Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38713438

ABSTRACT

Microglia and astrocytes are key players in neuroinflammation and ischemic stroke. A1 astrocytes are a subtype of astrocytes that are extremely neurotoxic and quickly kill neurons. Although the detrimental A1 astrocytes are present in many neurodegenerative diseases and are considered to accelerate neurodegeneration, their role in the pathophysiology of ischemic stroke is poorly understood. Here, we combined RNA-seq, molecular and immunological techniques, and behavioral tests to investigate the role of A1 astrocytes in the pathophysiology of ischemic stroke. We found that astrocyte phenotypes change from a beneficial A2 type in the acute phase to a detrimental A1 type in the chronic phase following ischemic stroke. The activated microglial IL1α, TNF, and C1q prompt commitment of A1 astrocytes. Inhibition of A1 astrocytes induction attenuates reactive gliosis and ameliorates morphological and functional defects following ischemic stroke. The crosstalk between astrocytic C3 and microglial C3aR contributes to the formation of A1 astrocytes and morphological and functional defects. In addition, NF-κB is activated following ischemic stroke and governs the formation of A1 astrocytes via direct targeting of inflammatory cytokines and chemokines. Taken together, we discovered that A2 astrocytes and A1 astrocytes are enriched in the acute and chronic phases of ischemic stroke respectively, and that the C3/C3aR/NF-κB signaling leads to A1 astrocytes induction. Therefore, the C3/C3aR/NF-κB signaling is a novel therapeutic target for ischemic stroke treatment.

2.
Heliyon ; 10(8): e28916, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38655362

ABSTRACT

Context: Shenyu Ningshen (SYNS) tablet is the first pure Chinese medicinal small compound preparation approved for clinical trials for the treatment of depression in China. Clinical experiments confirmed that the formulation had a significant Improvement effect against depression due to the deficiency of both qi and yin. It has been shown to exhibit noticeable anti-inflammatory effect in an animal model of depression. Our previous study showed that SYNS could effectively inhibit the inflammatory response in a depression model. Aim of the study: The purpose of this study was to investigate the protective effects of SYNS on neurons and explore whether the underlying mechanism was associated with A1s. Materials and methods: The depression model of solitary raising-chronic restraint stress (CRS) rats was established; body weight examination, sugar water preference test, open field test, and histological analysis were performed to preliminarily verify the efficacy of the formulation. Subsequently, neuronal nucleus (NeuN) and synaptic-associated proteins (MAP2 and PSD95) were labeled, and the protective effect of SYNS on hippocampal neurons was observed based on the fluorescence intensity of the above indicators. Western blotting, histological examination, and immunofluorescence were used to evaluate the inhibitory effects of SYNS on neuroinflammation and activation of A1s in CRS depression model. Results: SYNS improved behavioral indicators such as weight loss, pleasure loss, and reduced exercise volume in CRS rat model. SYNS restored the CRS-induced histopathological changes in the hippocampus. SYNS showed a certain degree of protective effect on synapses. Further, SYNS inhibited the activation of A1s by inhibiting neuroinflammatory factors in the hippocampus. Conclusion: Our results showed that SYNS had a certain degree of neuroprotective effect, which might be related to its inhibition of the inflammatory response and A1s.

3.
Mol Neurobiol ; 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38367135

ABSTRACT

M2 microglia transplantation has previously demonstrated beneficial effects on spinal cord injury (SCI) by regulating neuroinflammation and enhancing neuronal survival. Exosomes (EXOs), secreted by almost all cell types, embody partial functions and properties of their parent cells. However, the effect of M2 microglia-derived EXOs (M2-EXOs) on SCI recovery and the underlying molecular mechanisms remain unclear. In this study, we isolated M2-EXOs and intravenously introduced them into mice with SCI. Considering the reciprocal communication between microglia and astroglia in both healthy and injured central nervous systems (CNSs), we subsequently focused on the influence of M2-EXOs on astrocyte phenotype regulation. Our findings indicated that M2-EXOs promoted neuron survival and axon preservation, reduced the lesion area, inhibited A1 astrocyte activation, and improved motor function recovery in SCI mice. Moreover, they inhibited the nuclear translocation of p65 and the activation of the NF-κB signalling pathway in A1 astrocytes. Therefore, our research suggests that M2-EXOs mitigate the activation of neurotoxic A1 astrocytes by inhibiting the NF-κB signalling pathway, thereby improving spinal tissue preservation and motor function recovery following SCI. This positions M2-EXOs as a promising therapeutic strategy for SCI.

