Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Brain Behav Immun ; 120: 99-116, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38705494

ABSTRACT

INTRODUCTION: Despite improved management of traumatic brain injury (TBI), it still leads to lifelong sequelae and disability, particularly in children. Chronic neuroinflammation (the so-called tertiary phase), in particular, microglia/macrophage and astrocyte reactivity, is among the main mechanisms suspected of playing a role in the generation of lesions associated with TBI. The role of acute neuroinflammation is now well understood, but its persistent effect and impact on the brain, particularly during development, are not. Here, we investigated the long-term effects of pediatric TBI on the brain in a mouse model. METHODS: Pediatric TBI was induced in mice on postnatal day (P) 7 by weight-drop trauma. The time course of neuroinflammation and myelination was examined in the TBI mice. They were also assessed by magnetic resonance, functional ultrasound, and behavioral tests at P45. RESULTS: TBI induced robust neuroinflammation, characterized by acute microglia/macrophage and astrocyte reactivity. The long-term consequences of pediatric TBI studied on P45 involved localized scarring astrogliosis, persistent microgliosis associated with a specific transcriptomic signature, and a long-lasting myelination defect consisting of the loss of myelinated axons, a decreased level of myelin binding protein, and severe thinning of the corpus callosum. These results were confirmed by reduced fractional anisotropy, measured by diffusion tensor imaging, and altered inter- and intra-hemispheric connectivity, measured by functional ultrasound imaging. In addition, adolescent mice with pediatric TBI showed persistent social interaction deficits and signs of anxiety and depressive behaviors. CONCLUSIONS: We show that pediatric TBI induces tertiary neuroinflammatory processes associated with white matter lesions and altered behavior. These results support our model as a model for preclinical studies for tertiary lesions following TBI.

2.
Neurobiol Dis ; 187: 106315, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37783234

ABSTRACT

G protein-coupled receptor 17 (GPR17) and the WNT pathway are critical players of oligodendrocyte (OL) differentiation acting as essential timers in developing brain to achieve fully-myelinating cells. However, whether and how these two systems are related to each other is still unknown. Of interest, both factors are dysregulated in developing and adult brain diseases, including white matter injury and cancer, making the understanding of their reciprocal interactions of potential importance for identifying new targets and strategies for myelin repair. Here, by a combined pharmacological and biotechnological approach, we examined regulatory mechanisms linking WNT signaling to GPR17 expression in OLs. We first analyzed the relative expression of mRNAs encoding for GPR17 and the T cell factor/Lymphoid enhancer-binding factor-1 (TCF/LEF) transcription factors of the canonical WNT/ß-CATENIN pathway, in PDGFRα+ and O4+ OLs during mouse post-natal development. In O4+ cells, Gpr17 mRNA level peaked at post-natal day 14 and then decreased concomitantly to the physiological uprise of WNT tone, as shown by increased Lef1 mRNA level. The link between WNT signaling and GPR17 expression was further reinforced in vitro in primary PDGFRα+ cells and in Oli-neu cells. High WNT tone impaired OL differentiation and drastically reduced GPR17 mRNA and protein levels. In Oli-neu cells, WNT/ß-CATENIN activation repressed Gpr17 promoter activity through both putative WNT response elements (WRE) and upregulation of the inhibitor of DNA-binding protein 2 (Id2). We conclude that the WNT pathway influences OL maturation by repressing GPR17, which could have implications in pathologies characterized by dysregulations of the OL lineage including multiple sclerosis and oligodendroglioma.


Subject(s)
Oligodendrocyte Precursor Cells , Wnt Signaling Pathway , Mice , Animals , beta Catenin/metabolism , Oligodendrocyte Precursor Cells/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Cell Differentiation/physiology , Oligodendroglia/metabolism , RNA, Messenger/metabolism
3.
Ann Neurol ; 91(1): 48-65, 2022 01.
Article in English | MEDLINE | ID: mdl-34741343

