Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 374: 114703, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38281588

ABSTRACT

Germinal matrix hemorrhage (GMH) is a devasting neurological disease in premature newborns. After GMH, brain iron overload associated with hemoglobin degradation contributed to oxidative stress, causing disruption of the already vulnerable blood-brain barrier (BBB). Mitochondrial ferritin (FTMT), a novel mitochondrial outer membrane protein, is crucial in maintaining cellular iron homeostasis. We aimed to investigate the effect of FTMT upregulation on oxidative stress and BBB disruption associated with brain iron overload in rats. A total of 222 Sprague-Dawley neonatal rat pups (7 days old) were used to establish a collagenase-induced GMH model and an iron-overload model of intracerebral FeCl2 injection. Deferiprone was administered via gastric lavage 1 h after GMH and given daily until euthanasia. FTMT CRISPR Knockout and adenovirus (Ad)-FTMT were administered intracerebroventricularly 48 h before GMH and FeCl2 injection, respectively. Neurobehavioral tests, immunofluorescence, Western blot, Malondialdehyde measurement, and brain water content were performed to evaluate neurobehavior deficits, oxidative stress, and BBB disruption, respectively. The results demonstrated that brain expressions of iron exporter Ferroportin (FPN) and antioxidant glutathione peroxidase 4 (GPX4) as well as BBB tight junction proteins including Claudin-5 and Zona Occulta (ZO)-1 were found to be decreased at 72 h after GMH. FTMT agonist Deferiprone attenuated oxidative stress and preserved BBB tight junction proteins after GMH. These effects were partially reversed by FTMT CRISPR Knockout. Iron overload by FeCl2 injection resulted in oxidative stress and BBB disruption, which were improved by Ad-FTMT mediated FTMT overexpression. Collectively, FTMT upregulation is neuroprotective against brain injury associated with iron overload. Deferiprone reduced oxidative stress and BBB disruption by maintaining cellular iron homeostasis partially by the upregulating of FTMT after GMH. Deferiprone may be an effective treatment for patients with GMH.


Subject(s)
Blood-Brain Barrier , Iron Overload , Humans , Infant, Newborn , Rats , Animals , Blood-Brain Barrier/metabolism , Animals, Newborn , Rats, Sprague-Dawley , Up-Regulation , Deferiprone/metabolism , Deferiprone/pharmacology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Oxidative Stress , Iron/metabolism , Iron Overload/metabolism , Homeostasis , Ferritins/metabolism , Tight Junction Proteins/metabolism
2.
Exp Neurol ; 372: 114615, 2024 02.
Article in English | MEDLINE | ID: mdl-37995951

ABSTRACT

BACKGROUND: Activation of mast cells plays an important role in brain inflammation. CD300a, an inhibitory receptor located on mast cell surfaces, has been reported to reduce the production of pro-inflammatory cytokines and exert protective effects in inflammation-related diseases. Peroxisome proliferator-activated receptor ß/δ (PPARß/δ), a ligand-activated nuclear receptor, activation upregulates the transcription of CD300a. In this study, we aim to investigate the role of PPARß/δ in the attenuation of germinal matrix hemorrhage (GMH)-induced mast cell activation via CD300a/SHP1 pathway. METHODS: GMH model was induced by intraparenchymal injection of bacterial collagenase into the right hemispheric ganglionic eminence in P7 Sprague Dawley rats. GW0742, a PPARß/δ agonist, was administered intranasally at 1 h post-ictus. CD300a small interfering RNA (siRNA) and PPARß/δ siRNA were injected intracerebroventricularly 5 days and 2 days before GMH induction. Behavioral tests, Western blot, immunofluorescence, Toluidine Blue staining, and Nissl staining were applied to assess post-GMH evaluation. RESULTS: Results demonstrated that endogenous protein levels of PPARß/δ and CD300a were decreased, whereas chymase, tryptase, IL-17A and transforming growth factor ß1 (TGF-ß1) were elevated after GMH. GMH induced significant short- and long-term neurobehavioral deficits in rat pups. GW0742 decreased mast cell degranulation, improved neurological outcomes, and attenuated ventriculomegaly after GMH. Additionally, GW0742 increased expression of PPARß/δ, CD300a and phosphorylation of SHP1, decreased phosphorylation of Syk, chymase, tryptase, IL-17A and TGF-ß1 levels. PPARß/δ siRNA and CD300a siRNA abolished the beneficial effects of GW0742. CONCLUSIONS: GW0742 inhibited mast cell-induced inflammation and improved neurobehavior after GMH, which is mediated by PPARß/δ/CD300a/SHP1 pathway. GW0742 may serve as a potential treatment to reduce brain injury for GMH patients.


