Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
2.
Int J Mol Sci ; 24(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36982947

ABSTRACT

Inhaled nitric oxide (iNO) is a therapy used in neonates with pulmonary hypertension. Some evidence of its neuroprotective properties has been reported in both mature and immature brains subjected to injury. NO is a key mediator of the VEGF pathway, and angiogenesis may be involved in the reduced vulnerability to injury of white matter and the cortex conferred by iNO. Here, we report the effect of iNO on angiogenesis in the developing brain and its potential effectors. We found that iNO promotes angiogenesis in the developing white matter and cortex during a critical window in P14 rat pups. This shift in the developmental program of brain angiogenesis was not related to a regulation of NO synthases by exogenous NO exposure, nor the VEGF pathway or other angiogenic factors. The effects of iNO on brain angiogenesis were found to be mimicked by circulating nitrate/nitrite, suggesting that these carriers may play a role in transporting NO to the brain. Finally, our data show that the soluble guanylate cyclase/cGMP signaling pathway is likely to be involved in the pro-angiogenetic effect of iNO through thrombospondin-1, a glycoprotein of the extracellular matrix, inhibiting soluble guanylate cyclase through CD42 and CD36. In conclusion, this study provides new insights into the biological basis of the effect of iNO in the developing brain.


Subject(s)
Nitric Oxide , Rodentia , Animals , Rats , Nitric Oxide/metabolism , Animals, Newborn , Rodentia/metabolism , Soluble Guanylyl Cyclase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Brain/metabolism , Administration, Inhalation
4.
J Neural Transm (Vienna) ; 130(3): 473-479, 2023 03.
Article in English | MEDLINE | ID: mdl-36719463

ABSTRACT

On June 2022, the 2nd Webinar "Neurodevelopmental Disorders (NDD) without boundaries took place at the Imagine Institute in Paris and was broadcasted live and in replay. The aim of this webinar is to address NDD in a dimensional rather than in a categorical approach. Several speakers were invited to present their researches on the subject. Classifications in NDD were discussed: irritability in NDD, involvement of the immune system in neurodevelopment, nutrition and gut microbiota modulate brain inflammation and neurodevelopment, co-occurring conditions in autistic adolescents and adults without intellectual disability. Classifications in psychiatric disorders were asked: mapping the effect of genes on cognition and autism risk, epigenetics and symptomatic trajectory in neurodevelopmental disorders, the autism-schizophrenia continuum in two examples: minor neurological signs and EEG microstates, the cerebellum in schizophrenia and autism: from imaging to intervention perspectives. Both genetic and environmental factors, along with clinical and imaging features, argue toward a continnum between NDD but also with adult psychiatric presentations. This new paradigm could modify the therapeutic strategy, with the development of large-spectrum treatments or new psychotherapies addressing co-occuring symptoms. The complexity and the heterogeneity of NDD apply well to the next scientific and political challenges: developing international convergence to push back the frontiers of our knowledge. This article is a summary of the 2nd webinar "Neurodevelopmental Disorders (NDD) without boundaries: research and interventions beyond classifications" sponsored by the French National Academy of Medicine, the autism and neurodevelopmental disorders scientific interest group (GIS), the International Research Network Dev-O-Psy and the French Institute of Psychiatry (GDR3557). Oral presentations are available as a replay on the following website (in French): https://autisme-neurodev.org/evenements/2022/04/12/tnd-sans-frontieres-la-recherche-et-les-interventions-au-dela-des-classifications/ .


Subject(s)
Autistic Disorder , Intellectual Disability , Neurodevelopmental Disorders , Adult , Adolescent , Humans , Neurodevelopmental Disorders/therapy , Intellectual Disability/genetics , Psychotherapy
5.
BMC Womens Health ; 22(1): 525, 2022 12 17.
Article in English | MEDLINE | ID: mdl-36526995

ABSTRACT

BACKGROUND: Endometriosis is a pathophysiological condition characterized by glands and stroma outside the uterus in regions such as the bladder, ureter, fallopian tubes, peritoneum, ovaries, and even in extra pelvic sites. One of the main clinical problems of endometriosis is chronic pelvic pain (CPP), which considerably affects the patients' quality of life. Patients with endometriosis may, cyclically or non-cyclically (80% of cases) experience CPP. High levels of anxiety and depression have been described in patients with endometriosis related to CPP; however, this has not been evaluated in endometriosis women with different types of CPP. Therefore, the research question of this study was whether there is a difference in the emotional dysregulation due to the type of pain experienced by women with endometriosis? METHODS: This work was performed in the National Institute of Perinatology (INPer) in Mexico City from January 2019 to March 2020 and aimed to determine if there are differences in emotional dysregulation in patients with cyclical and non-cyclical CPP. 49 women from 18 to 52 years-old diagnosed with endometriosis presenting cyclical and non-cyclical CPP answered several batteries made up of Mini-Mental State Examination, Visual Analog Scale, Beck's Depression Inventory, State Trait-Anxiety Inventory, and Generalized Anxiety Inventory. Mann-Whitney U and Student's t-test for independent samples to compare the difference between groups was used. Relative risk estimation was performed to determine the association between non-cyclical and cyclical CPP with probability of presenting emotional dysregulation. RESULTS: We observed that patients with non-cyclical CPP exhibited higher levels of depression and anxiety (trait-state and generalized anxiety) than patients with cyclical pain, p < 0.05 was considered significant. No differences were observed in pain intensity, but there was a higher probability of developing emotional dysregulation (anxiety or depression) in patients with non-cyclical CPP. No differences were observed in cognitive impairment. CONCLUSIONS: Our data suggest that patients with non-cyclical (persistent) CPP present a higher emotional dysregulation than those with cyclical pain.


Subject(s)
Chronic Pain , Endometriosis , Humans , Female , Adolescent , Young Adult , Adult , Middle Aged , Endometriosis/diagnosis , Quality of Life/psychology , Pelvic Pain/etiology , Pelvic Pain/psychology , Anxiety/psychology
6.
Chirality ; 25(3): 180-4, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23280651

ABSTRACT

Coatline A (1) and α-epi-coatline A (4) co-occur in the trunk extract of Andira coriacea. Inspection of their chiroptical properties led to intriguing results. After a careful examination of the experimental data used for the previously reported absolute configuration of these compounds, some uncertainties were identified. A combined theoretical approach including conformational analyses and calculation of electronic circular dichroism (ECD) spectra, in addition with experimental data obtained for schoepfin A (5) and the new schoepfin D (6) isolated from Senna quinquangulata, allowed the revision of the absolute configuration of coatlines A (1) and B (2).


Subject(s)
Circular Dichroism , Quantum Theory , Molecular Conformation , Plant Bark/chemistry , Plant Extracts/chemistry
7.
Brain Pathol ; 22(1): 1-16, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21615591

ABSTRACT

Perinatal brain injury including white matter damage (WMD) is highly related to sensory, motor or cognitive impairments in humans born prematurely. Our aim was to examine the neuroanatomical, functional and behavioral changes in adult rats that experienced prenatal ischemia (PI), thereby inducing WMD. PI was induced by unilateral uterine artery ligation at E17 in pregnant rats. We assessed performances in gait, cognitive abilities and topographical organization of maps, and neuronal and glial density in primary motor and somatosensory cortices, the hippocampus and prefrontal cortex, as well as axonal degeneration and astrogliosis in white matter tracts. We found WMD in corpus callosum and brainstem, and associated with the hippocampus and somatosensory cortex, but not the motor cortex after PI. PI rats exhibited mild locomotor impairments associated with minor signs of spasticity. Motor map organization and neuronal density were normal in PI rats, contrasting with major somatosensory map disorganization, reduced neuronal density, and a marked reduction of inhibitory interneurons. PI rats exhibited spontaneous hyperactivity in open-field test and short-term memory deficits associated with abnormal neuronal density in related brain areas. Thus, this model reproduces in adult PI rats the main deficits observed in infants with a perinatal history of hypoxia-ischemia and WMD.


Subject(s)
Cognition Disorders/pathology , Cognition Disorders/physiopathology , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Neurons/pathology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Behavioral Symptoms/pathology , Behavioral Symptoms/physiopathology , Cognition Disorders/etiology , Disease Models, Animal , Female , Male , Nerve Fibers, Myelinated/pathology , Nerve Fibers, Myelinated/physiology , Neurons/physiology , Pregnancy , Rats , Rats, Sprague-Dawley
8.
Ann Neurol ; 69(2): 341-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21387379

ABSTRACT

OBJECTIVE: Perinatal infections and the systemic inflammatory response to them are critical contributors to white matter disease (WMD) in the developing brain despite the use of highly active antibiotics. Fluoroquinolones including ciprofloxacin (CIP) have intrinsic anti-inflammatory effects. We hypothesized that CIP, in addition to its antibacterial activity, could exert a neuroprotective effect by modulating white matter inflammation in response to sepsis. METHODS: We adapted an Escherichia coli sepsis model to 5-day-old rat pups (P5), to induce white matter inflammation without bacterial meningitis. We then compared the ability of CIP to modulate inflammatory-induced brain damage compared with cefotaxime (CTX) (treatment of reference). RESULTS: Compared with CTX, CIP was associated with reduced microglial activation and inducible nitric oxide synthase (iNOS) expression in the developing white matter in rat pups subjected to E. coli sepsis. In addition to reducing microglial activation, CIP was able to prevent myelination delay induced by E. coli sepsis and to promote oligodendroglial survival and maturation. We found that E. coli sepsis altered the transcription of the guidance molecules semaphorin 3A and 3F; CIP treatment was capable of reducing semaphorin 3A and 3F transcription levels to those seen in uninfected controls. Finally, in a noninfectious white matter inflammation model, CIP was associated with significantly reduced microglial activation and prevented WMD when compared to CTX. INTERPRETATION: These data strongly suggest that CIP exerts a beneficial effect in a model of E. coli sepsis-induced WMD in rat pups that is independent of its antibacterial activity but likely related to iNOS expression modulation.


Subject(s)
Anti-Infective Agents/therapeutic use , Ciprofloxacin/therapeutic use , Demyelinating Diseases/prevention & control , Escherichia coli Infections/complications , Nerve Fibers, Myelinated/drug effects , Sepsis/complications , Animals , Animals, Newborn , Demyelinating Diseases/etiology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Escherichia coli Infections/metabolism , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Semaphorin-3A/metabolism , Sepsis/metabolism , Statistics, Nonparametric
9.
J Neuropathol Exp Neurol ; 69(8): 828-37, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20613635

ABSTRACT

Inhaled nitric oxide (iNO) is one of the most promising therapies used in neonates, but there is little information available about its effect on the developing brain. We explored the effects of both iNO and endogenous NO on developing white matter in rodents. Rat or mouse pups and their mothers were placed in a chamber containing 5 to 20 ppm of NO for 7 days after birth. Neonatal exposure to iNO was associated with a transient increase in central nervous system myelination in rats and C57BL/6 mice without any deleterious effects at low doses (5 ppm) or behavioral consequences in adulthood. Exposure to iNO was associated with a proliferative effect on immature oligodendrocytes and a subsequent promaturational effect. The role of endogenous NO in myelination was investigated in animals treated with the nitric oxides synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME) in the neonatal period; this led to protracted myelination defects and subsequent behavioral deficits in adulthood. These effects were reversed by rescuing L-NAME-treated animals with iNO. Thus, we demonstrate considerable effect of both exogenous and endogenous NO on myelination in rodents. These data point to potential new avenues for neuroprotection in human perinatal brain damage.


Subject(s)
Brain/drug effects , Brain/growth & development , Free Radical Scavengers/pharmacology , Nerve Fibers, Myelinated/drug effects , Nitric Oxide/administration & dosage , Nitric Oxide/metabolism , Administration, Inhalation , Age Factors , Animals , Animals, Newborn , Antigens/metabolism , Behavioral Symptoms/chemically induced , Behavioral Symptoms/enzymology , Brain/cytology , Cell Proliferation/drug effects , Central Nervous System/cytology , Central Nervous System/drug effects , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , In Situ Nick-End Labeling/methods , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/ultrastructure , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Neuropsychological Tests , Nitric Oxide Synthase Type II/pharmacology , O Antigens/metabolism , Oligodendroglia/drug effects , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Space Perception/drug effects , Spatial Behavior/drug effects , Statistics, Nonparametric
10.
PLoS One ; 5(6): e10916, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20532231

ABSTRACT

BACKGROUND: Inhaled nitric oxide (iNO) is one of the most promising therapies used in neonates. However, little information is known about its impact on the developing brain submitted to excitotoxic challenge. METHODOLOGY/PRINCIPAL FINDINGS: We investigated here the effect of iNO in a neonatal model of excitotoxic brain lesions. Rat pups and their dams were placed in a chamber containing 20 ppm NO during the first week of life. At postnatal day (P)5, rat pups were submitted to intracranial injection of glutamate agonists. At P10, rat pups exposed to iNO exhibited a significant decrease of lesion size in both the white matter and cortical plate compared to controls. Microglia activation and astrogliosis were found significantly decreased in NO-exposed animals. This neuroprotective effect was associated with a significant decrease of several glutamate receptor subunits expression at P5. iNO was associated with an early (P1) downregulation of pCREB/pAkt expression and induced an increase in pAkt protein concentration in response to excitotoxic challenge (P7). CONCLUSION: This study is the first describe and investigate the neuroprotective effect of iNO in neonatal excitotoxic-induced brain damage. This effect may be mediated through CREB pathway and subsequent modulation of glutamate receptor subunits expression.


Subject(s)
Brain/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide/pharmacology , Administration, Inhalation , Animals , Animals, Newborn , Magnetic Resonance Spectroscopy , Neuroprotective Agents/administration & dosage , Nitric Oxide/administration & dosage , Rats
11.
PLoS One ; 4(9): e7128, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19771167

ABSTRACT

OBJECTIVE: To investigate the effects of melatonin treatment in a rat model of white matter damage (WMD) in the developing brain. Additionally, we aim to delineate the cellular mechanisms of melatonin effect on the oligodendroglial cell lineage. METHODS: A unilateral ligation of the uterine artery in pregnant rat at the embryonic day 17 induces fetal hypoxia and subsequent growth restriction (GR) in neonatal pups. GR and control pups received a daily intra-peritoneal injection of melatonin from birth to post-natal day (P) 3. RESULTS: Melatonin administration was associated with a dramatic decrease in microglial activation and astroglial reaction compared to untreated GR pups. At P14, melatonin prevented white matter myelination defects with an increased number of mature oligodendrocytes (APC-immunoreactive) in treated GR pups. Conversely, melatonin was not found to be associated with an increased density of total oligodendrocytes (Olig2-immunoreactive), suggesting that melatonin is able to promote oligodendrocyte maturation but not proliferation. These effects appear to be melatonin-receptor dependent and were reproduced in vitro. INTERPRETATION: These data suggest that melatonin has a strong protective effect on developing damaged white matter through decreased microglial activation and oligodendroglial maturation leading to a normalization of the myelination process. Consequently, melatonin should be a considered as an effective neuroprotective candidate not only in perinatal brain damage but also in inflammatory and demyelinating diseases observed in adults.


Subject(s)
Gene Expression Regulation, Developmental , Melatonin/physiology , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/metabolism , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Cell Lineage , Cells, Cultured , Immunohistochemistry/methods , Melatonin/metabolism , Microscopy, Fluorescence/methods , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
12.
J Clin Invest ; 119(10): 3089-101, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19729834

ABSTRACT

Cyclin I is an atypical cyclin because it is most abundant in postmitotic cells. We previously showed that cyclin I does not regulate proliferation, but rather controls survival of podocytes, terminally differentiated epithelial cells that are essential for the structural and functional integrity of kidney glomeruli. Here, we investigated the mechanism by which cyclin I safeguards against apoptosis and found that cyclin I bound and activated cyclin-dependent kinase 5 (Cdk5) in isolated mouse podocytes and neurons. Cdk5 activity was reduced in glomeruli and brain lysates from cyclin I-deficient mice, and inhibition of Cdk5 increased in vitro the susceptibility to apoptosis in response to cellular damage. In addition, levels of the prosurvival proteins Bcl-2 and Bcl-XL were reduced in podocytes and neurons from cyclin I-deficient mice, and restoration of Bcl-2 or Bcl-XL expression prevented injury-induced apoptosis. Furthermore, we found that levels of phosphorylated MEK1/2 and ERK1/2 were decreased in cyclin I-deficient podocytes and that inhibition of MEK1/2 restored Bcl2 and Bcl-XL protein levels. Of interest, this pathway was also defective in mice with experimental glomerulonephritis. Taken together, these data suggest that a cyclin I-Cdk5 complex forms a critical antiapoptotic factor in terminally differentiated cells that functions via MAPK signaling to modulate levels of the prosurvival proteins Bcl-2 and Bcl-XL.


Subject(s)
Cyclin I/physiology , Cyclin-Dependent Kinase 5/metabolism , Mitosis , Podocytes/metabolism , Proto-Oncogene Proteins/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis , Brain/metabolism , Caspase 3/metabolism , Cell Differentiation , Cell Survival , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , HEK293 Cells , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphotransferases/metabolism , Podocytes/enzymology , Podocytes/physiology , Protein Processing, Post-Translational , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , bcl-X Protein/genetics
13.
PLoS One ; 4(5): e5509, 2009.
Article in English | MEDLINE | ID: mdl-19434240

ABSTRACT

The neuropeptide somatostatin has been suggested to play an important role during neuronal development in addition to its established modulatory impact on neuroendocrine, motor and cognitive functions in adults. Although six somatostatin G protein-coupled receptors have been discovered, little is known about their distribution and function in the developing mammalian brain. In this study, we have first characterized the developmental expression of the somatostatin receptor sst2A, the subtype found most prominently in the adult rat and human nervous system. In the rat, the sst2A receptor expression appears as early as E12 and is restricted to post-mitotic neuronal populations leaving the ventricular zone. From E12 on, migrating neuronal populations immunopositive for the receptor were observed in numerous developing regions including the cerebral cortex, hippocampus and ganglionic eminences. Intense but transient immunoreactive signals were detected in the deep part of the external granular layer of the cerebellum, the rostral migratory stream and in tyrosine hydroxylase- and serotonin- positive neurons and axons. Activation of the sst2A receptor in vitro in rat cerebellar microexplants and primary hippocampal neurons revealed stimulatory effects on neuronal migration and axonal growth, respectively. In the human cortex, receptor immunoreactivity was located in the preplate at early development stages (8 gestational weeks) and was enriched to the outer part of the germinal zone at later stages. In the cerebellum, the deep part of the external granular layer was strongly immunoreactive at 19 gestational weeks, similar to the finding in rodents. In addition, migrating granule cells in the internal granular layer were also receptor-positive. Together, theses results strongly suggest that the somatostatin sst2A receptor participates in the development and maturation of specific neuronal populations during rat and human brain ontogenesis.


Subject(s)
Axons/metabolism , Brain/embryology , Brain/metabolism , Cell Movement , Neurons/cytology , Neurons/metabolism , Receptors, Somatostatin/metabolism , Animals , Animals, Newborn , Brain/cytology , Dendrites/metabolism , Fluorescent Antibody Technique , Gestational Age , Humans , Organ Specificity , Protein Transport , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
14.
Semin Fetal Neonatal Med ; 14(1): 28-34, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18986855

ABSTRACT

Bronchopulmonary dysplasia is a chronic lung disease that affects premature infants and contributes to their morbidity and mortality. With the advent of prenatal steroids and postnatal exogenous surfactant and less aggressive respiratory support, premature infants can develop chronic oxygen dependency without even acute respiratory distress. This 'new bronchopulmonary dysplasia' could be the result of impaired postnatal growth. Several experimental studies have suggested a possible role of the vascular endothelial growth factor/nitric oxide (VEGF/NO) pathway in restoring pulmonary angiogenesis and enhancing distal lung growth. The results of the clinical studies are, however, inconclusive, and it is currently unclear which subsets of premature infants might benefit from inhaled nitric oxide. Besides, severe intracranial haemorrhage and/or cystic periventricular leucomalacia may affect the most immature babies, many of whom are spared from severe initial respiratory disease. Recently, inhaled nitric oxide was shown to significantly decrease the incidence of these neurological events, and to improve the long-term outcome in a few clinical trials. At times neuroprotective, at times neurotoxic, nitric oxide is capable of divergent effects depending upon the extent of cerebral damage, the redox state of the cell, and the experimental model used. Recently, our group found that inhaled nitric oxide had remote effects including angiogenesis and maturation on the developing brain in rodent pups. Thus, we await the results of the recently completed randomised clinical trial of inhaled nitric oxide to prevent bronchopulmonary dysplasia (the European Nitric Oxide or 'EUNO' trial) where, besides the primary endpoint of chronic oxygen dependency reduction at 36 weeks' postconceptional age, long-term lung and brain will be followed-up until 7 years of age.


Subject(s)
Bronchodilator Agents/pharmacology , Bronchopulmonary Dysplasia/prevention & control , Nitric Oxide/pharmacology , Administration, Inhalation , Animals , Brain/drug effects , Brain/growth & development , Humans , Infant, Newborn , Infant, Premature , Lung/metabolism , Models, Animal , Vascular Endothelial Growth Factors/metabolism
15.
Pediatr Res ; 65(3): 296-300, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19092727

ABSTRACT

Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) have been implicated in normal brain development, adult stroke, and, more recently, perinatal brain injury. Here, our objective was to obtain comprehensive and comparative data on the ontogeny of MMP-2, MMP-9, TIMP-1, and TIMP-2 in the neocortex of male and female mice belonging to various strains, from embryonic life to adulthood. We used gelatin zymography, ELISA, and real-time PCR analyses. MMP-2, MMP-9, and TIMP-1 activity and/or expression peaked during embryonic life and the early neonatal period, whereas TIMP-2 peaked during the first two postnatal weeks. Comparable results were obtained in all the mouse strains except BALB/c, where MMP-2 levels were considerably lower at all ages compared with the other strains. No gender effect was observed on any of the study parameters. This comprehensive study will serve as a basis for future investigations into the role for MMPs and TIMPs in normal brain development and prenatal brain injury.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Matrix Metalloproteinases/metabolism , Neocortex/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Age Factors , Animals , Electrophoresis , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Mice, Mutant Strains , Neocortex/embryology , Neocortex/growth & development , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
16.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R801-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19109375

ABSTRACT

Anemia and hypoxia in rats result in an increase in factors potentially involved in cerebral angiogenesis. Therefore, the aim of this study was to assess the effect of chronic anemia and/or chronic hypoxia on cerebral cellular responses and angiogenesis in wild-type and anemic transgenic mice. These studies were done in erythropoietin-deficient mice (Epo-TAg(h)) in normoxia and following acute (one day) and chronic (14 days, barometric pressure = 420 mmHg) hypoxia. In normoxia, Epo-TAg(h) mice showed an increase in transcript and protein levels of hypoxia-inducible factor 1alpha (HIF-1alpha), vascular endothelial growth factor (VEGF), erythropoietin receptors (EpoR), phospho-STAT-5/STAT-5 ratio, and neuronal neuronal nitric oxide synthase (nNOS) along with a higher cerebral capillary density. In wild-type (WT) mice, acute hypoxia increased all of the studied factors, while in chronic hypoxia, HIF-1alpha, EpoR, phospho-STAT-5/STAT-5 ratio, nNOS, and inducible NOS remained elevated, with an increase in capillary density. Surprisingly, in Epo-TAg(h) mice, chronic hypoxia did not further increase any factor except the nitric oxide metabolites, while HIF-1alpha, EpoR, and phospho-STAT-5/STAT-5 ratio were reduced. Normoxic Epo-TAg(h) mice developed cerebral angiogenesis through the HIF-1alpha/VEGF pathway. In acute hypoxia, WT mice up-regulated all of the studied factors, including cerebral NO. Polycythemia and angiogenesis occurred with acclimatization to chronic hypoxia only in WT mice. In Epo-TAg(h), the decrease in HIF-1alpha, VEGF proteins, and phospho-STAT-5 ratio in chronic hypoxia suggest that neuroprotective and angiogenesis pathways are altered.


Subject(s)
Anemia/physiopathology , Brain/physiopathology , Erythropoietin/deficiency , Erythropoietin/genetics , Hypoxia/physiopathology , Animals , Body Weight/physiology , Cerebral Cortex/metabolism , Chronic Disease , Erythropoietin/metabolism , Hemoglobins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoassay , Immunohistochemistry , Male , Mice , Mice, Inbred CBA , Mice, Knockout , Nitric Oxide/metabolism , RNA/biosynthesis , RNA/isolation & purification , Receptors, Erythropoietin/biosynthesis , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
17.
Proc Natl Acad Sci U S A ; 105(43): 16779-84, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18922769

ABSTRACT

White-matter damage is a leading cause of neurological handicap. Although hypoxia-ischemia and excitotoxicity are major pathogenic factors, a role for genetic influences was suggested recently. Thus, protracted gestational hypoxia was associated with white-matter damage (WMD) in rat pups but not in mouse pups. Indeed, microglial activation and vessel-wall density on postnatal days (P)1 and P10 were found increased in both mouse and rat pups, but cell death, astrogliosis, and myelination were only significantly altered in hypoxic rat pups. We investigated whether this species-related difference was ascribable to effects of antenatal hypoxia on the expression of glutamate receptor subunits by using immunocytochemistry, PCR, and excitotoxic double hit insult. Quantitative PCR in hypoxic mouse pups on P1 showed 2- to 4-fold down-regulation of the AMPA-receptor subunits -1, 2, and -4; of the kainate-receptor subunit GluR7; and of the metabotropic receptor subunits mGluR1, -2, -3, -5, and -7. None of the glutamate-receptor subunits was down-regulated in the hypoxic rat pups. NR2B was the only NMDA-receptor subunit that was down-regulated in hypoxic mice but not in hypoxic rat on P1. Ifenprodil administration to induce functional inhibition of NMDA containing NR2B-subunit receptors prevented hypoxia-induced myelination delay in rat pups. Intracerebral injection of a glutamate agonist produced a larger decrease in ibotenate-induced excitotoxic lesions in hypoxic mouse pups than in normoxic mouse pups. Gestational hypoxia may regulate the expression of specific glutamate-receptor subunits in fetal mice but not in fetal rats. Therefore, genetic factors may influence the susceptibility of rodents to WMD.


Subject(s)
Gene Expression Regulation , Hypoxia/genetics , Nerve Fibers, Myelinated/pathology , Receptors, Glutamate/genetics , Animals , Animals, Newborn , Female , Genetic Predisposition to Disease , Hypoxia/etiology , Mice , Pregnancy , Protein Subunits/genetics , Rats , Receptors, AMPA/genetics , Receptors, Kainic Acid/genetics , Receptors, Metabotropic Glutamate/genetics , Species Specificity
18.
J Neuropathol Exp Neurol ; 67(10): 994-1000, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18800008

ABSTRACT

Intracerebral injection of ibotenate in newborn rodents produces brain damage that mimics that of infants with cerebral palsy. Because maternal infection may contribute to brain injury in preterm infants, we investigated brain damage after maternal inflammation and postnatal ibotenate treatment in a rat model of cerebral palsy. Pregnant rats were injected intraperitoneally with lipopolysaccharide at Days 19 and 20 of gestation. Neonates were given intracerebral injections of ibotenate at postnatal Day 4 and were then killed at Day 9. Lesion sizes were measured by cresyl violet staining, and microglial activation, astrogliosis, and myelination were evaluated by immunohistochemistry. The lipopolysaccharide groups had larger cortical and white matter lesions than the control group; they also had significantly greater microglial activation and astrogliosis and less white matter myelination in the lesioned hemispheres compared with the controls. Thus, maternal endotoxin exposure may affect prenatal development of the offspring and modulate the subsequent development of excitotoxic brain lesions. These results demonstrate the critical influence of prenatal immune events on neonatal central nervous system vulnerability and provide a model for studying the pathophysiology of cerebral damage in preterm infants and, specifically, the interplay between brain inflammation and excitotoxicity.


Subject(s)
Animals, Newborn/physiology , Brain/drug effects , Brain/pathology , Endotoxins/toxicity , Excitatory Amino Acid Agonists/toxicity , Ibotenic Acid/toxicity , Lipopolysaccharides/toxicity , Animals , CD11b Antigen/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Immunohistochemistry , Microglia/pathology , Myelin Basic Protein/metabolism , Myelin Sheath/pathology , Nerve Fibers/pathology , Pregnancy , Rats , Rats, Wistar
19.
J Magn Reson Imaging ; 28(4): 1046-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18821607

ABSTRACT

PURPOSE: To investigate whether USPIO-enhanced magnetic resonance imaging (MRI) detected reticuloendothelial system (RES) cells in newborn normal rats. MATERIALS AND METHODS: Newborn normal rats were imaged in vivo on a 1.5 T MR system, 2-96 hours after intraperitoneal Ferumoxtran-10 (n = 38) or saline injection (control group, n = 5). Signals from liver, spleen, and vertebral bone marrow were measured (T2-weighted Turbo Spin Echo) to describe the kinetics of enhancement. The pups were sacrificed and iron concentrations in plasma and peritoneal fluid were measured using atomic absorption spectrometry. Prussian blue-labeled cells density in liver, spleen, and vertebral bone marrow was assessed. RESULTS: Significant (P < 0.05) negative enhancement of the liver, spleen, and vertebral bone marrow was noted after Ferumoxtran-10 injection (2-96 hours for liver and spleen, 4-96 hours for bone marrow). Ferumoxtran-10 was absorbed from the peritoneum in the first 8 hours postinjection, entering the circulation with a plasma peak (8 hours); then Ferumoxtran-10 returned over the baseline in plasma (96 hours). Important intracellular iron deposition in liver and spleen was measured postinjection (3-96 hours, P < 0.05). Limited but significant intracellular iron deposition was noted in vertebral bone marrow postinjection (96 hours, P < 0.05), suggesting that Ferumoxtran-10 selectively labeled RES cells after 96 hours and produced nonspecific labeling at earlier timepoints. CONCLUSION: Ferumoxtran-10-enhanced MRI visualizes RES cells in vivo in newborn rats.


Subject(s)
Iron/pharmacokinetics , Magnetic Resonance Imaging/methods , Mononuclear Phagocyte System/cytology , Oxides/pharmacokinetics , Animals , Animals, Newborn , Bone Marrow/metabolism , Contrast Media/pharmacokinetics , Dextrans , Feasibility Studies , Ferrosoferric Oxide , Magnetite Nanoparticles , Rats , Rats, Sprague-Dawley , Spleen/metabolism , Statistics, Nonparametric , Tissue Distribution
20.
Pediatr Res ; 62(5): 604-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18049373

ABSTRACT

Caffeine is frequently administered in human preterm newborns. Although some data suggest a potential risk for the developing brain, its impact has not been fully evaluated. We used a murine model of postnatal caffeine treatment in which mouse pups received intraperitoneal injections of caffeine from postnatal days 3 to 10. Caffeine exposure resulted in a transient reduction of glial fibrillary acidic protein and S100beta protein expression in various brain areas during the first 2 postnatal weeks (19.8% and 23.2% reduction in the hippocampus at P15, respectively). This effect was dose-dependent and at least partly involved a reduction of glial proliferation, as a caffeine-induced decrease of 5-bromodeoxyuridine incorporation was observed in the dentate gyrus and subventricular zone (25.8% and 26.6%, respectively) and no increase of programmed cell death (cleaved caspase-3 immunostaining) was observed at postnatal day 7. This effect could be reproduced with an antagonist of A(2a) adenosine receptor (A(2a)R) and was blocked by co-injection of an agonist. These results suggest that postnatal caffeine treatment might induce an alteration of astrocytogenesis via A(2a)R blockade during brain development. Although no obvious neuritic abnormalities (microtubule-associated protein 2 and synaptophysin immunostaining) were observed, postnatal caffeine treatment could have long-term consequences on brain function.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Caffeine/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Body Weight/drug effects , Brain/cytology , Brain/growth & development , Brain/metabolism , Caffeine/administration & dosage , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Injections, Intraperitoneal , Mice , Microtubule-Associated Proteins/metabolism , Models, Animal , Nerve Growth Factors/metabolism , Organ Size/drug effects , Phenethylamines/pharmacology , Pyrimidines/pharmacology , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/metabolism , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Synaptophysin/metabolism , Time Factors , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...