ABSTRACT

OBJECTIVES: In the premature newborn, perinatal inflammation mediated by microglia contributes significantly to neurodevelopmental injuries including white matter injury (WMI). Brain inflammation alters development through neuroinflammatory processes mediated by activation of homeostatic microglia toward a pro-inflammatory and neurotoxic phenotype. Investigating immune regulators of microglial activation is crucial to find effective strategies to prevent and treat WMI. METHODS: Ex vivo microglial cultures and a mouse model of WMI induced by perinatal inflammation (interleukin-1-beta [IL-1ß] and postnatal days 1-5) were used to uncover and elucidate the role of microRNA-146b-5p in microglial activation and WMI. RESULTS: A specific reduction in vivo in microglia of Dicer, a protein required for microRNAs maturation, reduces pro-inflammatory activation of microglia and prevents hypomyelination in our model of WMI. Microglial miRNome analysis in the WMI model identified miRNA-146b-5p as a candidate modulator of microglial activation. Ex vivo microglial cell culture treated with the pro-inflammatory stimulus lipopolysaccharide (LPS) led to overexpression of immunomodulatory miRNA-146b-5p but its drastic reduction in the microglial extracellular vesicles (EVs). To increase miRNA-146b-5p expression, we used a 3DNA nanocarrier to deliver synthetic miRNA-146b-5p specifically to microglia. Enhancing microglial miRNA-146b-5p overexpression significantly decreased LPS-induced activation, downregulated IRAK1, and restored miRNA-146b-5p levels in EVs. In our WMI model, 3DNA miRNA-146b-5p treatment significantly prevented microglial activation, hypomyelination, and cognitive defect induced by perinatal inflammation. INTERPRETATIONS: These findings support that miRNA-146b-5p is a major regulator of microglia phenotype and could be targeted to reduce the incidence and the severity of perinatal brain injuries and their long-term consequences. ANN NEUROL 2022;91:48-65.


Subject(s)
Brain/pathology , MicroRNAs/metabolism , Microglia/pathology , White Matter/pathology , Animals , Mice , Neurogenesis/physiology
4.
J Neural Transm (Vienna) ; 130(3): 281-297, 2023 03.
Article in English | MEDLINE | ID: mdl-36335540

ABSTRACT

Approximately 15 million babies are born prematurely every year and many will face lifetime motor and/or cognitive deficits. Children born prematurely are at higher risk of developing perinatal brain lesions, especially white matter injuries (WMI). Evidence in humans and rodents demonstrates that systemic inflammation-induced neuroinflammation, including microglial and astrocyte reactivity, is the prominent processes of WMI associated with preterm birth. Thus, a new challenge in the field of perinatal brain injuries is to develop new neuroprotective strategies to target neuroinflammation to prevent WMI. Serotonin (5-HT) and its receptors play an important role in inflammation, and emerging evidence indicates that 5-HT may regulate brain inflammation by the modulation of microglial reactivity and astrocyte functions. The present study is based on a mouse model of WMI induced by intraperitoneal (i.p.) injections of IL-1ß during the first 5 days of life. In this model, certain key lesions of preterm brain injuries can be summarized by (i) systemic inflammation, (ii) pro-inflammatory microglial and astrocyte activation, and (iii) inhibition of oligodendrocyte maturation, leading to hypomyelination. We demonstrate that Htr7 mRNA (coding for the HTR7/5-HT7 receptor) is significantly overexpressed in the anterior cortex of IL-1ß-exposed animals, suggesting it as a potential therapeutic target. LP-211 is a specific high-affinity HTR7 agonist that crosses the blood-brain barrier (BBB). When co-injected with IL-1ß, LP-211 treatment prevented glial reactivity, the down-regulation of myelin-associated proteins, and the apparition of anxiety-like phenotypes. Thus, HTR7 may represent an innovative therapeutic target to protect the developing brain from preterm brain injuries.


Subject(s)
Brain Injuries , Premature Birth , White Matter , Animals , Mice , Pregnancy , Female , Child , Infant, Newborn , Humans , White Matter/pathology , Rodentia , Neuroinflammatory Diseases , Serotonin/metabolism , Premature Birth/metabolism , Premature Birth/pathology , Brain/metabolism , Brain Injuries/etiology , Brain Injuries/prevention & control , Inflammation/pathology , Microglia/metabolism
5.
Glia ; 70(9): 1699-1719, 2022 09.
Article in English | MEDLINE | ID: mdl-35579329

ABSTRACT

Preterm infants often show pathologies of the cerebellum, which are associated with impaired motor performance, lower IQ and poor language skills at school ages. Using a mouse model of inflammation-induced encephalopathy of prematurity driven by systemic administration of pro-inflammatory IL-1ß, we sought to uncover causes of cerebellar damage. In this model, IL-1ß is administered between postnatal day (P) 1 to day 5, a timing equivalent to the last trimester for brain development in humans. Structural MRI analysis revealed that systemic IL-1ß treatment induced specific reductions in gray and white matter volumes of the mouse cerebellar lobules I and II (5% false discovery rate [FDR]) from P15 onwards. Preceding these MRI-detectable cerebellar volume changes, we observed damage to oligodendroglia, with reduced proliferation of OLIG2+ cells at P10 and reduced levels of the myelin proteins myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) at P10 and P15. Increased density of IBA1+ cerebellar microglia were observed both at P5 and P45, with evidence for increased microglial proliferation at P5 and P10. Comparison of the transcriptome of microglia isolated from P5 cerebellums and cerebrums revealed significant enrichment of pro-inflammatory markers in microglia from both regions, but cerebellar microglia displayed a unique type I interferon signaling dysregulation. Collectively, these data suggest that perinatal inflammation driven by systemic IL-1ß leads to specific cerebellar volume deficits, which likely reflect oligodendrocyte pathology downstream of microglial activation. Further studies are now required to confirm the potential of protective strategies aimed at preventing sustained type I interferon signaling driven by cerebellar microglia as an important therapeutic target.


Subject(s)
Cerebellar Diseases , Infant, Premature, Diseases , Inflammation , Interferon Type I , Interleukin-1beta , Microglia , Animals , Brain Diseases/chemically induced , Brain Diseases/immunology , Brain Diseases/pathology , Cerebellar Diseases/chemically induced , Cerebellar Diseases/immunology , Cerebellar Diseases/pathology , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/pathology , Disease Models, Animal , Female , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/chemically induced , Infant, Premature, Diseases/immunology , Infant, Premature, Diseases/pathology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interferon Type I/immunology , Interleukin-1beta/adverse effects , Interleukin-1beta/pharmacology , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Pregnancy
6.
Int J Mol Sci ; 23(9)2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35563258

ABSTRACT

Preterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecules that block oligodendrocyte differentiation, leading to hypomyelination. Some preterm survivors can face lifetime motor and/or cognitive disabilities linked to periventricular white matter injuries (PWMIs). There is currently no recommendation concerning the mode of delivery in the case of PTB and its impact on brain development. Many animal models of induced-PTB based on LPS injections exist, but with a low survival rate. There is a lack of information regarding clinically used pharmacological substances to induce PTB and their consequences on brain development. Mifepristone (RU-486) is a drug used clinically to induce preterm labor. This study aims to elaborate and characterize a new model of induced-PTB and PWMIs by the gestational injection of RU-486 and the perinatal injection of pups with IL-1beta. A RU-486 single subcutaneous (s.c.) injection at embryonic day (E)18.5 induced PTB at E19.5 in pregnant OF1 mice. All pups were born alive and were adopted directly after birth. IL-1beta was injected intraperitoneally from postnatal day (P)1 to P5. Animals exposed to both RU-486 and IL-1beta demonstrated microglial reactivity and subsequent PWMIs. In conclusion, the s.c. administration of RU-486 induced labor within 24 h with a high survival rate for pups. In the context of perinatal inflammation, RU-486 labor induction significantly decreases microglial reactivity in vivo but did not prevent subsequent PWMIs.


Subject(s)
Microglia , Premature Birth , Animals , Animals, Newborn , Female , Humans , Inflammation , Lipopolysaccharides/toxicity , Mice , Mifepristone/pharmacology , Pregnancy
7.
BMC Infect Dis ; 19(1): 931, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31690258

ABSTRACT

BACKGROUND: The sepsis-induced immunodepression contributes to impaired clinical outcomes of various stress conditions. This syndrome is well documented and characterized by attenuated function of innate and adaptive immune cells. Several pharmacological interventions aimed to restore the immune response are emerging of which interferon-gamma (IFNγ) is one. It is of paramount relevance to obtain clinical information on optimal timing of the IFNγ-treatment, -tolerance, -effectiveness and outcome before performing a RCT. We describe the effects of IFNγ in a cohort of 18 adult and 2 pediatric sepsis patients. METHODS: In this open-label prospective multi-center case-series, IFNγ treatment was initiated in patients selected on clinical and immunological criteria early (< 4 days) or late (> 7 days) following the onset of sepsis. The data collected in 18 adults and 2 liver transplanted pediatric patients were: clinical scores, monocyte expression of HLA-DR (flow cytometry), lymphocyte immune-phenotyping (flow cytometry), IL-6 and IL-10 plasma levels (ELISA), bacterial cultures, disease severity, and mortality. RESULTS: In 15 out of 18 patients IFNγ treatment was associated with an increase of median HLA-DR expression from 2666 [IQ 1547; 4991] to 12,451 [IQ 4166; 19,707], while the absolute number of lymphocyte subpopulations were not affected, except for the decrease number of NK cells 94.5 [23; 136] to 32.5 [13; 90.8] (0.0625)]. Plasma levels of IL-6 464 [201-770] to 108 (89-140) ng/mL (p = 0.04) and IL-10 from IL-10 from 29 [12-59] to 9 [1-15] pg/mL decreased significantly. Three patients who received IFNγ early after ICU admission (<4 days) died. The other patients had a rapid clinical improvement assessed by the SOFA score and bacterial cultures that were repeatedly positive became negative. The 2 pediatric cases improved rapidly, but 1 died for hemorrhagic complication. CONCLUSION: Guided by clinical and immunological monitoring, adjunctive immunotherapy with IFNγ appears well-tolerated in our cases and improves immune host defense in sepsis induced immuno suppression. Randomized clinical studies to assess its potential clinical benefit are warranted.


Subject(s)
Immune Tolerance , Interferon-gamma/therapeutic use , Sepsis/drug therapy , Adult , Aged , Aged, 80 and over , Child , Female , HLA-DR Antigens/metabolism , Humans , Infant , Intensive Care Units , Interleukin-10/blood , Interleukin-6/blood , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Male , Middle Aged , Monocytes/cytology , Monocytes/metabolism , Prospective Studies , Pseudomonas aeruginosa/isolation & purification , Sepsis/microbiology
8.
Int J Mol Sci ; 20(15)2019 Aug 04.
Article in English | MEDLINE | ID: mdl-31382688

ABSTRACT

We recently reported that neonatal ischemia induces microglia/macrophage activation three days post-ischemia. We also found that female mice sustained smaller infarcts than males three months post-ischemia. The objective of our current study was to examine whether differential acute neuroinflammatory response and infiltrated immune cells occurs between male and females after three days post-ischemia. Permanent middle cerebral artery occlusion was induced in male and female postnatal 9-day-old (P9) mice, and mice were sacrificed three days after ischemia. Brains were analyzed for mRNA transcription after microglia magnetic cell sorting to evaluate M1 and M2 markers. FACS analysis was performed to assess myeloid infiltration and microglial expression of CX3 chemokine receptor 1 (CX3CR1). Inflammatory cytokine expression and microglia/macrophage activation were analyzed via in situ hybridization combined with immunofluorescence techniques. Lesion volume and cell death were measured. An increase in microglia/macrophages occurred in male versus female mice. The cells exhibited amoeboid morphology, and TNFα and ptgs2 (Cox-2) genes were more expressed in males. More myeloid cell infiltration was found in male versus female brains. However, we did not observe sex-dependent differences in the injured volume or cell death density. Our data show that sex differences in the acute microglial and immune responses to neonatal ischemia are likely both gene- and region-specific.


Subject(s)
Brain Ischemia/immunology , Immunity, Innate/genetics , Inflammation/immunology , Stroke/immunology , Animals , Animals, Newborn/immunology , Brain/immunology , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery , Inflammation/genetics , Inflammation/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Sex Characteristics , Stroke/genetics , Stroke/pathology
9.
Proc Natl Acad Sci U S A ; 110(44): 17957-62, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24133137

ABSTRACT

Neutrophils play a major role in inflammatory responses and immune defense against pathogens. Even though expression of inhibitory receptors has been reported on neutrophils, their role remains poorly defined. Here we show that primary human neutrophils expressed immunoglobulin-like transcript 4 (ILT4) inhibitory receptor and that this expression was induced during differentiation of the myelomonoblast PLB-985 cell line into "neutrophil-like" cells. Functional assays indicated that human leukocyte antigen G, the preferred ligand of ILT4, inhibited the phagocytic function of neutrophils. ILT4 engagement also impaired reactive oxygen species production induced through CD32a and both receptors were found colocalized into neutrophil lipid rafts. Moreover, neutrophil degranulation induced through inflammatory stimuli increased ILT4 expression as a result of the rapid translocation of an intracellular pool to the cell surface. Consequently to this ILT4 up-regulation, the human leukocyte antigen G-mediated inhibition of neutrophil phagocytic function was enhanced. Finally, we found that ILT4 up-regulation induced on healthy donor neutrophils following stimulation was impaired in presence of plasma from patients with sepsis. Similarly, ILT4 up-regulation was inhibited in neutrophils from septic patients. Altogether, our results reveal a unique mechanism of regulation of neutrophil functions through ILT4 and its exocytosis that may have implications in inflammatory disorders.


Subject(s)
Exocytosis/immunology , Gene Expression Regulation/immunology , Membrane Glycoproteins/immunology , Neutrophils/immunology , Receptors, Immunologic/immunology , Respiratory Burst/immunology , Apoptosis , Flow Cytometry , Humans , Microscopy, Confocal , Phagocytosis/immunology , Reactive Oxygen Species/metabolism , Sepsis/immunology , Sepsis/metabolism
10.
Cytometry B Clin Cytom ; 106(1): 58-63, 2024 01.
Article in English | MEDLINE | ID: mdl-37702371

ABSTRACT

BACKGROUND: Monocyte (m)HLA-DR expression appears to be a potent marker of immunosuppression in critically ill patients. The persistence of low mHLA-DR expression is associated with an increased risk of nosocomial infections and mortality. To adapt this measurement to pediatric requirements and provide extensive 24/7 access, we have developed a whole blood no-lyse no-wash micromethod (MM) and compared it with the standardized method (SM). METHODS: mHLA-DR was quantified by flow cytometry using Quantibrite™ Anti-HLA-DR PE/Monocyte PerCP-Cy™5.5 with either the SM performed in a diagnostic hematology laboratory using manufacturer protocol, or a whole blood no-lyse no-wash MM using an Attune flow cytometer located in the pediatric ICU. Median fluorescence intensity was measured in both techniques and converted to antibodies per cell (AB/C) calibrated with BD Quantibrite™ PE beads. Blood and Quantibrite™ reagent volume used with the MM was reduced by 5-fold compared to SM. In addition to Quantibrite™ Anti-Human HLA-DR PE/Monocyte PerCP-Cy™5.5, MM required anti-CD45 and anti-CD19 labeling. RESULTS: We determined the expression of mHLA-DR in 34 patients, 20 adults, and 14 children admitted to ICU. Correlation between MM and SM was excellent (Pearson's correlation: y = 0.8192x + 678.7, r = 0.9270, p < 0.0001). The estimated bias was 2467 ± 1.96 × 3307 AB/C; CI 95% [-4016; +8949]. CONCLUSIONS: The no-lyse no-wash whole blood microvolume method for measuring mHLA-DR expression allows for simplified sample preparation without compromising accuracy of the data. This method may simplify immune monitoring of critically ill patient by the deployment of a point of care method.


Subject(s)
Critical Illness , Monocytes , Adult , Humans , Child , Monocytes/metabolism , Flow Cytometry , HLA-DR Antigens , Immune Tolerance , Antibodies/metabolism
11.
J Vis Exp ; (185)2022 07 25.
Article in English | MEDLINE | ID: mdl-35938804

ABSTRACT

Microglia, as brain resident macrophages, are fundamental to several functions, including response to environmental stress and brain homeostasis. Microglia can adopt a large spectrum of activation phenotypes. Moreover, microglia that endorse pro-inflammatory phenotype is associated with both neurodevelopmental and neurodegenerative disorders. In vitro studies are widely used in research to evaluate potential therapeutic strategies in specific cell types. In this context studying microglial activation and neuroinflammation in vitro using primary microglial cultures is more relevant than microglial cell lines or stem-cell-derived microglia. However, the use of some primary cultures might suffer from a lack of reproducibility. This protocol proposes a reproducible and relevant method of magnetically isolating microglia from neonate pups. Microglial activation using several stimuli after 4 h and 24 h by mRNA expression quantification and a Cy3-bead phagocytic assay is demonstrated here. The current work is expected to provide an easily reproducible technique for isolating physiologically relevant microglia from juvenile developmental stages.


Subject(s)
Brain , Microglia , Animals , Magnetic Phenomena , Mice , Primary Cell Culture , Reproducibility of Results
12.
iScience ; 25(11): 105291, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36304101

ABSTRACT

Low monocyte (m)HLA-DR expression is associated with mortality in sepsis. G-286A∗rs3087456 polymorphism in promoter III of HLA class II transactivator (CIITA), the master regulator of HLA, has been associated with autoimmune diseases but its role in sepsis has never been demonstrated. In 203 patients in septic shock, GG genotype was associated with 28-day mortality and mHLA-DR remained low whereas it increased in patients with AA or AG genotype. In ex vivo cells, mHLA-DR failed to augment in GG in comparison with AG or AA genotype on exposure to IFN-γ. Promoter III transcript levels were similar in control monocytes regardless of genotype and exposure to IFN-γ. Promoter III activity was decreased in GG genotype in monocyte cell line but restored after stimulation with IFN-γ. Hereby, we demonstrated that G-286A∗rs3087456 significantly impact mHLA-DR expression in patients with septic shock in part through CIITA promoter III activity, that can be rescued using IFN-γ.

13.
Biochem Pharmacol ; 186: 114461, 2021 04.
Article in English | MEDLINE | ID: mdl-33571501

ABSTRACT

Acquired perinatal brain injuries are a set of conditions that remains a key challenge for neonatologists and that have significant social, emotional and financial implications for our communities. In our perspective article, we will introduce perinatal brain injury focusing specifically on the events leading to brain damage in preterm born infants and outcomes for these infants. Then we will summarize and discuss the preclinical and clinical studies testing the efficacy of stem cells as neuroprotectants in the last ten years in perinatal brain injury. There are no therapies to treat brain damage in preterm born infants and a primary finding from this review is that there is a scarcity of stem cell trials focused on overcoming brain injuries in these infants. Overall, across all forms of perinatal brain injury there is a remarkable heterogeneity in previous and on-going preclinical and clinical studies in terms of the stem cell type, animal models/patient selection, route and time of administration. Despite the quality of many of the studies this variation makes it difficult to reach a valid consensus for future developments. However, it is clear that stem cells (and stem cell derived exosomes) can reduce perinatal brain injury and our field needs to work collectively to refine an effective protocol for each type of injury. The use of standardized stem cell products and testing these products across multiple models of injury will provide a stronger framework for clinical trials development.


Subject(s)
Brain Injuries/therapy , Clinical Trials as Topic/methods , Disease Models, Animal , Infant, Premature/growth & development , Stem Cell Transplantation/methods , Animals , Brain Injuries/immunology , Brain Injuries/pathology , Cord Blood Stem Cell Transplantation/methods , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant, Newborn , Pregnancy , Stem Cells/immunology
14.
Nutrients ; 13(11)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34835975

ABSTRACT

Microglial activation is a key modulator of brain vulnerability in response to intra-uterine growth restriction (IUGR). However, the consequences of IUGR on microglial development and the microglial proteome are still unknown. We used a model of IUGR induced by a gestational low-protein diet (LPD) in rats. Microglia, isolated from control and growth-restricted animals at P1 and P4, showed significant changes in the proteome between the two groups. The expression of protein sets associated with fetal growth, inflammation, and the immune response were significantly enriched in LPD microglia at P1 and P4. Interestingly, upregulation of protein sets associated with the oxidative stress response and reactive oxygen species production was observed at P4 but not P1. During development, inflammation-associated proteins were upregulated between P1 and P4 in both control and LPD microglia. By contrast, proteins associated with DNA repair and senescence pathways were upregulated in only LPD microglia. Similarly, protein sets involved in protein retrograde transport were significantly downregulated in only LPD microglia. Overall, these data demonstrate significant and multiple effects of LPD-induced IUGR on the developmental program of microglial cells, leading to an abnormal proteome within the first postnatal days.


Subject(s)
Fetal Growth Retardation/metabolism , Microglia/metabolism , Proteome/metabolism , Animals , Animals, Newborn , Body Weight , Cluster Analysis , Diet, Protein-Restricted , Inflammation/pathology , Oxidative Stress , Rats, Sprague-Dawley
15.
Cell Death Dis ; 12(2): 166, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558485

ABSTRACT

A leading cause of preterm birth is the exposure to systemic inflammation (maternal/fetal infection), which leads to neuroinflammation and white matter injury (WMI). A wide range of cytokines and chemokines are expressed and upregulated in oligodendrocytes (OLs) in response to inflammation and numerous reports show that OLs express several receptors for immune related molecules, which enable them to sense inflammation and to react. However, the role of OL immune response in WMI is unclear. Here, we focus our study on toll-like receptor-3 (TLR3) that is activated by double-strand RNA (dsRNA) and promotes neuroinflammation. Despite its importance, its expression and role in OLs remain unclear. We used an in vivo mouse model, which mimics inflammation-mediated WMI of preterm born infants consisting of intraperitoneal injection of IL-1ß from P1 to P5. In the IL-1ß-treated animals, we observed the upregulation of Tlr3, IL-1ß, IFN-ß, Ccl2, and Cxcl10 in both PDGFRα+ and O4+ sorted cells. This upregulation was higher in O4+ immature OLs (immOLs) as compared to PDGFRα+ OL precursor cells (OPCs), suggesting a different sensitivity to neuroinflammation. These observations were confirmed in OL primary cultures: cells treated with TLR3 agonist Poly(I:C) during differentiation showed a stronger upregulation of Ccl2 and Cxcl10 compared to cells treated during proliferation and led to decreased expression of myelin genes. Finally, OLs were able to modulate microglia phenotype and function depending on their maturation state as assessed by qPCR using validated markers for immunomodulatory, proinflammatory, and anti-inflammatory phenotypes and by phagocytosis and morphological analysis. These results show that during inflammation the response of OLs can play an autonomous role in blocking their own differentiation: in addition, the immune activation of OLs may play an important role in shaping the response of microglia during inflammation.


Subject(s)
Cell Differentiation , Cell Proliferation , Encephalitis/metabolism , Leukoencephalopathies/metabolism , Oligodendroglia/metabolism , Toll-Like Receptor 3/metabolism , White Matter/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Encephalitis/pathology , Female , Inflammation Mediators/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/immunology , Leukoencephalopathies/pathology , Male , Mice , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Oligodendroglia/drug effects , Oligodendroglia/immunology , Oligodendroglia/pathology , Poly I-C/pharmacology , Pregnancy , Premature Birth , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Toll-Like Receptor 3/agonists , White Matter/drug effects , White Matter/immunology , White Matter/pathology
16.
Cell Rep ; 31(2): 107506, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294449

ABSTRACT

A distinctive feature of neocortical development is the highly coordinated production of different progenitor cell subtypes, which are critical for ensuring adequate neurogenic outcome and the development of normal neocortical size. To further understand the mechanisms that underlie neocortical growth, we focused our studies on the microcephaly gene Mcph1, and we report here that Mcph1 (1) exerts its functions in rapidly dividing apical radial glial cells (aRGCs) during mouse neocortical development stages that precede indirect neurogenesis; (2) is expressed at mitochondria; and (3) controls the proper proliferation and survival of RGCs, potentially through crosstalk with cellular metabolic pathways involving the stimulation of mitochondrial activity via VDAC1/GRP75 and AKT/HK2/VDAC1 and glutaminolysis via ATF4/PCK2. We currently report the description of a MCPH-gene implication in the interplay between bioenergetic pathways and neocortical growth, thus pointing to alterations of cellular metabolic pathways, in particular glutaminolysis, as a possible cause of microcephalic pathogenesis.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Microcephaly/genetics , Microcephaly/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cytoskeletal Proteins/metabolism , Female , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microcephaly/physiopathology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neuroglia/metabolism , Neurons/metabolism , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
17.
Crit Care Med ; 37(10): 2746-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19707128

ABSTRACT

OBJECTIVES: To test early measurement of human leukocyte antigen-DR expression on circulating monocytes (mHLA-DR) as prognostic marker, and the trend of mHLA-DR recovery for the prediction of late secondary infection risk in a large intensive care unit population. DESIGN: Prospective, observational study over 16 mos. SETTING: Intensive care unit in a tertiary teaching hospital. INCLUSION CRITERIA: Simplified Acute Physiology Score II >15, age >18 yrs. MEASUREMENTS AND MAIN RESULTS: The mHLA-DR was measured by flow cytometry within the first 3 days and twice a week until discharge. We used a logistic regression model for outcome prediction, and a competing risk approach to test the relationship between mHLA-DR recovery (log (mHLA-DR) slope) and incidence of secondary infection. A total of 283 consecutive patients suffering from various pathologies were monitored (Simplified Acute Physiology Score II = 39, Sepsis-related Organ Failure Assessment of 5 on day 0). Early mHLA-DR was decreased in the whole population, however, more deeply in sepsis (p < .0001). Low mHLA-DR was associated with mortality in the whole population (p = .003), as in subgroups (nonseptic, neurologic, and septic), but not when adjusted on Simplified Acute Physiology Score II. In patients with a length of stay of >7 days (n = 70), the lower the slope of mHLA-DR recovery, the higher the incidence of the first secondary infection (adjusted on early mHLA-DR, p = .04). CONCLUSIONS: For a given severity, mHLA-DR proved not to a predictive marker of outcome, but a weak trend of mHLA-DR recovery was associated with an increased risk of secondary infection. Monitoring immune functions through mHLA-DR in intensive care unit patients therefore could be useful to identify a high risk of secondary infection.


Subject(s)
Bacterial Infections/immunology , Cross Infection/immunology , HLA-DR Antigens/blood , Intensive Care Units , Monocytes/immunology , Shock, Septic/immunology , Systemic Inflammatory Response Syndrome/immunology , Adult , Aged , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/mortality , Bacterial Infections/therapy , Cross Infection/mortality , Cross Infection/therapy , Drug Therapy, Combination , Female , Hospital Mortality , Humans , Immune Tolerance/immunology , Immunization, Passive , Length of Stay/statistics & numerical data , Male , Middle Aged , Multiple Organ Failure/immunology , Multiple Organ Failure/mortality , Multiple Organ Failure/therapy , Pneumonia, Ventilator-Associated/immunology , Pneumonia, Ventilator-Associated/mortality , Predictive Value of Tests , Prognosis , Reference Values , Risk Factors , Shock, Septic/mortality , Shock, Septic/therapy , Systemic Inflammatory Response Syndrome/mortality , Systemic Inflammatory Response Syndrome/therapy
18.
Sci Rep ; 9(1): 15660, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666644

ABSTRACT

Biomarkers in sepsis for severity, prediction of outcome or reversibility of organ dysfunction are warranted. Measurements of plasma DAMP levels at admission can reflect the severity of cellular damage in septic shock, which might predict the prognosis and reduce the risk of overtreating patients with costly therapies. We measured plasma levels of two DAMPs, S100A8/S100A9 and S100A12 during the first 24 h of admission of septic shock patients. Forty-nine septic shock patients with a similar SOFA scores were selected from our sepsis database to compare a similar proportion of survivors and non-survivors. Plasma levels of S100A8/S100A9 and S100A12 were compared with healthy volunteers using in-house ELISA. Plasma levels of S100A8/S100A9 and S100A12 (5.71 [2.60-13.63] µg/mL and 0.48 [0.22-1.05] µg/mL) were higher in septic shock patients than in healthy volunteers (1.18 [0.74-1.93] µg/mL and 0.09 [0.02-0.39] µg/mL) (P < 0.0001 and P = 0.0030). Levels of S100A8/S100A9 and S100A12 in non-survivors at day 28 (11.70 [2.85-24.36] µg/mL and 0.62 [0.30-1.64] µg/mL) were significantly higher than in survivors (4.59 [2.16-7.47] µg/mL and 0.30 [0.20-0.49] µg/mL) (P = 0.0420 and P = 0.0248) and correlated well (Spearman r = 0.879, P < 0.0001). The high level of plasma calgranulins at admission in septic shock, were higher in non-survivors compared to survivors. These markers could indicate a higher risk of death when SOFA scores are similar and help the stratification of patients for improved care and therapy selection.