Subject(s)
PPAR delta , PPAR-beta , Humans , Rats , Animals , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Animals, Newborn , Mast Cells/metabolism , Chymases , Interleukin-17 , Rats, Sprague-Dawley , Transforming Growth Factor beta1 , Tryptases , Cerebral Hemorrhage , Thiazoles/pharmacology , Inflammation , RNA, Small Interfering
3.
Stroke ; 54(9): 2420-2433, 2023 09.
Article in English | MEDLINE | ID: mdl-37465997

ABSTRACT

BACKGROUND: Hematoma clearance has been a proposed therapeutic strategy for hemorrhagic stroke. This study investigated the impact of CX3CR1 (CX3C chemokine receptor 1) activation mediated by r-FKN (recombinant fractalkine) on hematoma resolution, neuroinflammation, and the underlying mechanisms involving AMPK (AMP-activated protein kinase)/PPARγ (peroxisome proliferator-activated receptor gamma) pathway after experimental germinal matrix hemorrhage (GMH). METHODS: A total of 313 postnatal day 7 Sprague Dawley rat pups were used. GMH was induced using bacterial collagenase by a stereotactically guided infusion. r-FKN was administered intranasally at 1, 25, and 49 hours after GMH for short-term neurological evaluation. Long-term neurobehavioral tests (water maze, rotarod, and foot-fault test) were performed 24 to 28 days after GMH with the treatment of r-FKN once daily for 7 days. To elucidate the underlying mechanism, CX3CR1 CRISPR, or selective CX3CR1 inhibitor AZD8797, was administered intracerebroventricularly 24 hours preinduction of GMH. Selective inhibition of AMPK/PPARγ signaling in microglia via intracerebroventricularly delivery of liposome-encapsulated specific AMPK (Lipo-Dorsomorphin), PPARγ (Lipo-GW9662) inhibitor. Western blot, Immunofluorescence staining, Nissl staining, Hemoglobin assay, and ELISA assay were performed. RESULTS: The brain expression of FKN and CX3CR1 were elevated after GMH. FKN was expressed on both neurons and microglia, whereas CX3CR1 was mainly expressed on microglia after GMH. Intranasal administration of r-FKN improved the short- and long-term neurobehavioral deficits and promoted M2 microglia polarization, thereby attenuating neuroinflammation and enhancing hematoma clearance, which was accompanied by an increased ratio of p-AMPK (phosphorylation of AMPK)/AMPK, Nrf2 (nuclear factor erythroid 2-related factor 2), PPARγ, CD36 (cluster of differentiation 36), CD163 (hemoglobin scavenger receptor), CD206 (the mannose receptor), and IL (interleukin)-10 expression, and decreased CD68 (cluster of differentiation 68), IL-1ß, and TNF (tumor necrosis factor) α expression. The administration of CX3CR1 CRISPR or CX3CR1 inhibitor (AZD8797) abolished the protective effect of FKN. Furthermore, selective inhibition of microglial AMPK/PPARγ signaling abrogated the anti-inflammation effects of r-FKN after GMH. CONCLUSIONS: CX3CR1 activation by r-FKN promoted hematoma resolution, attenuated neuroinflammation, and neurological deficits partially through the AMPK/PPARγ signaling pathway, which promoted M1/M2 microglial polarization. Activating CX3CR1 by r-FKN may provide a promising therapeutic approach for treating patients with GMH.


Subject(s)
Chemokine CX3CL1 , Infant, Newborn, Diseases , Rats , Animals , Humans , Infant, Newborn , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/pharmacology , PPAR gamma/metabolism , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Rats, Sprague-Dawley , Neuroinflammatory Diseases , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Microglia/metabolism , Hematoma/metabolism , CX3C Chemokine Receptor 1/metabolism
4.
J Correct Health Care ; 29(2): 135-142, 2023 04.
Article in English | MEDLINE | ID: mdl-36930850

ABSTRACT

In using an approach encompassing intersectionality and interconnectedness, we highlight how the experiences of Indigenous mothers and mother figures in contact with the law are a result of various historical and contemporary events. We highlight a need for a wholistic approach to eliminate the overrepresentation of Indigenous Peoples, including parents, in the criminal justice system. There is a lack of research and discussion on Indigenous women and their experience with the justice system and by using an Indigenous lens, we can explore the establishment of culturally safe resources and care wherein gender inclusivity is prioritized. Our team of researchers and advocates intends for this article to contribute and spark dialogue on Indigenous Peoples, particularly mothers and mother figures and their interactions with the justice system. Although this article mainly focuses on federal programs and policies in Canada, insights on the barriers to care can be applied into policy and practice across multiple settler states.


Subject(s)
Criminal Law , Mothers , Humans , Female , Canada , Population Groups
5.
Exp Neurol ; 360: 114276, 2023 02.
Article in English | MEDLINE | ID: mdl-36402169

ABSTRACT

AIMS: Germinal matrix hemorrhage (GMH) is a disastrous clinical event for newborns. Neuroinflammation plays an important role in the development of neurological deficits after GMH. The purpose of this study is to investigate the anti-inflammatory role of secukinumab after GMH and its underlying mechanisms involving PKCß/ERK/NF-κB signaling pathway. METHODS: A total of 154 Sprague-Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of bacterial collagenase. Secukinumab was administered intranasally post-GMH. PKCß activator PMA and p-ERK activator Ceramide C6 were administered intracerebroventricularly at 24 h prior to GMH induction, respectively. Neurobehavioral tests, western blot and immunohistochemistry were used to evaluate the efficacy of Secukinumab in both short-term and long-term studies. RESULTS: Endogenous IL-17A, IL-17RA, PKCß and p-ERK were increased after GMH. Secukinumab treatment improved short- and long-term neurological outcomes, reduced the synthesis of MPO and Iba-1 in the perihematoma area, and inhibited the synthesis of proinflammatory factors, such as NF-κB, IL-1ß, TNF-α and IL-6. Additionally, PMA and ceramide C6 abolished the beneficial effects of Secukinumab. CONCLUSION: Secukinumab treatment suppressed neuroinflammation and attenuated neurological deficits after GMH, which was mediated through the downregulation of the PKCß/ERK/NF-κB pathway. Secukinumab treatment may provide a promising therapeutic strategy for GMH patients.


Subject(s)
NF-kappa B , Neuroinflammatory Diseases , Animals , Rats , Rats, Sprague-Dawley , Animals, Newborn , Cerebral Hemorrhage/metabolism
6.
Exp Neurol ; 359: 114257, 2023 01.
Article in English | MEDLINE | ID: mdl-36279933

ABSTRACT

Germinal matrix hemorrhage (GMH) is one of the leading causes of morbidity and mortality in preterm infants in the United States, with little progress made in its clinical management. Blood clots disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage are key contributors towards post-hemorrhagic hydrocephalus development. n-formyl peptide receptor 2 (FPR2), a G-protein-coupled receptor, has been associated with the activation of p-ERK1/2, which in turn promotes the transcription of the DUSP1 gene, which may play a role in CD36 signaling. CD36 scavenger, a transmembrane glycoprotein, plays an essential role in microglia phagocytic blood clot clearance after GMH. FPR2's role in blood clot clearance after hemorrhagic stroke is unknown. We hypothesize that FPR2 activation by FPR2 agonist Annexin A1 (AnxA1) will enhance hematoma resolution via the upregulation of the CD36 signaling pathway, thereby improving short- and long-term neurological outcomes. Bacterial collagenase (0.3 U) was infused intraparenchymally into the right hemispheric ganglionic eminence in P7 rat pups to induce GMH. AnxA1 and FPR2 Inhibitor (Boc2) were given at 1-h post-GMH via intranasal administration. FPR2 CRISPR was given 48-h prior to GMH induction. Short-term neurological deficits were assessed using negative geotaxis test. Hematoma volume was assessed using hemoglobin assay. Protein expression was assessed using western blots. Long-term neurocognitive deficits and motor coordination were assessed using Morris water maze, rotarod, and foot fault tests. We have demonstrated that AnxA1 treatment enhances hematoma resolution and improved short and long-term outcomes. Lastly, FPR2 agonist AnxA1 treatment resulted in the upregulation of the FPR2/p-ERK(1/2)/DUSP1/CD36 signaling pathway.


Subject(s)
Annexin A1 , Receptors, Formyl Peptide , Animals , Humans , Infant, Newborn , Rats , Annexin A1/genetics , Annexin A1/metabolism , CD36 Antigens/genetics , Cerebral Hemorrhage/complications , Dual Specificity Phosphatase 1/metabolism , Hematoma , Infant, Premature , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Signal Transduction , Extracellular Signal-Regulated MAP Kinases
7.
Front Sociol ; 7: 790397, 2022.
Article in English | MEDLINE | ID: mdl-35586263

ABSTRACT

This article explores the unique and understudied experiences of Indigenous women living in Toronto, Canada during the first year of the COVID-19 pandemic. The purpose of this study is to better document the impacts of COVID-19 on the mental health and wellbeing of Indigenous women in Toronto, Canada to better understand unmet needs, as well as lay the groundwork for more targeted research and potential interventions based on these needs. Using in-depth semi-structured interviews with thirteen Indigenous women, we shed light on the negative effects this pandemic has had on this population. We find that COVID-19 has negatively affected people's mental health, substance use and access to health services. This research speaks to the growing body of work that discusses the harmful effects of COVID-19 generally and how this pandemic has specifically affected Indigenous peoples.

8.
J Neuroinflammation ; 18(1): 160, 2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34275493

ABSTRACT

BACKGROUND: Germinal matrix hemorrhage (GMH) is defined by the rupture of immature blood vessels in the germinal matrix, where subsequent hemorrhage enters the subependymal zone and the cerebral lateral ventricles. The consequent blood clot has been identified as the causative factor of secondary brain injury, which triggers a series of complex parallel and sequential harmful mechanisms, including neuroinflammation. The orphan G-protein-coupled receptor 40 (GPR40), a free fatty acid (FFA) receptor 1, has been shown to exert anti-inflammatory effects when activated and improved outcomes in animal models of stroke. We aimed to investigate the anti-inflammatory effects of GPR40 and its underlying mechanisms after GMH. METHODS: GMH model was induced in 7-day-old rat pups by an intraparenchymal injection of bacterial collagenase. GPR40 agonist, GW9508, was administered intranasally 1 h, 25 h, and 49 h after GMH induction. CRISPR targeting GPR40, PAK4, and KDM6B were administered through intracerebroventricular injection 48 h before GMH induction. Neurologic scores, microglia polarization, and brain morphology were evaluated by negative geotaxis, right reflex, rotarod test, foot fault test, Morris water maze, immunofluorescence staining, Western blots, and nissl staining respectfully. RESULTS: The results demonstrated that GW9508 improved neurological and morphological outcomes after GMH in the short (24 h, 48 h, 72h) and long-term (days 21-27). However, the neuroprotective effects of treatment were abolished by GW1100, a selective GPR40 antagonist. GW9508 treatment increased populations of M2 microglia and decreased M1 microglia in periventricular areas 24 h after GMH induction. GW9508 upregulated the phosphorylation of PAK4, CREB, and protein level of KDM6B, CD206, IL-10, which was also met with the downregulation of inflammatory markers IL-1ß and TNF-α. The mechanism study demonstrated that the knockdown of GPR40, PAK4, and KDM6B reversed the neuroprotective effects brought on by GW9508. This evidence suggests that GPR40/PAK4/CREB/KDM6B signaling pathway in microglia plays a role in the attenuation of neuroinflammation after GMH. CONCLUSIONS: In conclusion, the present study demonstrates that the activation of GPR40 attenuated GMH-induced neuroinflammation through the activation of the PAK4/CREB/KDM6B signaling pathway, and M2 microglia may be a major mediator of this effect. Thus, GPR40 may serve as a potential target in the reduction of the inflammatory response following GMH, thereby improving neurological outcomes in the short- and long-term.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Microglia/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Animals, Newborn , Behavior Rating Scale , Cerebral Hemorrhage/immunology , Cytokines , Disease Models, Animal , Fluorescent Antibody Technique , Jumonji Domain-Containing Histone Demethylases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
9.
Oxid Med Cell Longev ; 2021: 5913424, 2021.
Article in English | MEDLINE | ID: mdl-33532035

ABSTRACT

AIMS: Blood clots play the primary role in neurological deficits after germinal matrix hemorrhage (GMH). Previous studies have shown a beneficial effect in blood clot clearance after hemorrhagic stroke. The purpose of this study is to investigate interleukin-19's role in hematoma clearance after GMH and its underlying mechanism of IL-20R1/ERK/Nrf2 signaling pathway. METHODS: A total of 240 Sprague-Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of bacterial collagenase. rIL-19 was administered intranasally 1 hour post-GMH. IL-20R1 CRISPR was administered intracerebroventricularly, or Nrf2 antagonist ML385 was administered intraperitoneally 48 hours and 1 hour before GMH induction, respectively. Neurobehavior, Western blot, immunohistochemistry, histology, and hemoglobin assay were used to evaluate treatment regiments in the short- and long-term. RESULTS: Endogenous IL-19, IL-20R1, IL-20R2, and scavenger receptor CD163 were increased after GMH. rIL-19 treatment improved neurological deficits, reduced hematoma volume and hemoglobin content, reduced ventriculomegaly, and attenuated cortical thickness loss. Additionally, treatment increased ERK, Nrf2, and CD163 expression, whereas IL-20R1 CRISPR-knockdown plasmid and ML385 inhibited the effects of rIL-19 on CD163 expression. CONCLUSION: rIL-19 treatment improved hematoma clearance and attenuated neurological deficits induced by GMH, which was mediated through the upregulation of the IL-20R1/ERK/Nrf2 pathways. rIL-19 treatment may provide a promising therapeutic strategy for the GMH patient population.


Subject(s)
Cerebral Hemorrhage/drug therapy , Interleukins/therapeutic use , Receptors, Interleukin/agonists , Animals , Animals, Newborn , Cerebral Hemorrhage/congenital , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Hematoma/congenital , Hematoma/drug therapy , Hematoma/metabolism , Hematoma/pathology , Interleukins/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , NF-E2-Related Factor 2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin/metabolism , Recombinant Proteins/pharmacology , Remission Induction
10.
Cult Health Sex ; 23(9): 1215-1226, 2021 09.
Article in English | MEDLINE | ID: mdl-32701035

ABSTRACT

This paper examines the experiences of girls who engage in intimate partner relationships behind bars and describes how institutional actors respond to these partnerships. Current research on sexuality in prisons and detention centres largely focuses on the experiences of men. Those studies that focus on women largely ignore the experiences of underage women. Moreover, current theoretical and empirical work in this area demonstrates how monitoring sexuality is a form of social control and is largely punished behind bars. We discuss how this process works and how girls negotiate relationships in one California Juvenile Detention Centre, drawing on two years of ethnographic research and interviews with incarcerated women. We find that correctional staff often ignore sexual relationships behind bars and provide little or no sexuality and sexual health education. Additionally, we discuss the benefits and challenges to girls participating in relationships in secure confinement. Our work contributes to the small but growing body of research on sexuality behind bars.


Subject(s)
Jails , Sexual Partners , Female , Humans , Male , Prisons , Sexual Behavior , Sexuality
11.
J Neuroinflammation ; 17(1): 250, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32859236

ABSTRACT

BACKGROUND: Mast cells play an important role in early immune reactions in the brain by degranulation and the consequent inflammatory response. Our aim of the study is to investigate the effects of rh-relaxin-2 on mast cells and the underlying mechanisms in a germinal matrix hemorrhage (GMH) rat model. METHODS: One hundred seventy-three P7 rat pups were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Clodronate liposome was administered through intracerebroventricular (i.c.v.) injections 24 h prior to GMH to inhibit microglia. Rh-relaxin-2 was administered intraperitoneally at 1 h and 13 h after GMH. Small interfering RNA of RXFP1 and PI3K inhibitor LY294002 were given by i.c.v. injection. Post-GMH evaluation included neurobehavioral function, Western blot analysis, immunofluorescence, Nissl staining, and toluidine blue staining. RESULTS: Our results demonstrated that endogenous relaxin-2 was downregulated and that RXFP1 level peaked on the first day after GMH. Administration of rh-relaxin-2 improved neurological functions, attenuated degranulation of mast cells and neuroinflammation, and ameliorated post-hemorrhagic hydrocephalus (PHH) after GMH. These effects were associated with RXFP1 activation, increased expression of PI3K, phosphorylated AKT and TNFAIP3, and decreased levels of phosphorylated NF-κB, tryptase, chymase, IL-6, and TNF-α. However, knockdown of RXFP1 and PI3K inhibition abolished the protective effects of rh-relaxin-2. CONCLUSIONS: Our findings showed that rh-relaxin-2 attenuated degranulation of mast cells and neuroinflammation, improved neurological outcomes, and ameliorated hydrocephalus after GMH through RXFP1/PI3K-AKT/TNFAIP3/NF-κB signaling pathway.


Subject(s)
Intracranial Hemorrhages/metabolism , Mast Cells/drug effects , NF-kappa B/metabolism , Recombinant Proteins/pharmacology , Relaxin/pharmacology , Signal Transduction/drug effects , Animals , Disease Models, Animal , Mast Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
13.
J Neurosci Res ; 98(1): 105-120, 2020 01.
Article in English | MEDLINE | ID: mdl-30793349

ABSTRACT

In addition to being the leading cause of morbidity and mortality in premature infants, germinal matrix hemorrhage (GMH) is also the leading cause of acquired infantile hydrocephalus. The pathophysiology of posthemorrhagic hydrocephalus (PHH) development after GMH is complex and vaguely understood, although evidence suggests fibrosis and gliosis in the periventricular and subarachnoid spaces disrupts normal cerebrospinal fluid (CSF) dynamics. Theories explaining general hydrocephalus etiology have substantially evolved from the original bulk flow theory developed by Dr. Dandy over a century ago. Current clinical and experimental evidence supports a new hydrodynamic theory for hydrocephalus development involving redistribution of vascular pulsations and disruption of Starling forces in the brain microcirculation. In this review, we discuss CSF flow dynamics, history and development of theoretical hydrocephalus pathophysiology, and GMH epidemiology and etiology as it relates to PHH development. We highlight known mechanisms and propose new avenues that will further elucidate GMH pathophysiology, specifically related to hydrocephalus.


Subject(s)
Choroid Plexus/metabolism , Hydrocephalus/metabolism , Intracranial Hemorrhages/metabolism , Signal Transduction/physiology , Choroid Plexus/pathology , Humans , Hydrocephalus/etiology , Hydrocephalus/pathology , Infant , Infant, Newborn , Infant, Premature , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/pathology
14.
J Neurosci Res ; 98(1): 168-178, 2020 01.
Article in English | MEDLINE | ID: mdl-31157469

ABSTRACT

Spontaneous intracerebral hemorrhage (ICH) is the deadliest stroke subtype and neuroinflammation is a critical component of the pathogenesis following ICH. Annexin A1-FPR2 signaling has been shown to play a protective role in animal stroke models. This study aimed to assess whether Annexin A1 attenuated neuroinflammation and brain edema after ICH and investigate the underlying mechanisms. Male CD-1 mice were subjected to collagenase-induced ICH. Annexin A1 was administered at 0.5 hr after ICH. Brain water content measurement, short-term and long-term neurobehavioral tests, Western blot and immnunofluorescence were performed. Results showed that Annexin A1 effectively attenuated brain edema, improved short-term neurological function and ameliorated microglia activation after ICH. Annexin A1 also improved memory function at 28 days after ICH. However, these beneficial effects were abolished with the administration of FPR2 antagonist Boc-2. Furthermore, AnxA1/FPR2 signaling may confer protective effects via inhibiting p38-associated inflammatory cascade. Our study demonstrated that Annexin A1/FPR2/p38 signaling pathway played an important role in attenuating neuroinflammation after ICH and that Annexin A1 could be a potential therapeutic strategy for ICH patients.


Subject(s)
Annexin A1/pharmacology , Brain Edema/drug therapy , Cerebral Hemorrhage/drug therapy , Cyclooxygenase 2/metabolism , Receptors, Formyl Peptide/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Annexin A1/therapeutic use , Brain/drug effects , Brain/metabolism , Brain Edema/etiology , Brain Edema/metabolism , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/metabolism , Collagenases , Disease Models, Animal , Male , Maze Learning/drug effects , Mice , Neurons/drug effects , Neurons/metabolism
15.
J Neurosci Res ; 98(1): 121-128, 2020 01.
Article in English | MEDLINE | ID: mdl-30667078

ABSTRACT

Currently, there is no effective treatment for germinal matrix hemorrhage and intraventricular hemorrhage (GMH-IVH), a common and often fatal stroke subtype in premature infants. Secondary brain injury after GMH-IVH is known to involve blood clots that contribute to inflammation and neurological deficits. Furthermore, the subsequent blood clots disrupt normal cerebrospinal fluid circulation and absorption after GMH-IVH, contributing to posthemorrhagic hydrocephalus (PHH). Clinically, GMH-IVH severity is graded on a I to IV scale: Grade I is confined to the germinal matrix, grade II includes intraventricular hemorrhage, grade III includes intraventricular hemorrhage with extension into dilated ventricles, and grade IV includes intraventricular hemorrhage with extension into dilated ventricles as well as parenchymal hemorrhaging. GMH-IVH hematoma volume is the best prognostic indicator, where patients with higher grades have worsened outcomes. Various preclinical studies have shown that rapid hematoma resolution quickly ameliorates inflammation and improves neurological outcomes. Current experimental evidence identifies alternatively activated microglia as playing a pivotal role in hematoma clearance. In this review, we discuss the pathophysiology of GMH-IVH in the development of PHH, microglia/macrophage's role in the neonatal CNS, and established/potential therapeutic targets that enhance M2 microglia/macrophage phagocytosis of blood clots after GMH-IVH.


Subject(s)
Brain/metabolism , Intracranial Hemorrhages/metabolism , Macrophages/metabolism , Microglia/metabolism , Brain/pathology , Humans , Hydrocephalus/etiology , Hydrocephalus/metabolism , Hydrocephalus/pathology , Infant, Newborn , Infant, Premature , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/pathology , Macrophages/pathology , Microglia/pathology
16.
CNS Neurosci Ther ; 25(10): 1151-1161, 2019 10.
Article in English | MEDLINE | ID: mdl-31020769

ABSTRACT

AIMS: Reactive astrogliosis plays a critical role in neurological deficits after germinal matrix hemorrhage (GMH). It has been reported that interleukin-17A and IL-17A receptor IL-17RA/(C/EBPß)/SIRT1 signaling pathway enhances reactive astrogliosis after brain injuries. We evaluated the effects of secukinumab on reactive astrogliosis in a rat pup model of GMH. METHODS: A total of 146 Sprague Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of collagenase. Secukinumab was administered intranasally 1 hour post-GMH. C/EBPß CRISPR or SIRT1 antagonist EX527 was administrated intracerebroventricularly (icv) 48 hours and 1 hour before GMH induction, respectively. Neurobehavior, Western blot, histology, and immunohistochemistry were used to assess treatment regiments in the short term and long term. RESULTS: The endogenous IL-17A, IL-17RA, C/EBPß, and GFAP and proliferation marker CyclinD1 were increased, while SIRT1 expression was decreased after GMH. Secukinumab treatment improved neurological deficits, reduced ventriculomegaly, and increased cortical thickness. Additionally, treatment increased SIRT1 expression and lowered proliferation proteins PCNA and CyclinD1 as well as GFAP expression. C/EBPß CRISPR activation plasmid and EX527 reversed the antireactive astrogliosis effects of secukinumab. CONCLUSION: Secukinumab attenuated reactive astrogliosis and reduced neurological deficits after GMH, partly by regulating IL-17RA/(C/EBPß)/SIRT1 pathways. Secukinumab may provide a promising therapeutic strategy for GMH patients.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Cerebral Hemorrhage/metabolism , Disease Models, Animal , Gliosis/metabolism , Receptors, Interleukin-17/metabolism , Sirtuin 1/metabolism , Administration, Intranasal , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Hemorrhage/drug therapy , Female , Gliosis/drug therapy , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
17.
Cell Death Dis ; 10(2): 97, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718467

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) is a devastating neurological event that contributes to the prolonged neurodevelopmental consequences in infants. Therapeutic strategies focused on attenuating neuronal apoptosis in the penumbra appears to be promising. Given the increasingly recognized neuroprotective roles of adipokines in HIE, we investigated the potential anti-apoptotic roles of a novel member of adipokines, Chemerin, in an experimental model of HIE. In the present study, 10-day-old rat pups underwent right common carotid artery ligation followed by 2.5 h hypoxia. At 1 h post hypoxia, pups were intranasally administered with human recombinant chemerin (rh-chemerin). Here, we showed that rh-chemerin prevented the neuronal apoptosis and degeneration as evidenced by the decreased expression of the pro-apoptotic markers, cleaved caspase 3 and Bax, as well as the numbers of Fluoro-Jade C and TUNEL-positive neurons. Furthermore, rh-Chemerin reversed neurological and morphological impairments induced by hypoxia-ischemia in neonatal rats at 24 h and 4 weeks after HIE. In addition, chemerin-mediated neuronal survival correlated with the elevation of chemerin receptor 23 (chemR23), phosphorylated calmodulin-dependent protein kinase kinase 2 (CAMKK2), as well as phosphorylated adenosine monophosphate-activated protein kinase (AMPK). Specific inhibition of chemR23, CAMKK2, and AMPK abolished the anti-apoptotic effects of rh-chemerin at 24 h after HIE, demonstrating that rh-chemerin ameliorated neuronal apoptosis partially via activating chemR23/CAMKK2/AMPK signaling pathway. Neuronal apoptosis is a well-established contributing factor of pathological changes and the neurological impairment after HIE. These results revealed mechanisms of neuroprotection by rh-chemerin, and indicated that activation of chemR23 might be harnessed to protect from neuronal apoptosis in HIE.


Subject(s)
Apoptosis/drug effects , Chemokines/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Neurons/drug effects , Receptors, Chemokine/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Caspase 3/metabolism , Chemokines/metabolism , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , In Situ Nick-End Labeling , Infant, Newborn , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Phosphorylation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
18.
Brain Behav Immun ; 79: 174-185, 2019 07.
Article in English | MEDLINE | ID: mdl-30711510

ABSTRACT

Neuroinflammation occurs after germinal matrix hemorrhage (GMH) and induces secondary brain injury. Interferon-α (IFN-α) has been shown to exert anti-inflammatory effects in infectious diseases via activating IFNAR and its downstream signaling. We aimed to investigate the anti-inflammatory effects of Recombinant human IFN-α (rh-IFN-α) and the underlying mechanisms in a rat GMH model. Two hundred and eighteen P7 rat pups of both sexes were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Rh-IFN-α was administered intraperitoneally. Small interfering RNA (siRNA) of IFNAR, and siRNA of tumor necrosis factor receptor associated factor 3 (TRAF3) were administered through intracerebroventricular (i.c.v.) injections. JAK1 inhibitor ruxolitinib was given by oral lavage. Post-GMH evaluation included neurobehavioral function, Nissl staining, Western blot analysis, and immunofluorescence. Our results showed that endogenous IFN-α and phosphorylated IFNAR levels were increased after GMH. Administration of rh-IFN-α improved neurological functions, attenuated neuroinflammation, inhibited microglial activation, and ameliorated post-hemorrhagic hydrocephalus after GMH. These observations were concomitant with IFNAR activation, increased expression of phosphorylated JAK1, phosphorylated STAT1 and TRAF3, and decreased levels of phosphorylated NF-κB, IL-6 and TNF-α. Specifically, knockdown of IFNAR, JAK1 and TRAF3 abolished the protective effects of rh-IFN-α. In conclusion, our findings demonstrated that rh-IFN-α treatment attenuated neuroinflammation, neurological deficits and hydrocephalus formation through inhibiting microglial activation after GMH, which might be mediated by IFNAR/JAK1-STAT1/TRAF3/NF-κB signaling pathway. Rh-IFN-α may be a promising therapeutic agent to attenuate brain injury via its anti-inflammatory effect.


Subject(s)
Cerebral Intraventricular Hemorrhage/immunology , Interferon-alpha/metabolism , Neuroimmunomodulation/physiology , Animals , Animals, Newborn , Brain Injuries/metabolism , Cerebral Intraventricular Hemorrhage/chemically induced , Cerebral Intraventricular Hemorrhage/physiopathology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , I-kappa B Proteins/metabolism , Inflammation/metabolism , Interferon-alpha/pharmacology , Interferon-alpha/physiology , Janus Kinase 1/metabolism , Janus Kinase 1/physiology , Male , Microglia/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Neuroimmunomodulation/immunology , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/physiology , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/metabolism
19.
J Cereb Blood Flow Metab ; 39(1): 97-107, 2019 01.
Article in English | MEDLINE | ID: mdl-28792282

ABSTRACT

CD200 has been reported to be neuroprotective in neurodegenerative diseases. However, the potential protective effects of CD200 in germinal matrix hemorrhage (GMH) have not been investigated. We examined the anti-inflammatory mechanisms of CD200 after GMH. A total of 167 seven-day-old rat pups were used. The time-dependent effect of GMH on the levels of CD200 and CD200 Receptor 1 (CD200R1) was evaluated by western blot. CD200R1 was localized by immunohistochemistry. The short-term (24 h) and long-term (28 days) outcomes were evaluated after CD200 fusion protein (CD200Fc) treatment by neurobehavioral assessment. CD200 small interfering RNA (siRNA) and downstream of tyrosine kinase 1 (Dok1) siRNA were injected intracerebroventricularly. Western blot was employed to study the mechanisms of CD200 and CD200R1. GMH induced significant developmental delay and caused impairment in both cognitive and motor functions in rat pups. CD200Fc ameliorated GMH-induced damage. CD200Fc increased expression of Dok1 and decreased IL-1beta and TNF-alpha levels. CD200R1 siRNA and Dok1 siRNA abolished the beneficial effects of CD200Fc, as demonstrated by enhanced expression levels of IL-1beta and TNF-alpha. CD200Fc inhibited GMH-induced inflammation and this effect may be mediated by CD200R1/Dok1 pathway. Thus, CD200Fc may serve as a potential treatment to ameliorate brain injury for GMH patients.


Subject(s)
Cerebral Hemorrhage/drug therapy , DNA-Binding Proteins/drug effects , Inflammation/pathology , Microglia/drug effects , Phosphoproteins/drug effects , RNA-Binding Proteins/drug effects , Receptors, Immunologic/agonists , Animals , Animals, Newborn , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Cerebral Hemorrhage/pathology , Developmental Disabilities/etiology , Immunoglobulin G/therapeutic use , Immunohistochemistry , Injections, Intraventricular , Interleukin-1beta/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Rats , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
20.
Brain Behav Immun ; 70: 179-193, 2018 05.
Article in English | MEDLINE | ID: mdl-29499303

ABSTRACT

Chemerin, an adipokine, has been reported to reduce the production of pro-inflammatory cytokines and neutrophil infiltration. This study investigated the role of Chemerin and its natural receptor, ChemR23, as well as its downstream mediator calmodulin-dependent protein kinase kinase 2 (CAMKK2)/adenosine monophosphate-activated protein kinase (AMPK) /Nuclear factor erythroid 2-related factor 2 (Nrf2) following germinal matrix hemorrhage (GMH) in neonatal rats, with a specific focus on inflammation. GMH was induced by intraparenchymal injection of bacterial collagenase (0.3U) in P7 rat pups. The results demonstrated that human recombinant Chemerin (rh-Chemerin) improved neurological and morphological outcomes after GMH. Rh-Chemerin promoted accumulation and proliferation of M2 microglia in periventricular regions at 72 h. Rh-Chemerin increased phosphorylation of CAMKK2, AMPK and expression of Nrf2, and decreased IL-1beta, IL-6 and TNF-alpha levels. Selective inhibition of ChemR23/CAMKK2/AMPK signaling in microglia via intracerebroventricular delivery of liposome-encapsulated specific ChemR23 (Lipo-alpha-NETA), CAMKK2 (Lipo-STO-609) and AMPK (Lipo-Dorsomorphin) inhibitor increased the expression levels of IL-1beta, IL-6 and TNF- alpha, demonstrating that ChemR23/CAMKK2/AMPK signaling in microglia suppressed inflammatory response after GMH. Cumulatively, these data showed that rh-Chemerin ameliorated GMH-induced inflammatory response by promoting ChemR23/CAMKK2/AMPK/Nrf2 pathway, and M2 microglia may be a major mediator of this effect. Thus, rh-Chemerin can serve as a potential agent to reduce the inflammatory response following GMH.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/therapy , Chemokines/physiology , Intercellular Signaling Peptides and Proteins/physiology , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Anemia, Neonatal , Animals , Animals, Newborn , Brain/embryology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Male , Microglia , NF-E2-Related Factor 2/metabolism , Neuroimmunomodulation/physiology , Phosphorylation , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...