4.
Neurochem Res ; 48(12): 3571-3584, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37556038

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease with selective degeneration of motor neurons. It has been reported that an increase in the levels of inflammatory cytokines and glial cells such as reactive astrocytes is closely involved in the pathological progression of ALS. Recently, the levels of neuropathic cytotoxic (A1) astrocytes among reactive astrocytes have reportedly increased in the central nervous system of ALS mice, which induce motor neuron degeneration through the production of inflammatory cytokines and secretion of neuropathic factors. Hence, elucidating the induction mechanism of A1 astrocytes in ALS is important to understand the mechanism of disease progression in ALS. In this study, we observed that the expression of peroxiredoxin 6 (PRDX6), a member of the peroxiredoxin family, was markedly upregulated in astrocytes of the lumbar spinal cord of SOD1G93A mice model for ALS. Additionally, when PRDX6 was transiently transfected into the mouse astrocyte cell line C8-D1A and human astrocytoma cell line U-251 MG, the mRNA expression of complement C3 (a marker for A1 astrocyte phenotype) and inflammatory cytokines was increased. Furthermore, the mRNA expression of C3 and inflammatory cytokine was increased in C8-D1A and U-251 MG cells stably expressing PRDX6, and the increased mRNA expression was significantly suppressed by MJ33 (lithium[1-hexadecoxy-3-(2,2,2-trifluoroethoxy) propan-2-yl] methyl phosphate), an inhibitor of the phospholipase A2 activity of PRDX6. Our results suggest that the expression of PRDX6 in astrocytes plays an important role in the induction of A1 astrocytes and expression of inflammatory cytokines in the ALS mice model.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Neurotoxicity Syndromes , Mice , Humans , Animals , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Peroxiredoxin VI/genetics , Peroxiredoxin VI/metabolism , Neurodegenerative Diseases/metabolism , Mice, Transgenic , Spinal Cord/metabolism , Cytokines/metabolism , Disease Models, Animal , Neurotoxicity Syndromes/metabolism , RNA, Messenger/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase/metabolism
5.
ACS Appl Mater Interfaces ; 15(25): 29876-29888, 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37334941

ABSTRACT

Resistance to traditional antiepileptic drugs is a major challenge in chronic epilepsy treatment. MicroRNA-based gene therapy is a promising alternative but has demonstrated limited efficacy due to poor blood-brain barrier permeability, cellular uptake, and targeting efficiency. Adenosine is an endogenous antiseizure agent deficient in the epileptic brain due to elevated adenosine kinase (ADK) activity in reactive A1 astrocytes. We designed a nucleic acid nanoantiepileptic drug (tFNA-ADKASO@AS1) based on a tetrahedral framework nucleic acid (tFNA), carrying an antisense oligonucleotide targeting ADK (ADKASO) and A1 astrocyte-targeted peptide (AS1). This tFNA-ADKASO@AS1 construct effectively reduced brain ADK, increased brain adenosine, mitigated aberrant mossy fiber sprouting, and reduced the recurrent spontaneous epileptic spike frequency in a mouse model of chronic temporal lobe epilepsy. Further, the treatment did not induce any neurotoxicity or major organ damage. This work provides proof-of-concept for a novel antiepileptic drug delivery strategy and for endogenous adenosine as a promising target for gene-based modulation.


Subject(s)
Epilepsy , Nucleic Acids , Mice , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Astrocytes/metabolism , Adenosine Kinase/genetics , Adenosine Kinase/metabolism , Nucleic Acids/metabolism , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/metabolism , Adenosine/pharmacology
6.
Free Radic Biol Med ; 195: 329-342, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36610561

ABSTRACT

Epilepsy is a common neurological disorder with a complex etiology. Ferroptosis, a new form of programmed cell death, is characterized by the accumulation of lipid peroxides and associated with seizures. However, the underlying mechanism of ferroptosis in epilepsy remains elusive. Here, we found that GPX4-GSH-dependent neuronal ferroptosis was detected in epileptic mice, which was attenuated with ferroptosis inhibitors. Moreover, activated neurotoxic A1 astrocytes facilitated seizure-related neuronal ferroptosis in epileptic brains. Inhibition of ferroptosis blocked A1 astrocyte-induced neurotoxicity. A1 astrocyte-secreted CXCL10 enhanced STAT3 phosphorylation but suppressed SLC7A11 in neurons via CXCR3, leading to ferroptosis-associated lipid peroxidation in a GPX4-dependent manner. This was in line with clinical findings, showing a significant correlation between neuronal ferroptosis and A1 astrocytes in epileptic patients. In summary, the present data show that A1 astrocyte-induced neuronal ferroptosis contributes to the pathogenesis of epilepsy, which offers a novel therapeutic target for precision medicine.


Subject(s)
Epilepsy , Ferroptosis , Mice , Animals , Ferroptosis/genetics , Astrocytes/metabolism , Apoptosis , Epilepsy/genetics , Epilepsy/metabolism , Neurons/metabolism
7.
Aging Dis ; 13(3): 943-959, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35656116

ABSTRACT

Astrocytes play an essential role in the modulation of blood-brain barrier function. Neurological diseases induce the transformation of astrocytes into a neurotoxic A1 phenotype, exacerbating brain injury. However, the effect of A1 astrocytes on the BBB dysfunction after stroke is unknown. Adult male ICR mice (n=97) were subjected to 90-minute transient middle cerebral artery occlusion (tMCAO). Immunohistochemical staining of A1 (C3d) and A2 (S100A10) was performed to characterize phenotypic changes in astrocytes over time after tMCAO. The glucagon-like peptide-1 receptor agonist semaglutide was intraperitoneally injected into mice to inhibit A1 astrocytes. Infarct volume, atrophy volume, neurobehavioral outcomes, and BBB permeability were evaluated. RNA-seq was adopted to explore the potential targets and signaling pathways of A1 astrocyte-induced BBB dysfunction. Astrocytic C3d expression was increased, while expression of S100A10 was decreased in the first two weeks after tMCAO, reflecting a shift in the astrocytic phenotype. Semaglutide treatment reduced the expression of CD16/32 in microglia and C3d in astrocytes after ischemic stroke (p<0.05). Ischemia-induced brain infarct volume, atrophy volume and neuroinflammation were reduced in the semaglutide-treated mice, and neurobehavioral outcomes were improved compared to control mice (p<0.05). We further demonstrated that semaglutide treatment reduced the gap formation of tight junction proteins ZO-1, claudin-5 and occludin, as well as IgG leakage three days following tMCAO (p<0.05). In vitro experiments revealed that A1 astrocyte-conditioned medium disrupted BBB integrity. RNA-seq showed that A1 astrocytes were enriched in inflammatory factors and chemokines and significantly modulated the TNF and chemokine signaling pathways, which are closely related to barrier damage. We concluded that astrocytes undergo a phenotypic shift over time after ischemic stroke. C3d+/GFAP+ astrocytes aggravate BBB disruption, suggesting that inhibiting C3d+/GFAP+ astrocyte formation represents a novel strategy for the treatment of ischemic stroke.

8.
Int Immunopharmacol ; 108: 108771, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35461109

ABSTRACT

Astrocytes (AST) play an important role in the pathogenesis of neurological disorders, and their activation is involved in the progression of multiple sclerosis (MS). (6aS, 10S, 11aR, 11bR, 11cS)-10-methylaminododecahydro-3a, 7a-diaza-benzo (de) anthracene-8-thione (MASM), a novel derivative of matrine, exhibits vast pharmacological activities, such as anti-tumor, anti-fibrosis and immune regulation. In this study, we demonstrate that MASM is a promising agent for the treatment of experimental autoimmune encephalomyelitis (EAE). MASM not only inhibited inflammatory responses in LPS-stimulated astrocytes, but also suppressed the formation of reactive A1 astrocyte and maintained astrocytic functions, including the ability to promote synapse formation and phagocytose synapses and myelin debris. Importantly, MASM could significantly alleviate the development of EAE, with significant inhibition of inflammation, demyelination, axon loss and the body weight loss. Meanwhile, MASM also inhibited the activation of astrocytes and improved the function of BBB in vivo. These findings provide novel insights into the protective effect of MASM on EAE, which may be a promising drug candidate for treatment of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Neoplasms , Animals , Anthracenes , Astrocytes , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Thiones/pharmacology
9.
Cell Signal ; 89: 110183, 2022 01.
Article in English | MEDLINE | ID: mdl-34728368

ABSTRACT

Ror2 plays an important role in neuronal development, neuronal plasticity, and neuropathic pain. In our previous pilot study, we found that Ror2 and GFAP (a marker of astrocytes) protein levels increased in thoracic dorsal root ganglia from postoperative day (POD) 7 to POD 21 in rats with chronic post-thoracotomy pain (CPTP). In the present study, we aimed to further explore the roles of Ror2 and activated astrocytes during CPTP development. Ror2, c-JUN, and C3aR levels increased and the activated astrocytes were mainly expressed as the A1 phenotype in the spinal cord dorsal horn of the rats with CPTP. The knockdown of Ror2 in the spinal cord astrocytes alleviated thoracotomy-induced mechanical hyperalgesia and cold allodynia as well as reverted the A1/A2 ratio of the reactive astrocytes, downregulating the expression of c-JUN and C3aR in rats with CPTP. These results suggest that Ror2 in the spinal cord astrocytes mediates the transformation of A1/A2 reactive astrocytes via regulating the expressions of the c-JUN and C3aR in CPTP. Furthermore, the suppression of Ror2 could be utilized as a new strategy to help prevent CPTP.


Subject(s)
Astrocytes , Chronic Pain , Animals , Astrocytes/metabolism , Chronic Pain/metabolism , Pilot Projects , Rats , Rats, Sprague-Dawley , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Thoracotomy
10.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1014176

ABSTRACT

Aim To evaluate the effects of different doses of IL-36Ra on pain behavior and the polarization of spinal A1 astrocytes in mice with inflammatory pain.Methods A total of 32 male C57BL/6 mice were divided into four groups: CFA+Saline group, CFA+IL-36Ra 50 ng group, CFA+IL-36Ra 100 ng group and CFA+IL-36Ra 200 ng group by random grouping.The inflammatory pain model was established by injection of complete Freund's adjuvant(CFA)into the plantar surface of the right hind paw of mice.The drugs were given daily from the 1st day to the 7th day after CFA injection in each group by intrathecal injection.The changes in the mechanical withdrawal threshold(MWT)and the radiant heat stimulating paw withdrawal latency(PWL)of the mice were detected before and 1, 3, 5 and 7 days after the CFA injection.Reverse transcription polymerase chain reaction was used to detect the expression changes of A1 and A2 astrocyte markers after IL-36Ra treatment.Immunohistochemistry was used to test the effect of IL-36Ra on the co-expression level of A1 astrocyte marker C3 and GFAP in the spinal dorsal horn.Results MWT and PWL of the ipsilateral paw significantly decreased after the CFA injection, and IL-36Ra(100 ng, 200 ng)treatment could significantly improve the mechanical allodynia and thermal hyperalgesia of CFA mice.After treatment for 7 days, IL-36Ra 200 ng successfully reversed the increase of GFAP and Lcn2 expression in the spinal cord of CFA mice, which demonstrated IL-36Ra could inhibit the activation of astrocytes.IL-36Ra significantly inhibited the expression of A1 astrocyte maker Serping1, H2-T23 in spinal cord but showed no effects on the expression of A2 astrocytes marker with each dose.Furthermore, IL-36Ra inhibited the expression of C3 within the astrocytes in the spinal dorsal horn of CFA mice.Conclusion IL-36Ra attenuates the inflammatory pain via inhibiting the polarization of A1 reactive astrocytes in the spinal cord of mice with inflammatory pain.

11.
J Chem Neuroanat ; 117: 102004, 2021 11.
Article in English | MEDLINE | ID: mdl-34280490

ABSTRACT

Type 1 astrocytes (A1), which are highly proinflammatory and neurotoxic, are prevalent in multiple sclerosis (MS). In addition, in MS and its animal model, experimental autoimmune encephalomyelitis (EAE), immune cells must cross the blood-brain barrier (BBB) and infiltrate into the parenchyma of the central nervous system (CNS) in order to induce neurological deficits. We have previously reported that treatment of EAE with matrine (MAT), a quinazine alkaloid derived from Sophorae Flavescens, effectively inhibited CNS inflammation and promoted neuroregeneration. However, the impact of MAT treatment on astrocyte phenotype is not known. In the present study, we showed that MAT treatment inhibited the generation of neurotoxic A1 astrocytes and promoted neuroprotective A2 astrocytes in the CNS of EAE, most likely by inhibiting production of the A1-inducing cytokine cocktail. MAT also downregulated the expression of vascular endothelial growth factor-A (VEGF-A) and upregulated tight junction proteins Claudin 5 and Occludin, thus protecting the BBB from CNS inflammation-induced damage. Moreover, MAT treatment promotes the formation of astrocyte tight junctions at glia limitans, thereby limiting parenchymal invasion of the CNS by immune cells. Taken together, the inhibition of A1 astrogliogenesis, and the dual effects on the BBB and astrocytic glia limitans, may be the mechanisms whereby MAT significantly improves EAE clinical scores and neuroprotection.


Subject(s)
Alkaloids/pharmacology , Astrocytes/drug effects , Autoimmunity/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Phenotype , Quinolizines/pharmacology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Autoimmunity/physiology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Brain/immunology , Brain/metabolism , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Female , Guinea Pigs , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Matrines
12.
Front Neurosci ; 15: 628917, 2021.
Article in English | MEDLINE | ID: mdl-34135725

ABSTRACT

Traumatic spinal cord injury (TSCI) leads to pathological changes such as inflammation, edema, and neuronal apoptosis. Methylprednisolone (MP) is a glucocorticoid that has a variety of beneficial effects, including decreasing inflammation and ischemic reaction, as well as inhibiting lipid peroxidation. However, the efficacy and mechanism of MP in TSCI therapy is yet to be deciphered. In the present study, MP significantly attenuated the apoptotic effects of H2O2 in neuronal cells. Western blot analysis demonstrated that the levels of apoptotic related proteins, Bax and cleaved caspase-3, were reduced while levels of anti-apoptotic Bcl-2 were increased. In vivo TUNEL assays further demonstrated that MP effectively protected neuronal cells from apoptosis after TSCI, and was consistent with in vitro studies. Furthermore, we demonstrated that MP could decrease expression levels of IBA1, Il-1α, TNFα, and C3 and suppress A1 neurotoxic reactive astrocyte activation in TSCI mouse models. Neurological function was evaluated using the Basso Mouse Scale (BMS) and Footprint Test. Results demonstrated that the neurological function of MP-treated injured mice was significantly increased. In conclusion, our study demonstrated that MP could attenuate astrocyte cell death, decrease microglia activation, suppress A1 astrocytes activation, and promote functional recovery after acute TSCI in mouse models.

13.
Mol Neurobiol ; 58(9): 4506-4519, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34076838

ABSTRACT

To identify potential regulators and investigate the molecular mechanism of macrophage polarization affecting astrocyte activation from the perspective of non-coding RNA regulation, we isolated mouse bone marrow mononuclear cells (BMMNCs)-induced macrophages toward M1 or M2a polarization. Long non-coding RNA NEAT1 and IL-33 expression levels were significantly upregulated in M2a macrophages; NEAT1 knockdown in M2a macrophages markedly reduced the protein levels of IL-33 and M2a markers, IL-4 and IL-13 concentrations, and the bacterial killing capacity of M2a macrophages. NEAT1 acted as a competing endogenous RNA (ceRNA) to regulate IL-33 expression by sponging miR-224-5p in M2a macrophages; NEAT1 knockdown upregulated miR-224-5p expression, while miR-224-5p inhibition increased the protein content and concentration of IL-33. miR-224-5p inhibition exerted the opposite effects on the protein levels of IL-33 and M2a markers, IL-4 and IL-13 concentrations, and the bacterial killing capacity of M2a macrophages compared to NEAT1 knockdown; the effects of NEAT1 knockdown were significantly reversed by miR-224-5p inhibition. M2a macrophage conditioned medium (CM) significantly suppressed the activation of A1 astrocytes. NEAT1 knockdown M2a macrophage CM led to enhanced A1 astrocyte activation while miR-224-5p-silenced M2a macrophage CM led to a blockade of A1 astrocyte activation; the effects of NEAT1 knockdown M2a macrophage CM on A1 astrocyte activation were significantly reversed by miR-224-5p inhibition in M2a macrophages. The NEAT1/miR-224-5p/IL-33 axis modulates macrophage M2a polarization, therefore affecting A1 astrocyte activation.


Subject(s)
Astrocytes/metabolism , Interleukin-33/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction/physiology , Animals , Astrocytes/cytology , Cell Polarity , Macrophage Activation , Macrophages/cytology , Mice
14.
Biochem Biophys Rep ; 25: 100905, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33553683

ABSTRACT

Functional improvement after spinal cord injury remains an unsolved difficulty. Glial scars, a major component of SCI lesions, are very effective in improving the rate of this recovery. Such scars are a result of complex interaction mechanisms involving three major cells, namely, astrocytes, oligodendrocytes, and microglia. In recent years, scientists have identified two subtypes of reactive astrocytes, namely, A1 astrocytes that induce the rapid death of neurons and oligodendrocytes, and A2 astrocytes that promote neuronal survival. Moreover, recent studies have suggested that the macrophage polarization state is more of a continuum between M1 and M2 macrophages. M1 macrophages that encourage the inflammation process kill their surrounding cells and inhibit cellular proliferation. In contrast, M2 macrophages promote cell proliferation, tissue growth, and regeneration. Furthermore, the ability of oligodendrocyte precursor cells to differentiate into adult oligodendrocytes or even neurons has been reviewed. Here, we first scrutinize recent findings on glial cell subtypes and their beneficial or detrimental effects after spinal cord injury. Second, we discuss how we may be able to help the functional recovery process after injury.

15.
Front Neurosci ; 14: 824, 2020.
Article in English | MEDLINE | ID: mdl-32848579

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a multifactorial disease, characterized by a progressive loss of motor neurons that eventually leads to paralysis and death. The current ALS-approved drugs modestly change the clinical course of the disease. The mechanism by which motor neurons progressively degenerate remains unclear but entails a non-cell autonomous process. Astrocytes impaired biological functionality were implicated in multiple neurodegenerative diseases, including ALS, frontotemporal dementia (FTD), Parkinson's disease (PD), and Alzheimer disease (AD). In ALS disease patients, A1 reactive astrocytes were found to play a key role in the pathology of ALS disease and death of motor neurons, via loss or gain of function or acquired toxicity. The contribution of astrocytes to the maintenance of motor neurons by diverse mechanisms makes them a promising therapeutic candidate for the treatment of ALS. Therapeutic approaches targeting at modulating the function of endogenous astrocytes or replacing lost functionality by transplantation of healthy astrocytes, may contribute to the development of therapies which might slow down or even halt the progression ALS diseases. The proposed mechanisms by which astrocytes can potentially ameliorate ALS progression and the status of ALS clinical studies involving astrocytes are discussed.

16.
Cell ; 180(5): 833-846.e16, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32142677

ABSTRACT

Cognitive dysfunction and reactive microglia are hallmarks of traumatic brain injury (TBI), yet whether these cells contribute to cognitive deficits and secondary inflammatory pathology remains poorly understood. Here, we show that removal of microglia from the mouse brain has little effect on the outcome of TBI, but inducing the turnover of these cells through either pharmacologic or genetic approaches can yield a neuroprotective microglial phenotype that profoundly aids recovery. The beneficial effects of these repopulating microglia are critically dependent on interleukin-6 (IL-6) trans-signaling via the soluble IL-6 receptor (IL-6R) and robustly support adult neurogenesis, specifically by augmenting the survival of newborn neurons that directly support cognitive function. We conclude that microglia in the mammalian brain can be manipulated to adopt a neuroprotective and pro-regenerative phenotype that can aid repair and alleviate the cognitive deficits arising from brain injury.


Subject(s)
Brain Injuries, Traumatic/therapy , Interleukin-6/genetics , Receptors, Interleukin-6/genetics , Regeneration/genetics , Animals , Brain/growth & development , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Cognitive Dysfunction/therapy , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Signal Transduction/genetics
17.
Front Neurosci ; 13: 429, 2019.
Article in English | MEDLINE | ID: mdl-31130839

ABSTRACT

Two types of reactive astrocytes, A1 and A2 astrocytes, are induced following neuroinflammation and ischemia. In this study, we evaluated the effects of the fibroblast growth factor (FGF)2/FGF receptor (FGFR)1 pathway on A1 and A2 astrocytes in the rat hippocampus using double-labeling immunofluorescence following infrasound exposure. A1 astrocytes were induced in the CA1 region of the hippocampus after exposure to infrasound for 3 days. The number of microglial cells was also increased, and we investigated if these might be responsible for the reactivity of A1 astrocytes. Accordingly, expression levels of C3 and Iba-1, as markers of A1 astrocytes and microglial cells, respectively, were both up-regulated in rat hippocampus following infrasound exposure, as demonstrated by western blot. We also explored the effect of the FGF2/FGFR1 pathway on A1 astrocyte reactivity by pretreating rats with FGF2 or the specific FGFR1 antagonist, PD173074. A1 astrocytes were gradually down-regulated by activation of the FGF2/FGFR1 pathway and were up-regulated by inhibition of the FGF2/FGFR1 pathway after infrasound damage. These results further our understanding of the role of reactive astrocytes in infrasound-induced central nervous system injury and will thus facilitate the development of new treatments for these injuries.

18.
Arch Pharm Res ; 42(5): 416-425, 2019 May.
Article in English | MEDLINE | ID: mdl-30830660

ABSTRACT

Glial cells outnumber neurons in the brain and play important roles in the neuroinflammation that accompanies brain damage in neurodegenerative diseases. In Parkinson's disease (PD), dopaminergic neuronal loss is accompanied by inflammatory changes in microglia, astrocytes, innate immune cells, and infiltrating peripheral immune cells. Neuroinflammation is probably a fundamental immune response to protect neurons from harm and compensate for neuronal damage, but at the same time, its neurotoxic effects exacerbate neuron damage. Furthermore, neuroinflammatory response is regulated by immune cells, such as microglia, astrocytes, and peripheral immune cells, and by cytokines and chemokines. Accordingly, it is crucial that we understand how such immune cells in the brain regulate neuroinflammatory responses in PD pathology. This review describes the roles played by glia-mediated neuroinflammation in PD, both good and bad, and the therapeutic strategies used to treat PD.


Subject(s)
Brain/pathology , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Parkinson Disease/prevention & control , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Brain/cytology , Brain/drug effects , Brain/immunology , Cytokines/antagonists & inhibitors , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Parkinson Disease/etiology , Parkinson Disease/immunology , Parkinson Disease/pathology , Peroxisome Proliferator-Activated Receptors/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptors/metabolism
19.
Brain Behav Immun ; 70: 76-87, 2018 05.
Article in English | MEDLINE | ID: mdl-29604346

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disorder characterized by the central nervous system (CNS) infiltration of myelin-specific pathogenic T cells followed by brain inflammation in association with demyelination. Similarly, experimental autoimmune encephalomyelitis (EAE), the animal model of MS, also exhibits increased CNS infiltration of pathogenic T cells, including Th1 and Th17, leading to detrimental effects of neuroinflammation and demyelination. We previously reported that 3H-1,2-dithiole-3-thione (D3T), the structurally-simplest of the sulfur-containing dithiolethiones, exerted a promising therapeutic effect in EAE. In the current study we report that 5-Amino-3-thioxo-3H-(1,2)dithiole-4-carboxylic acid ethyl ester (ACDT), a substituted derivative of D3T, exhibits anti-inflammatory properties in EAE. ACDT, administered post immunization, delayed disease onset and reduced disease severity in chronic C57BL/6 EAE, and ACDT, administered during disease remission, suppressed disease relapse in relapsing-remitting SJL/J EAE. Further analysis of the cellular and molecular mechanisms underlying the protective effects of ACDT in EAE revealed that ACDT inhibited pathogenic T cell infiltration, suppressed microglia activation, repressed neurotoxic A1 astrocyte generation, lessened blood-brain barrier disruption, and diminished MMP3/9 production in the CNS of EAE. In summary, we demonstrate that ACDT suppresses neuroinflammation and ameliorates disease severity in EAE through multiple cellular mechanisms. Our findings suggest the potential of developing ACDT as a novel therapeutic agent for the treatment of MS/EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Thiones/therapeutic use , Thiophenes/therapeutic use , Animals , Central Nervous System , Disease Models, Animal , Female , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Myelin Sheath , Th1 Cells/drug effects , Th17 Cells/drug effects , Thiones/chemical synthesis , Thiones/pharmacology , Thiophenes/chemical synthesis , Thiophenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...