Subject(s)
Calgranulin A/blood , Calgranulin B/blood , Patient Admission , S100A12 Protein/blood , Shock, Septic/blood , Shock, Septic/mortality , Adult , Aged , Female , Humans , Leukocyte L1 Antigen Complex/blood , Male , Middle Aged , Prognosis , Risk , Shock, Septic/diagnosis , Survival Analysis
19.
Intensive Care Med ; 34(7): 1313-20, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18365175

ABSTRACT

OBJECTIVE: Arginine vasopressin (AVP) is being used increasingly to treat vasodilatory hypotension, although its effects on hepatosplanchnic perfusion have been debated. DESIGN: Prospective study in a university-based experimental research laboratory. SUBJECTS AND INTERVENTIONS: We compared the effect of AVP on systemic, gut, and liver blood flow in anesthetized and ventilated rabbits given either saline or endotoxin. Incremental i.v. boluses of AVP ranging from 1 to 1,000[Symbol: see text]ng were administered 90[Symbol: see text]min post-endotoxin or saline. MEASUREMENTS AND RESULTS: Endotoxin induced a shock state with a transient decrease of mesenteric artery blood flow velocity (pulsed Doppler, in centimeters per second, V(mes)) but had no effect on liver surface microcirculation (laser Doppler in TPU, MicroFl(liver)). Gut microcirculatory (MicroFl(gut)) changes became independent of mean arterial pressure (MAP) after endotoxin. In control rabbits (n = 5), increasing doses of AVP elevated MAP but reduced aortic blood flow (pulsed Doppler, VAo), V(mes), and MicroFl(gut) (p < 0.05). In endotoxic animals (n = 6), AVP produced a similar rise in MAP (p < 0.05), while V(mes) and MicroFl(gut) only decreased for AVP doses above 100[Symbol: see text]ng (p < 0.05). Liver microcirculation was only minimally affected by AVP, although significantly, both in control and endotoxin animals. CONCLUSION: Preservation of mesenteric blood flow as well as gut and liver microcirculation, with therapeutic doses of AVP during endotoxemia, supports its use as a hemodynamic agent during septic shock.


Subject(s)
Arginine Vasopressin/therapeutic use , Hypotension/drug therapy , Intestines/blood supply , Liver/blood supply , Microcirculation/drug effects , Splanchnic Circulation/drug effects , Vasoconstrictor Agents/therapeutic use , Animals , Arginine Vasopressin/pharmacology , Disease Models, Animal , Hemodynamics/drug effects , Rabbits , Shock, Septic/drug therapy , Vasoconstrictor Agents/pharmacology
20.
Intensive Care Med ; 34(8): 1371-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18392609

ABSTRACT

OBJECTIVE: To assess blood leucocytes gene profiling during recovery phase of septic shock; to test the relation between encoding gene expression and protein level. STUDY DESIGN: Gene expression levels were studied at days 0, 1, 7 and 28 (D0, 1, 7 and 28) on a dedicated microarray of 340 genes involved in inflammatory processes. SETTINGS: 16-bed intensive care unit, Lariboisière University hospital. PATIENTS: Seventeen septic shock patients enrolled when at least one additional organ dysfunction occurred. MEASUREMENTS AND RESULTS: Changes over time were compared with D0 via the ratio Dx/D0. The time-related gene expression study showed significant changes in ten genes. Among them, S100A8 and S100A12 had a reduced expression over time compared with D0, whereas CD74's expression increased. The microarray results were validated by RT-qPCR for four genes. The S100A8 plasma levels decrease along recovery in parallel with the gene expression decrease. The CD74 gene expression evolution significantly correlated with HLA-DR monocyte expression. CONCLUSIONS: These results are the first description of variations in expression of key inflammatory genes in the course of the septic shock recovery period.


Subject(s)
Calgranulin A/blood , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Shock, Septic/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Calgranulin A/genetics , Calgranulin A/physiology , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Histocompatibility Antigens Class II/genetics , Humans , Leukocytes , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Shock, Septic/mortality , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL