evaluated the effects of prenatal heroin exposure and subsequent precipitated withdrawal symptoms on microglial reactivity in the nucleus accumbens (NAc), dorsal hippocampus (HC), and ventral tegmental area (VTA) in rat neonates, as well as cognitive functioning at three developmental time points using the Morris Water Maze (MWM) task. METHODS: Heroin or saline (2 mg/kg) was randomly assigned and administered to six pregnant Sprague Dawley rat dams via osmotic minipump. A total of 63 rat neonates underwent naloxone-precipitated (5 mg/kg, subcutaneous injection) withdrawal testing at postnatal day 10 (PN10). Following withdrawal testing, neonates were randomly assigned to undergo perfusion and subsequent immunohistochemistry experiments to fluoresce Iba-1 for microglia detection, or to undergo the MWM task at three separate developmental time points (PN21-23; PN37; PN60) for cognitive testing. RESULTS: Results suggest that in-utero heroin exposure led to an increase in ultrasonic vocalizations during naloxone-precipitated withdrawal; a sensitive index of withdrawal in rat neonates. Additional results suggest increased microglial reactivity in the HC and VTA, but not the NAc, as well as reduced performance during the MWM in the group exposed to heroin in-utero. DISCUSSION: Together, these data suggest that in-utero opioid exposure is associated with microglial reactivity in brain regions associated with learning and memory, and may be associated with later cognitive deficits. Further research is needed to characterize these findings, which may inform future therapeutic strategies for this vulnerable population.


Assuntos
Cognição , Heroína , Microglia , Efeitos Tardios da Exposição Pré-Natal , Ratos Sprague-Dawley , Animais , Heroína/toxicidade , Heroína/efeitos adversos , Microglia/efeitos dos fármacos , Gravidez , Feminino , Ratos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Cognição/efeitos dos fármacos , Síndrome de Abstinência a Substâncias , Masculino , Animais Recém-Nascidos , Hipocampo/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
16.
Horm Behav ; 165: 105630, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39186858

RESUMO

Maternal behavior experienced in early life provides essential scaffolding to infant psychobiology with life-long effects on neurobiological and behavioral outcomes. However, infants are not passive recipients of caregiving. Evidence in rodents suggests that pups actively contribute to dam-pup interactions by soliciting maternal care with auditory, tactile, and hormonal cues. The limited bedding and nesting material (LBN) rearing manipulation induces changes in maternal care that have been attributed to maternal stress caused by the low-resource environment. The goal of the current study was to determine whether LBN also alters pup cues for maternal behavior, with implications for the mechanism of LBN-induced effects. Rat dams and pups were randomly assigned to LBN or Control rearing conditions on postnatal day (P) 0-6 and pups were fostered to the same or different condition on P6-13. LBN increased pup-directed maternal behaviors measured through 24 h monitoring using machine learning based automated analysis. LBN altered several pup cues known to affect maternal behavior including reducing pup core body temperature, reducing body weight, and altering pup vocalizations on P6 and P12. P6-13 LBN-exposed pups had elevated serum testosterone, which positively correlated with maternal licking and grooming. LBN reduced pup movement between nest attendance onset and the start of nursing, which was negatively related to dam nursing latency and contributed to longer nursing latency in LBN dams. P0-6 pup exposure to LBN also led to longer nest attendance bouts and shorter licking and grooming bouts on P7 and P9, suggesting lasting effects of LBN on pups. These data demonstrate that LBN changes pup behavioral and hormonal signals consistent with eliciting more maternal care, contributing to augmented pup-directed behaviors. This bidirectional interplay may be a critical mechanism involved in the lasting effects of early life environments.


Assuntos
Animais Recém-Nascidos , Sinais (Psicologia) , Comportamento Materno , Animais , Comportamento Materno/fisiologia , Feminino , Ratos , Masculino , Comportamento Animal/fisiologia , Testosterona/sangue , Ratos Long-Evans , Meio Ambiente , Ratos Sprague-Dawley , Vocalização Animal/fisiologia
17.
Histochem Cell Biol ; 162(5): 385-402, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39153131

RESUMO

Somatostatin (SST) is a peptide expressed in the peripheral and central nervous systems, as well as in endocrine and immune cells. The aim of the current study is to determine the percentage of SST immunoreactive (IR) neurons and their colocalization with choline acetyltransferase (ChAT), neuronal nitric oxide synthase (nNOS), neuropeptide Y (NPY), and glial fibrillary acidic protein (GFAP) in the myenteric plexus (MP) and submucous plexus (SP) of the small intestine (SI) and large intestine (LI) of rats across different age groups from newborn to senescence using immunohistochemistry. In the MP of the SI and LI, the percentage of SST-IR neurons significantly increased during early postnatal development from 12 ± 2.4 (SI) and 13 ± 3.0 (LI) in newborn rats to 23 ± 1.5 (SI) and 18 ± 1.6 (LI) in 20-day-old animals, remaining stable until 60 days of age. The proportion of SST-IR cells then decreased in aged 2-year-old animals to 14 ± 2.0 (SI) and 10 ± 2.6 (LI). In the SP, the percentage of SST-IR neurons significantly rose from 22 ± 3.2 (SI) and 23 ± 1.7 (LI) in newborn rats to 42 ± 4.0 in 20-day-old animals (SI) and 32 ± 4.9 in 30-day-old animals (LI), before declining in aged 2-year-old animals to 21 ± 2.6 (SI) and 28 ± 7.4 (LI). Between birth and 60 days of age, 97-98% of SST-IR neurons in the MP and SP colocalized with ChAT in both plexuses of the SI and LI. The percentage of SST/ChAT neurons decreased in old rats to 85 ± 5.0 (SI) and 90 ± 3.8 (LI) in the MP and 89 ± 3.2 (SI) and 89 ± 1.6 (LI) in the SP. Conversely, in young rats, only a few SST-IR neurons colocalized with nNOS, but this percentage significantly increased in 2-year-old rats. The percentage of SST/NPY-IR neurons exhibited considerable variation throughout postnatal development, with no significant differences across different age groups in both the MP and SP of both intestines. No colocalization of SST with GFAP was observed in any of the studied animals. In conclusion, the expression of SST in enteric neurons increases in young rats and decreases in senescence, accompanied by changes in SST colocalization with ChAT and nNOS.


Assuntos
Neurônios , Somatostatina , Animais , Ratos , Somatostatina/metabolismo , Somatostatina/análise , Neurônios/metabolismo , Neurônios/citologia , Masculino , Imuno-Histoquímica , Ratos Sprague-Dawley , Animais Recém-Nascidos
18.
Res Vet Sci ; 178: 105379, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39173242

RESUMO

In premature births, deficiency and/or inactivation of surfactant and incomplete development of lung occur, leading to pulmonary complications and greater need for ventilatory interventions. Prenatal corticosteroid therapy is used to improve neonatal lung function and, thus, may reduce mortality and lower incidence and severity of lung injury. Therefore, this study aimed to assess the need for ventilatory support in preterm lambs subjected or not to prenatal betamethasone treatment, and to evaluate the effectiveness on neonatal survival. Lambing was induced and 13 premature lambs were assigned to Corticosteroid Group (n = 8; lambs from ewes subjected previously to 0.5 mg/kg betamethasone, IM, at 133 days of pregnancy) and Control Group (n = 5; non-treated lambs). Lambs were evaluated for vitality, neurologic reflexes, vital functions and birth weight. Three ventilatory modalities were preconized for critical lambs, according to specific criteria: mask oxygen therapy, self-inflating bag with tracheal tube and mechanical ventilation. Non-treated lambs had lower vitality score, muscle tonus and respiratory rate compared to Corticosteroid Group. Ventilatory support was needed for 3 Control lambs and only 1 Corticosteroid neonate. Corticosteroid lamb required significant less time-frame between birth and onset of ventilatory assistance and remained under ventilation for a shorter time. Percentage of ventilated non-treated lambs correlated negatively with birth weight, muscle tone, heart and respiratory rate. In conclusion, antenatal betamethasone treatment reduces the need for ventilatory assistance in premature lambs. Additionally, mortality is low when a protocol for inducing pulmonary maturity (maternal corticosteroid therapy) and/or ventilatory interventions are employed, ensuring the survival of premature lambs.


Assuntos
Animais Recém-Nascidos , Betametasona , Respiração Artificial , Animais , Betametasona/uso terapêutico , Betametasona/administração & dosagem , Gravidez , Feminino , Respiração Artificial/veterinária , Ovinos , Nascimento Prematuro/veterinária , Nascimento Prematuro/prevenção & controle , Corticosteroides/uso terapêutico , Corticosteroides/administração & dosagem , Doenças dos Ovinos/prevenção & controle , Doenças dos Ovinos/tratamento farmacológico , Carneiro Doméstico
19.
J Neuroinflammation ; 21(1): 199, 2024 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-39128994

RESUMO

Infection during the perinatal period can adversely affect brain development, predispose infants to ischemic stroke and have lifelong consequences. We previously demonstrated that diet enriched in n-3 polyunsaturated fatty acids (n-3 PUFA) transforms brain lipid composition in the offspring and protects the neonatal brain from stroke, in part by blunting injurious immune responses. Critical to the interface between the brain and systemic circulation is the vasculature, endothelial cells in particular, that support brain homeostasis and provide a barrier to systemic infection. Here, we examined whether maternal PUFA-enriched diets exert reprograming of endothelial cell signalling in postnatal day 9 mice after modeling aspects of infection using LPS. Transcriptome analysis was performed on microvessels isolated from brains of pups from dams maintained on 3 different maternal diets from gestation day 1: standard, n-3 enriched or n-6 enriched diets. Depending on the diet, in endothelial cells LPS produced distinct regulation of pathways related to immune response, cell cycle, extracellular matrix, and angiogenesis. N-3 PUFA diet enabled higher immune reactivity in brain vasculature, while preventing imbalance of cell cycle regulation and extracellular matrix cascades that accompanied inflammatory response in standard diet. Cytokine analysis revealed a blunted LPS response in blood and brain of offspring from dams on n-3 enriched diet. Analysis of cerebral vasculature in offspring in vivo revealed no differences in vessel density. However, vessel complexity was decreased in response to LPS at 72 h in standard and n-6 diets. Thus, LPS modulates specific transcriptomic changes in brain vessels of offspring rather than major structural vessel characteristics during early life. N-3 PUFA-enriched maternal diet in part prevents an imbalance in homeostatic processes, alters inflammation and ultimately mitigates changes to the complexity of surface vessel networks that result from infection. Importantly, maternal diet may presage offspring neurovascular outcomes later in life.


Assuntos
Animais Recém-Nascidos , Ácidos Graxos Ômega-3 , Transcriptoma , Animais , Camundongos , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Gravidez , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Inflamação/metabolismo , Inflamação/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Endotoxinas/toxicidade
20.
Neurobiol Dis ; 200: 106629, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39111704

RESUMO

Hypoxic-ischemic encephalopathy (HIE) in neonates causes mortality and neurologic morbidity, including poor cognition with a complex neuropathology. Injury to the cholinergic basal forebrain and its rich innervation of cerebral cortex may also drive cognitive pathology. It is uncertain whether genes associated with adult cognition-related neurodegeneration worsen outcomes after neonatal HIE. We hypothesized that neocortical damage caused by neonatal HI in mice is ushered by persistent cholinergic innervation and interneuron (IN) pathology that correlates with cognitive outcome and is exacerbated by genes linked to Alzheimer's disease. We subjected non-transgenic (nTg) C57Bl6 mice and mice transgenically (Tg) expressing human mutant amyloid precursor protein (APP-Swedish variant) and mutant presenilin (PS1-ΔE9) to the Rice-Vannucci HI model on postnatal day 10 (P10). nTg and Tg mice with sham procedure were controls. Visual discrimination (VD) was tested for cognition. Cortical and hippocampal cholinergic axonal and IN pathology and Aß plaques, identified by immunohistochemistry for choline acetyltransferase (ChAT) and 6E10 antibody respectively, were counted at P210. Simple ChAT+ axonal swellings were present in all sham and HI groups; Tg mice had more than their nTg counterparts, but HI did not affect the number of axonal swellings in APP/PS1 Tg mice. In contrast, complex ChAT+ neuritic clusters (NC) occurred only in Tg mice; HI increased that burden. The abundance of ChAT+ clusters in specific regions correlated with decreased VD. The frequency of attritional ChAT+ INs in the entorhinal cortex (EC) was increased in Tg shams relative to their nTg counterparts, but HI obviated this difference. Cholinergic IN pathology in EC correlated with NC number. The Aß deposition in APP/PS1 Tg mice was not exacerbated by HI, nor did it correlate with other metrics. Adult APP/PS1 Tg mice have significant cortical cholinergic axon and EC ChAT+ IN pathologies; some pathology was exacerbated by neonatal HI and correlated with VD. Mechanisms of neonatal HI induced cognitive deficits and cortical neuropathology may be modulated by genetic risk, perhaps accounting for some of the variability in outcomes.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Animais Recém-Nascidos , Neurônios Colinérgicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex , Animais , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Camundongos , Neocórtex/metabolismo , Neocórtex/patologia , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Neurônios Colinérgicos/patologia , Neurônios Colinérgicos/metabolismo , Presenilina-1/genética , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/genética , Lesões Encefálicas/patologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/genética , Colina O-Acetiltransferase/metabolismo , Colina O-Acetiltransferase/genética , Humanos , Masculino , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA