Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117.572
Filter
1.
Chem Biol Drug Des ; 103(4): e14520, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570710

ABSTRACT

Quercetin, a bioactive natural compound renowned for its potent anti-inflammatory, antioxidant, and antiviral properties, has exhibited therapeutic potential in various diseases. Given that bronchopulmonary dysplasia (BPD) development is closely linked to inflammation and oxidative stress, and quercetin, a robust antioxidant known to activate NRF2 and influence the ferroptosis pathway, offers promise for a wide range of age groups. Nonetheless, the specific role of quercetin in BPD remains largely unexplored. This study aims to uncover the target role of quercetin in BPD through a combination of network pharmacology, molecular docking, computer analyses, and experimental evaluations.


Subject(s)
Bronchopulmonary Dysplasia , Ferroptosis , Hyperoxia , Animals , Infant, Newborn , Humans , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/metabolism , Hyperoxia/drug therapy , Hyperoxia/metabolism , Quercetin/pharmacology , Quercetin/therapeutic use , Molecular Docking Simulation , Cyclooxygenase 2 , Animals, Newborn , Antioxidants , Network Pharmacology
2.
J Neurosci Res ; 102(4): e25329, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38597144

ABSTRACT

There is a need for new treatments to reduce brain injuries derived from neonatal hypoxia/ischemia. The only viable option used in the clinic today in infants born at term is therapeutic hypothermia, which has a limited efficacy. Treatments with exogenous RNase have shown great promise in a range of different adult animal models including stroke, ischemia/reperfusion injury, or experimental heart transplantation, often by conferring vascular protective and anti-inflammatory effects. However, any neuroprotective function of RNase treatment in the neonate remains unknown. Using a well-established model of neonatal hypoxic/ischemic brain injury, we evaluated the influence of RNase treatment on RNase activity, gray and white matter tissue loss, blood-brain barrier function, as well as levels and expression of inflammatory cytokines in the brain up to 6 h after the injury using multiplex immunoassay and RT-PCR. Intraperitoneal treatment with RNase increased RNase activity in both plasma and cerebropinal fluids. The RNase treatment resulted in a reduction of brain tissue loss but did not affect the blood-brain barrier function and had only a minor modulatory effect on the inflammatory response. It is concluded that RNase treatment may be promising as a neuroprotective regimen, whereas the mechanistic effects of this treatment appear to be different in the neonate compared to the adult and need further investigation.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Infant, Newborn , Infant , Humans , Animals, Newborn , Ribonucleases/metabolism , Ribonucleases/pharmacology , Brain Injuries/drug therapy , Brain/metabolism , Ischemia/drug therapy , Neuroprotective Agents/pharmacology , Disease Models, Animal
3.
Eur J Pharmacol ; 971: 176539, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38565342

ABSTRACT

Hypoxic-ischemic brain damage (HIBD) is a cerebral injury resulting from the combination of ischemia and hypoxia in neonatal brain tissue. Presently, there exists no efficacious remedy for HIBD. A mounting body of evidence indicates that dynamic metabolites formed during metabolic procedures assume a vital role in neuronal maturation and recuperation. However, it remains unclear whether any endogenous metabolites are involved in the pathogenesis of HIBD. Here, an untargeted metabolomics analysis was conducted by gas chromatography-mass spectrometry and liquid chromatography-mass spectrometry (GC/LC-MS) in OGD/R (oxygen-glucose deprivation/reoxygenation)-induced HT-22 cells. We observed that ferroptosis signaling plays an essential role in HI-induced neuronal injury. Interestingly, we also found that the differentially expressed metabolite, 2-phosphoglyceric acid, significantly improved the neuronal cell survival of OGD/R HT-22 cells by inhibiting ferroptosis. Moreover, 2-phosphoglyceric acid effectively rescued the cell activity of HT-22 cells treated with the ferroptosis inducer RSL-3. Furthermore, 2-phosphoglyceric acid alleviated cerebral infarction and reduced HIBD-induced neuronal cell loss of the central nervous system in neonatal rats by regulating GPX4 expression. Taken together, we found that 2-phosphoglyceric acid, which was downregulated in HT-22 cells induced by OGD/R, exerted neuronal protective effects on OGD/R-treated HT-22 cells and HIBD-induced neonatal rats by inhibiting hypoxic-ischemic-induced ferroptosis through the regulation of the GPX4/ACSL4 axis.


Subject(s)
Hypoxia-Ischemia, Brain , Rats , Animals , Animals, Newborn , Rats, Sprague-Dawley , Hypoxia-Ischemia, Brain/metabolism , Hypoxia/metabolism , Brain/metabolism
4.
Sci Rep ; 14(1): 7924, 2024 04 04.
Article in English | MEDLINE | ID: mdl-38575644

ABSTRACT

Neonatal hypoxic-ischemic brain injury (HIBI) results in part from excess reactive oxygen species and iron-dependent lipid peroxidation (i.e. ferroptosis). The vitamin D precursor 7-dehydrocholesterol (7-DHC) may inhibit iron-dependent lipid peroxidation. Primary neurons underwent oxygen and glucose deprivation (OGD) injury and treatment with 7-DHC-elevating medications such as cariprazine (CAR) or vehicle. Postnatal day 9 mice underwent sham surgery or carotid artery ligation and hypoxia and received intraperitoneal CAR. In neurons, CAR administration resulted in significantly increased cell survival compared to vehicle controls, whether administered 48 h prior to or 30 min after OGD, and was associated with increased 7-DHC. In the mouse model, malondialdehyde and infarct area significantly increased after HIBI in the vehicle group, which were attenuated by post-treatment with CAR and were negatively correlated with tissue 7-DHC concentrations. Elevating 7-DHC concentrations with CAR was associated with improved cellular and tissue viability after hypoxic-ischemic injury, suggesting a novel therapeutic avenue.


Subject(s)
Dehydrocholesterols , Ferroptosis , Hypoxia-Ischemia, Brain , Animals , Mice , Animals, Newborn , Brain , Hypoxia/complications , Oxygen/therapeutic use , Ischemia/complications , Iron/therapeutic use
5.
Cir Pediatr ; 37(2): 61-66, 2024 Apr 01.
Article in English, Spanish | MEDLINE | ID: mdl-38623798

ABSTRACT

INTRODUCTION: Necrotizing enterocolitis (NEC) is a life-threatening condition that afflicts neonates. Breastfeeding has demonstrated to play a protective role against it. By administering lipopolysaccharides (LPS) orally in newborn rats (NBR), we have developed an experimental model to induce NEC-like gut damage. Our aim was to assess the macroscopic and microscopic appearance of the gut, to evaluate the presence of NEC and study the role of breast milk (BM). MATERIALS AND METHODS: NBR were divided into 3 groups: Group A (control, n= 10) remained with the mother, group B (LPS, n= 25) was isolated after birth, gavage-fed with special rat formula and oral LPS, then submitted to stress (hypoxia after gavage) and group c (BM, n= 12) was breastfed once after birth, then isolated, and submitted to stress like group B. On day 4, NBR were sacrificed, and intestine was harvested and assessed. RESULTS: In the control group NEC was not present either macroscopically or histologically. Both groups submitted to stress (B and C) presented a global incidence of NEC of 73%. Most of group B developed histologic signs of NEC (85%) and group C showed a statistically lower incidence of NEC (50%, p= 0.04), playing the BM a protective role against NEC (OR= 0.19; 95% CI: 0.40-0.904). CONCLUSION: Our model showed a significant incidence of NEC in NBR (73%) with the same protective role of BM as in newborn humans, achieving a reliable and reproducible experimental NEC model. This will allow us to investigate new potential therapeutic targets for a devastating disease that currently lacks treatment.


INTRODUCCION: La enterocolitis necrotizante (ECN) es una enfermedad potencialmente mortal que afecta a los neonatos, y frente a la que la leche materna ha demostrado tener un papel protector. Administrando lipopolisacáridos (LPS) por vía oral en ratas recién nacidas (RRN), hemos desarrollado un modelo experimental para inducir un daño intestinal similar al que provoca la ECN con objeto de evaluar el aspecto macroscópico y microscópico del intestino, y de ese modo, analizar la presencia de ECN y estudiar el papel que desempeña la leche materna (LM). MATERIAL Y METODOS: Las RRN se dividieron en tres grupos: el grupo A (control, n= 10) permaneció con su madre; el grupo B (LPS, n= 25) fue aislado tras el nacimiento, alimentado por sonda con una fórmula especial para ratas y LPS oral, y sometido a estrés (hipoxia tras sonda); y el grupo C (LM, n= 12) fue alimentado con leche materna tras el nacimiento y posteriormente aislado y sometido a estrés al igual que el grupo B. El día 4 se sacrificó a las RRN y se recuperaron sus intestinos para su posterior evaluación. RESULTADOS: En el grupo de control, no se observó ECN ni macroscópica ni histológicamente, mientras que los dos grupos sometidos a estrés (B y C) presentaron una incidencia global de la ECN del 73%. La mayoría de los sujetos del grupo B desarrollaron signos histológicos de ECN (85%), y los del grupo C registraron una incidencia de la ECN estadísticamente menor (50%, p= 0,04), lo que significa que la LM desempeña una función protectora frente a la ECN (OR= 0,19; IC 95%: 0,40-0,904). CONCLUSION: Nuestro modelo reveló una incidencia significativa de la ECN en RRN (73%), desempeñando la LM la misma función protectora que en el caso de los humanos recién nacidos, lo que significa que este modelo experimental de ECN es fiable y reproducible. Gracias a dicho logro, podremos investigar nuevos y potenciales objetivos terapéuticos para una peligrosa enfermedad que, a día de hoy, carece de tratamiento.


Subject(s)
Enterocolitis, Necrotizing , Lipopolysaccharides , Female , Animals , Infant, Newborn , Rats , Humans , Animals, Newborn , Lipopolysaccharides/therapeutic use , Milk, Human , Enterocolitis, Necrotizing/epidemiology , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/prevention & control , Disease Models, Animal
6.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(4): 394-402, 2024 Apr 15.
Article in Chinese | MEDLINE | ID: mdl-38660904

ABSTRACT

OBJECTIVES: To compare the repair effects of different doses of human umbilical cord mesenchymal stem cells (hUC-MSCs) on white matter injury (WMI) in neonatal rats. METHODS: Two-day-old Sprague-Dawley neonatal rats were randomly divided into five groups: sham operation group, WMI group, and hUC-MSCs groups (low dose, medium dose, and high dose), with 24 rats in each group. Twenty-four hours after successful establishment of the neonatal rat white matter injury model, the WMI group was injected with sterile PBS via the lateral ventricle, while the hUC-MSCs groups received injections of hUC-MSCs at different doses. At 14 and 21 days post-modeling, hematoxylin and eosin staining was used to observe pathological changes in the tissues around the lateral ventricles. Real-time quantitative polymerase chain reaction was used to detect the quantitative expression of myelin basic protein (MBP) and glial fibrillary acidic protein (GFAP) mRNA in the brain tissue. Immunohistochemistry was employed to observe the expression levels of GFAP and neuron-specific nuclear protein (NeuN) in the tissues around the lateral ventricles. TUNEL staining was used to observe cell apoptosis in the tissues around the lateral ventricles. At 21 days post-modeling, the Morris water maze test was used to observe the spatial learning and memory capabilities of the neonatal rats. RESULTS: At 14 and 21 days post-modeling, numerous cells with nuclear shrinkage and rupture, as well as disordered arrangement of nerve fibers, were observed in the tissues around the lateral ventricles of the WMI group and the low dose group. Compared with the WMI group, the medium and high dose groups showed alleviated pathological changes; the arrangement of nerve fibers in the medium dose group was relatively more orderly compared with the high dose group. Compared with the WMI group, there was no significant difference in the expression levels of MBP and GFAP mRNA in the low dose group (P>0.05), while the expression levels of MBP mRNA increased and GFAP mRNA decreased in the medium and high dose groups. The expression level of MBP mRNA in the medium dose group was higher than that in the high dose group, and the expression level of GFAP mRNA in the medium dose group was lower than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the protein expression of GFAP and NeuN in the low dose group (P>0.05), while the expression of NeuN protein increased and GFAP protein decreased in the medium and high dose groups. The expression of NeuN protein in the medium dose group was higher than that in the high dose group, and the expression of GFAP protein in the medium dose group was lower than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the number of apoptotic cells in the low dose group (P>0.05), while the number of apoptotic cells in the medium and high dose groups was less than that in the WMI group, and the number of apoptotic cells in the medium dose group was less than that in the high dose group (P<0.05). Compared with the WMI group, there was no significant difference in the escape latency time in the low dose group (P>0.05); starting from the third day of the latency period, the escape latency time in the medium dose group was less than that in the WMI group (P<0.05). The medium and high dose groups crossed the platform more times than the WMI group (P<0.05). CONCLUSIONS: Low dose hUC-MSCs may yield unsatisfactory repair effects on WMI in neonatal rats, while medium and high doses of hUC-MSCs have significant repair effects, with the medium dose demonstrating superior efficacy.


Subject(s)
Animals, Newborn , Mesenchymal Stem Cell Transplantation , Rats, Sprague-Dawley , Umbilical Cord , White Matter , Animals , Rats , Humans , Umbilical Cord/cytology , White Matter/pathology , White Matter/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/analysis , Mesenchymal Stem Cells , Myelin Basic Protein/genetics , Myelin Basic Protein/analysis , Myelin Basic Protein/metabolism , Male , Apoptosis , Female , RNA, Messenger/analysis , RNA, Messenger/metabolism
7.
J Vis Exp ; (205)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38526075

ABSTRACT

Intrathecal injection is a commonly employed procedure in both pediatric and adult clinics, serving as an effective means to administer medications and treatments. By directly delivering medications and treatments into the cerebrospinal fluid of the central nervous system, this method achieves higher localized drug concentrations while reducing systemic side-effects compared to other routes such as intravenous, subcutaneous, or intramuscular injections. Its importance extends beyond clinical settings, as intrathecal injection plays a vital role in preclinical studies focused on treating neurogenetic disorders in rodents and other large animals, including non-human primates. However, despite its widespread application, intrathecal injection in young, particularly neonatal pups, poses significant technical challenges due to their small size and fragile nature. Successful and reliable administration of intrathecal injections in newborn mice requires meticulous attention to detail and careful consideration of various factors. Thus, there is a crucial need for a standardized protocol that not only provides instructions but also highlights key technical considerations and good laboratory practices to ensure procedural consistency, as well as the safety and welfare of the animals. To address this unmet need, we present a detailed and comprehensive protocol for performing intrathecal injections specifically in newborn pups on postnatal day 1 (P1). By following the step-by-step instructions, researchers can confidently perform intrathecal injections in neonatal pups, enabling the accurate delivery of drugs, antisense oligos, and viruses for gene replacement or genome editing-based treatments. Furthermore, the importance of adhering to good laboratory practices is emphasized to maintain the well-being of animals and ensure reliable experimental outcomes. This protocol aims to address the technical challenges associated with intrathecal injections in neonatal mice, ultimately facilitating advances in the field of neurogenetic research that aims to develop potential therapeutic interventions.


Subject(s)
Drug Delivery Systems , Gene Editing , Adult , Animals , Mice , Humans , Child , Animals, Newborn , Injections, Spinal/methods , Central Nervous System , Pharmaceutical Preparations
8.
Pediatr Surg Int ; 40(1): 80, 2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38493431

ABSTRACT

BACKGROUND AND AIM: Necrotizing Enterocolitis (NEC) is an inflammation-associated ischemic necrosis of the intestine. To investigate the effects of extra virgin olive oil (EVOO) on inflammation, oxidative stress, apoptosis, and histological changes in NEC-induced newborn rats. MATERIALS AND METHODS: 24 rats were randomly divided into three groups: control, NEC and NEC + EVOO. NEC induction was performed using hypoxia-hyperoxia, formula feeding, and cold stress. The NEC + EVOO group received 2 ml/kg EVOO with high phenolic content by gavage twice a day for 3 days. 3 cm of bowel including terminal ileum, cecum, and proximal colon was excised. RESULTS: Weight gain and clinical disease scores were significantly higher in the NEC + EVOO group than in the NEC group (p < 0.001). EVOO treatment caused significant decreases in IL1ß, IL6 levels (p = 0.016, p = 0.029 respectively) and EGF, MDA levels (p = 0.032, p = 0.013 respectively) compared to NEC group. Significant decreases were observed in IL6 gene expression in the NEC + EVOO group compared to the NEC group (p = 0.002). In the group NEC + EVOO, the number of Caspase-3 positive cells was found to be significantly reduced (p < 0.001) and histopathological examination revealed minimal changes and significantly lower histopathological scores (p < 0.001). CONCLUSION: Phenol-rich EVOO prevents intestinal damage caused by NEC by inhibiting inflammation, oxidative stress, apoptosis.


Subject(s)
Enterocolitis, Necrotizing , Interleukin-6 , Rats , Animals , Olive Oil/therapeutic use , Olive Oil/pharmacology , Interleukin-6/metabolism , Enterocolitis, Necrotizing/pathology , Oxidative Stress , Apoptosis , Inflammation , Phenols/pharmacology , Phenols/therapeutic use , Models, Theoretical , Animals, Newborn
9.
Aging (Albany NY) ; 16(5): 4670-4683, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38446592

ABSTRACT

Surgery and anesthesia are vital medical interventions, but concerns over their potential cognitive side effects, particularly with the use of inhalational anesthetics like sevoflurane, have surfaced. This study delves into the neuroprotective potential of Echinatin against sevoflurane-induced neurotoxicity and the underlying mechanisms. Echinatin, a natural compound, has exhibited anti-inflammatory, antioxidant, and anticancer properties. Sevoflurane, while a popular anesthetic, is associated with perioperative neurocognitive disorders (PND) and neurotoxicity. Our investigation began with cellular models, where Echinatin demonstrated a significant reduction in sevoflurane-induced apoptosis. Mechanistically, we identified ferroptosis, a novel form of programmed cell death characterized by iron accumulation and lipid peroxidation, as a key player in sevoflurane-induced neuronal injury. Echinatin notably suppressed ferroptosis in sevoflurane-exposed cells, suggesting a pivotal role in neuroprotection. Expanding our research to a murine model, we observed perturbations in iron homeostasis, inflammatory cytokines, and antioxidants due to sevoflurane exposure. Echinatin treatment effectively restored iron balance, mitigated inflammation, and preserved antioxidant levels in vivo. Behavioral assessments using the Morris water maze further confirmed Echinatin's neuroprotective potential, as it ameliorated sevoflurane-induced spatial learning and memory impairments. In conclusion, our study unveils Echinatin as a promising candidate for mitigating sevoflurane-induced neurotoxicity. Through the regulation of ferroptosis, iron homeostasis, and inflammation, Echinatin demonstrates significant neuroprotection both in vitro and in vivo. These findings illuminate the potential for Echinatin to enhance the safety of surgical procedures involving sevoflurane anesthesia, minimizing the risk of cognitive deficits and neurotoxicity.


Subject(s)
Chalcones , Ferroptosis , Methyl Ethers , Neurotoxicity Syndromes , Rats , Animals , Mice , Sevoflurane/toxicity , Methyl Ethers/pharmacology , Methyl Ethers/toxicity , Antioxidants/pharmacology , Animals, Newborn , Rats, Sprague-Dawley , Homeostasis , Inflammation/metabolism , Hippocampus/metabolism
10.
Gut Microbes ; 16(1): 2333808, 2024.
Article in English | MEDLINE | ID: mdl-38533575

ABSTRACT

Premature infants lack a normal intestinal microbial community and also at risk of perinatal hypoxic-ischemic (HI) brain injury, which is considered to be one of the major factors for motor, sensory, and cognitive deficits. We hypothesized that neonatal gut microbiota composition modulated the immune reaction and severity of neonatal H-I brain injury. Neonatal C57BL/6J mouse pups were exposed to H-I protocol consisting of permanent left carotid artery ligation, followed by 8% hypoxia for 60 min. Microbial manipulation groups included 1) antibiotic treatment, E18 (maternal) to P5; 2) antibiotic treatment E18 to P5 + E. coli gavage; 3) antibiotic treatment E18 to P5 + B. infantis gavage; and 4) saline to pups with dams getting fresh water. The extent of brain injury and recovery was measured on MRI. Edematous injury volume was significantly higher in E. coli group than that in B. infantis group and in fresh water group. Gene expression in brains of pro-inflammatory cytokines (IL1ß, IL6, IL2, TNF-α and toll-like receptors 2-6) were elevated to a greater extent in the E. coli group at P10, no injury, and at P13, 72 hours after H-I relative to sham control and B. infantis groups. Significant effects of microbiome and brain injury and interaction of these factors were found in abundance of major phyla. The neuroinflammatory response and brain injury after neonatal hypoxia-ischemia are affected by intestinal microbiota, providing opportunities for therapeutic intervention through targeting the early colonization and development of the gut microbiota.


Subject(s)
Brain Injuries , Gastrointestinal Microbiome , Hypoxia-Ischemia, Brain , Animals , Rats , Mice , Infant, Newborn , Pregnancy , Female , Humans , Animals, Newborn , Rats, Wistar , Escherichia coli , Mice, Inbred C57BL , Brain Injuries/metabolism , Ischemia/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Brain/metabolism , Hypoxia/metabolism , Anti-Bacterial Agents/pharmacology
11.
Nutrients ; 16(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38542762

ABSTRACT

The parenteral nutrition (PN) received by premature newborns is contaminated with peroxides that induce global DNA hypermethylation via oxidative stress. Exposure to peroxides could be an important factor in the induction of chronic diseases such as those observed in adults who were born preterm. As endogenous H2O2 is a major regulator of glucose-lipid metabolism, our hypothesis was that early exposure to PN induces permanent epigenetic changes in H2O2 metabolism. Three-day-old guinea pigs were fed orally (ON), PN or glutathione-enriched PN (PN+GSSG). GSSG promotes endogenous peroxide detoxification. After 4 days, half the animals were sacrificed, and the other half were fed ON until 16 weeks of age. The liver was harvested. DNA methylation and mRNA levels were determined for the SOD2, GPx1, GCLC, GSase, Nrf2 and Keap1 genes. PN induced GPx1 hypermethylation and decreased GPx1, GCLC and GSase mRNA. These findings were not observed in PN+GSSG. PN+GSSG induced Nrf2 hypomethylation and increased Nrf2 and SOD2 mRNA. These observations were independent of age. In conclusion, in neonatal guinea pigs, PN induces epigenetic changes, affecting the expression of H2O2 metabolism genes. These changes persist for at least 15 weeks after PN. This disruption may signify a permanent reduction in the capacity to detoxify peroxides.


Subject(s)
Hydrogen Peroxide , NF-E2-Related Factor 2 , Animals , Guinea Pigs , Hydrogen Peroxide/metabolism , Glutathione Disulfide/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Animals, Newborn , Parenteral Nutrition/adverse effects , Glutathione/metabolism , Peroxides/metabolism , Dietary Supplements , Epigenesis, Genetic , RNA, Messenger/genetics
12.
Sci Total Environ ; 925: 171790, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38508253

ABSTRACT

Fenvalerate (FEN), a type II pyrethroid pesticide, finds extensive application in agriculture, graziery and public spaces for pest control, resulting in severe environmental pollution. As an environmental endocrine disruptor with estrogen-like activity, exposure to FEN exhibited adverse effects on ovarian functions. Additionally, the presence of the metabolite of FEN in women's urine shows a positive association with the risk of primary ovarian insufficiency (POI). In mammals, the primordial follicle pool established during the early life serves as a reservoir for storing all available oocytes throughout the female reproductive life. The initial size of the primordial follicle pool and the rate of its depletion affect the occurrence of POI. Nevertheless, there is very limited research about the impact of FEN exposure on primordial folliculogenesis. In this study, pregnant mice were orally administrated with 0.2, 2.0 and 20.0 mg/kg FEN from 16.5 to 18.5 days post-coitus (dpc). Ovaries exposed to FEN exhibited the presence of large germ-cell cysts that persist on 1 days post-parturition (1 dpp), followed by a significant reduction in the total number of oocytes in pups on 5 dpp. Moreover, the levels of m6A-RNA and its associated proteins METTL3 and YTHDF2 were significantly increased in the ovaries exposed to FEN. The increased YTHDF2 promoted the assembly of the cytoplasmic processing bodies (P-body) in the oocytes, accompanied with altered expression of transcripts. Additionally, when YTHDF2 was knocked-down in fetal ovary cultures, the primordial folliculogenesis disrupted by FEN exposure was effectively restored. Further, the female offspring exposed to FEN displayed ovarian dysfunctions reminiscent of POI in early adulthood, characterized by decreases in ovarian coefficient and female hormone levels. Therefore, the present study revealed that exposure to FEN during late pregnancy disrupted primordial folliculogenesis by YTHDF2-mediated P-body assembly, causing enduring adverse effects on female fertility.


Subject(s)
Nitriles , Ovarian Reserve , Pyrethrins , Humans , Pregnancy , Animals , Female , Mice , Adult , Animals, Newborn , Processing Bodies , Oocytes/metabolism , Pyrethrins/toxicity , Pyrethrins/metabolism , Mammals/metabolism , Methyltransferases , RNA-Binding Proteins
13.
Anim Biotechnol ; 35(1): 2320726, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38436999

ABSTRACT

Colostrum is the initial secretion of the mammary glands following parturition, which offers main food, protection, and biological active substances for the new born. The most threatening episode of neonate's life is the initial two weeks after birth. This period is associated with high neonatal mortality and morbidity. These worthwhile losses lead to a poor prolificacy rate, low profitability, and ultimately poor performance in animal production. Hence, both diseases and mortality cause valuable losses in terms of production and economic losses. The survival of neonate is correlated with their immune status and passive immune transfer (PIT). Colostrum provides the primary source of nutrition and immunity (PIT) that protects neonates against infections. It must be given as soon as possible after birth since its immunoglobulins are absorbed within the first 16-27 hours after birth, ideally within 2-4 hours. As a result, immunoglobulin (PIT) is the most important component of distressing infectious immunity, and a passable concentration of immunoglobulin in the blood of newborn lambs is linked to their health and survival rate. In this review, we summarized the importance of colostrum in early life and its association with neonatal lamb's survival, profitability and productivity of sheep farming.


Subject(s)
Colostrum , Immunoglobulin G , Pregnancy , Female , Animals , Sheep , Animals, Newborn
14.
Stem Cell Res Ther ; 15(1): 80, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38486338

ABSTRACT

BACKGROUND: Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among preterm infants. Human induced pluripotent stem cells (hiPSCs) have shown promise in repairing injury in animal BPD models. Evidence suggests they exert their effects via paracrine mechanisms. We aim herein to assess the effectiveness of extracellular vesicles (EVs) derived from hiPSCs and their alveolar progenies (diPSCs) in attenuating hyperoxic injury in a preterm lung explant model. METHODS: Murine lung lobes were harvested on embryonic day 17.5 and maintained in air-liquid interface. Following exposure to 95% O2 for 24 h, media was supplemented with 5 × 106 particles/mL of EVs isolated from hiPSCs or diPSCs by size-exclusion chromatography. On day 3, explants were assessed using Hematoxylin-Eosin staining with mean linear intercept (MLI) measurements, immunohistochemistry, VEGFa and antioxidant gene expression. Statistical analysis was conducted using one-way ANOVA and Multiple Comparison Test. EV proteomic profiling was performed, and annotations focused on alveolarization and angiogenesis signaling pathways, as well as anti-inflammatory, anti-oxidant, and regenerative pathways. RESULTS: Exposure of fetal lung explants to hyperoxia induced airspace enlargement, increased MLI, upregulation of anti-oxidants Prdx5 and Nfe2l2 with decreased VEGFa expression. Treatment with hiPSC-EVs improved parenchymal histologic changes. No overt changes in vasculature structure were observed on immunohistochemistry in our in vitro model. However, VEGFa and anti-oxidant genes were upregulated with diPSC-EVs, suggesting a pro-angiogenic and cytoprotective potential. EV proteomic analysis provided new insights in regard to potential pathways influencing lung regeneration. CONCLUSION: This proof-of-concept in vitro study reveals a potential role for hiPSC- and diPSC-EVs in attenuating lung changes associated with prematurity and oxygen exposure. Our findings pave the way for a novel cell free approach to prevent and/or treat BPD, and ultimately reduce the global burden of the disease.


Subject(s)
Bronchopulmonary Dysplasia , Extracellular Vesicles , Hyperoxia , Induced Pluripotent Stem Cells , Lung Injury , Animals , Mice , Humans , Infant, Newborn , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Animals, Newborn , Induced Pluripotent Stem Cells/metabolism , Lung Injury/therapy , Lung Injury/etiology , Antioxidants/metabolism , Proteomics , Infant, Premature , Lung/pathology , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Extracellular Vesicles/metabolism
15.
J Clin Invest ; 134(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38488005

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung disease common in extreme preterm infants and is characterized by alveolar simplification. Current BPD research mainly focuses on alveolar type 2 (AT2) cells, myofibroblasts, and the endothelium. However, a notable gap exists in the involvement of AT1 cells, which constitute a majority of the alveolar surface area. In this issue of the JCI, Callaway and colleagues explored the role of TGF-ß signaling in AT1 cells for managing the AT1-to-AT2 transition and its involvement in the integration of mechanical forces with the pulmonary matrisome during development. The findings implicate AT1 cells in the pathogenesis of BPD.


Subject(s)
Bronchopulmonary Dysplasia , Animals , Infant , Infant, Newborn , Humans , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Animals, Newborn , Infant, Premature , Lung/pathology , Transforming Growth Factor beta
16.
BMC Genomics ; 25(1): 301, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38515015

ABSTRACT

BACKGROUND: Iron deficiency (ID) during the fetal-neonatal period results in long-term neurodevelopmental impairments associated with pervasive hippocampal gene dysregulation. Prenatal choline supplementation partially normalizes these effects, suggesting an interaction between iron and choline in hippocampal transcriptome regulation. To understand the regulatory mechanisms, we investigated epigenetic marks of genes with altered chromatin accessibility (ATAC-seq) or poised to be repressed (H3K9me3 ChIP-seq) in iron-repleted adult rats having experienced fetal-neonatal ID exposure with or without prenatal choline supplementation. RESULTS: Fetal-neonatal ID was induced by limiting maternal iron intake from gestational day (G) 2 through postnatal day (P) 7. Half of the pregnant dams were given supplemental choline (5.0 g/kg) from G11-18. This resulted in 4 groups at P65 (Iron-sufficient [IS], Formerly Iron-deficient [FID], IS with choline [ISch], and FID with choline [FIDch]). Hippocampi were collected from P65 iron-repleted male offspring and analyzed for chromatin accessibility and H3K9me3 enrichment. 22% and 24% of differentially transcribed genes in FID- and FIDch-groups, respectively, exhibited significant differences in chromatin accessibility, whereas 1.7% and 13% exhibited significant differences in H3K9me3 enrichment. These changes mapped onto gene networks regulating synaptic plasticity, neuroinflammation, and reward circuits. Motif analysis of differentially modified genomic sites revealed significantly stronger choline effects than early-life ID and identified multiple epigenetically modified transcription factor binding sites. CONCLUSIONS: This study reveals genome-wide, stable epigenetic changes and epigenetically modifiable gene networks associated with specific chromatin marks in the hippocampus, and lays a foundation to further elucidate iron-dependent epigenetic mechanisms that underlie the long-term effects of fetal-neonatal ID, choline, and their interactions.


Subject(s)
Iron Deficiencies , Iron , Pregnancy , Female , Animals , Rats , Male , Iron/metabolism , Chromatin/genetics , Chromatin/metabolism , Animals, Newborn , Rats, Sprague-Dawley , Epigenesis, Genetic , Choline/pharmacology , Choline/metabolism , Hippocampus
17.
J Biochem Mol Toxicol ; 38(4): e23680, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38511245

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease in newborns, which severely influences the health of infants and lacks effective clinical treatment strategies. The pathogenesis of BPD is correlated to enhanced inflammation and activated oxidative stress (OS). The application of antioxidants and anti-inflammatory treatment could be hot spots for BPD treatment. Nesfatin-1, a peptide with a suppressive property against inflammation, was tested herein for its potential therapeutic value in BPD. Neonatal SD rats were stimulated with hyperoxia, followed by being intraperitoneally administered with 20 µg/kg/day Nesfatin-1 for 2 weeks. Decreased RAC value in lung tissues, increased wet weight/dry weight (W/D) pulmonary ratio and bronchoalveolar lavage fluid (BALF) proteins, elevated cytokine release in BALF, increased malondialdehyde (MDA) content, and declined superoxide dismutase (SOD) activity were observed in BPD rats, all of which were sharply mitigated by Nesfatin-1. Rat epithelial type II cells (AECIIs) were handled with hyperoxia, and then cultured with 1 and 10 nM Nesfatin-1. Reduced cell viability, elevated lactate dehydrogenase production, elevated cytokine secretion, elevated MDA content, and decreased SOD activity were observed in hyperoxia-handled AECIIs, all of which were markedly alleviated by Nesfatin-1. Furthermore, activated nuclear factor-κB (NF-κB) signaling observed in both BPD rats and hyperoxia-handled AECIIs were notably repressed by Nesfatin-1. Collectively, Nesfatin-1 alleviated hyperoxia-triggered BPD by repressing inflammation and OS via the NF-κB signaling pathway.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Animals , Humans , Infant, Newborn , Rats , Animals, Newborn , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/metabolism , Cytokines/metabolism , Disease Models, Animal , Hyperoxia/metabolism , Inflammation/metabolism , Lung/metabolism , NF-kappa B/metabolism , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/metabolism
18.
Neurosci Lett ; 825: 137707, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38431039

ABSTRACT

Visfatin play an essential role in the central regulation of appetite in birds. This study aimed to determine role of intracerebroventricular (ICV) injection of the visfatin on food intake and its possible interaction with neuropeptide Y (NPY) and nitric oxide system in neonatal broiler chicken. In experiment 1, neonatal chicken received ICV injection visfatin (1, 2 and 4 µg). In experiment 2, chicken received ICV injection of B5063 (NPY1 receptor antagonist 1.25 µg), visfatin (4 µg) and co-injection of the B5063 + Visfatin. In experiments 3-6, SF22 (NPY2 receptor antagonist 1.25 µg), SML0891 (NPY5 receptor antagonist 1.25 µg), L-NAME (nitric oxide synthase inhibitor, 100 nmol) and L-arginine (Precursor of nitric oxide, 200 nmol) were injected instead of B5063. Then the amount of cumulative food was measured at 30, 60 and 120 min after injection. Obtained data showed, injection visfatin (2 and 4 µg) increased food intake compared to control group (P < 0.05). Co-injection of the B5063 + Visfatin decreased visfatin-induced hyperphagia compared to control group (P < 0.05). Co-injection of the L-NAME + Visfatin amplified visfatin-induced hyperphagia compared to control group (P < 0.05). The result showed that visfatin has hyperphagic role and this effect mediates via NPY1 and nitric oxide system in neonatal chicken.


Subject(s)
Chickens , Neuropeptide Y , Animals , Animals, Newborn , Neuropeptide Y/pharmacology , Chickens/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide , Nicotinamide Phosphoribosyltransferase , Eating , Receptors, Neuropeptide Y , Hyperphagia , Feeding Behavior/physiology
19.
Sci Rep ; 14(1): 6416, 2024 03 17.
Article in English | MEDLINE | ID: mdl-38494527

ABSTRACT

Neonatal hypoxia has a negative impact on the developing brain during the sensitive period. Inflammation plays a key role in the physiological response to hypoxic stress. Considering the anti-inflammatory properties of alpha-pinene, which has received a lot of attention in recent years, in this research we focused on the impact of alpha-pinene on the behavioral responses and proinflammatory factors in rats subjected to the neonatal hypoxia. This study involved Wistar rats (7-day-old) that were divided into six experimental groups, including a control group, groups receiving different doses of alpha-pinene (5 and 10 mg/kg), a hypoxia group receiving 7% O2 and 93% N2, 90 min duration for 7 days, and groups receiving alpha-pinene 30 min before hypoxia. All injections were done intraperitoneally. The rats were evaluated for proinflammatory factors 24 h after exposure to hypoxia (PND14) and at the end of the behavioral test (PND54). The results showed that hypoxia led to decreased motor activity, coordination, and memory, as well as increased inflammation. However, the rats that received alpha-pinene showed improved behavioral responses and reduced inflammation compared to the hypoxia group (all cases p < 0.05). This suggests that alpha-pinene may have a protective effect via anti-inflammatory properties against the negative impacts of hypoxia on the developing brain.


Subject(s)
Bicyclic Monoterpenes , Hypoxia-Ischemia, Brain , Rats , Animals , Rats, Wistar , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia/drug therapy , Inflammation/drug therapy , Anti-Inflammatory Agents/therapeutic use , Animals, Newborn
20.
STAR Protoc ; 5(1): 102919, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38427567

ABSTRACT

Early-life nutrition fundamentally influences newborn development and health. Here, we present a protocol for nutritional intervention in neonatal rats using the "pup-in-a-cup" artificial rearing system. We describe steps for rat milk substitute preparation, cheek cannulation and maintenance, and nutritional manipulation during the suckling period. This protocol enables investigation into the role of nutritional factors in newborns by artificially rearing rats away from the mother with experimental diets starting at postnatal day 4 for up to 18 days. For complete details on the use and execution of this protocol, please refer to Wang et al.,1 Choudhary et al.,2 and Mu et al.3,4.


Subject(s)
Animals, Newborn , Rats , Animals
SELECTION OF CITATIONS
SEARCH DETAIL
...