Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Orthop Surg Res ; 19(1): 396, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982418

ABSTRACT

BACKGROUND: The progression of osteoporosis (OP) can dramatically increase the risk of fractures, which seriously disturb the life of elderly individuals. Specific protein 1 (SP1) is involved in OP progression. However, the mechanism by which SP1 regulates OP progression remains unclear. OBJECTIVE: This study investigated the mechanism underlying the function of SP1 in OP. METHODS: SAMP6 mice were used to establish an in vivo model of age-dependent OP, and BALB/c mice were used as controls. BMSCs were extracted from two subtypes of mice. Hematoxylin and eosin staining were performed to mark the intramedullary trabecular bone structure to evaluate histological changes. ChIP assay was used to assess the targeted regulation between SP1 and miR-133a-3p. The binding sites between MAPK3 and miR-133a-3p were verified using a dual-luciferase reporter assay. The mRNA levels of miR-133a-3p and MAPK3 were detected using quantitative reverse transcription polymerase chain reaction (RT-qPCR). The protein expression of SP1, MAPK3, Colla1, OCN, and Runx2 was examined using Western blotting. Alkaline phosphatase (ALP) kit and Alizarin Red S staining were used to investigate ALP activity and mineralized nodules, respectively. RESULTS: The levels of SP1 and miR-133a-3p were upregulated, whereas the expression of MAPK3 was downregulated in BMSCs from SAMP6 mice, and miR-133a-3p inhibitor accelerated osteogenic differentiation in BMSCs. SP1 directly targeted miR-133a-3p, and MAPK3 was the downstream mRNA of miR-133a-3p. Mechanically, SP1 accelerated osteogenic differentiation in BMSCs via transcriptional mediation of the miR-133a-3p/MAPK3 axis. CONCLUSION: SP1 regulates osteogenic differentiation by mediating the miR-133a-3p/MAPK3 axis, which would shed new light on strategies for treating senile OP.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , MicroRNAs , Mitogen-Activated Protein Kinase 3 , Osteogenesis , Osteoporosis , Sp1 Transcription Factor , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mice, Inbred BALB C , Cells, Cultured , Disease Models, Animal , Male
2.
Heart Vessels ; 37(6): 1085-1096, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35320391

ABSTRACT

Atherosclerosis (AS) is the basic lesion underlying the occurrence and development of cerebrovascular diseases. Abnormal proliferation of vascular smooth muscle cells (VSMCs) plays a crucial role in AS. We aimed to explore the role of SNHG16 in AS and the molecular mechanism of VSMC involvement in the regulation of AS. The expression levels of SNHG16, miR-30c-5p and SDC2 were detected by qRT-PCR. CCK-8, wound healing and Transwell assays were used to assess ox-LDL-induced VSMC proliferation, migration, and invasion, respectively. Western blot analysis was used to detect SDC2 and MEK/ERK pathway-related protein levels. A dual-luciferase reporter assay confirmed the binding of SNHG16 with miR-30c-5p and miR-30c-5p with SDC2. SNHG16 and SDC2 expression was upregulated in patients with AS and ox-LDL-induced VSMCs, while miR-30c-5p was downregulated. Ox-LDL-induced VSMC proliferation and migration were increased, and the MEK/ERK signalling pathway was activated. MiR-30c-5p was targeted to SNHG16 and SDC2. Downregulating SNHG16 or upregulating miR-30c-5p inhibited ox-LDL-induced VSMC proliferation and migration and inhibited MEK/ERK signalling pathway activation. In contrast, downregulating miR-30c-5p or upregulating SDC2 reversed the effects of downregulating SNHG16 or upregulating miR-30c-5p. Furthermore, downregulating SDC2 inhibited ox-LDL-induced proliferation and migration of VSMCs and inhibited activation of the MEK/ERK signalling pathway, while upregulating lncRNA SNHG16 reversed the effects of downregulating SDC2. Downregulation of SNHG16 inhibited VSMC proliferation and migration in AS by targeting the miR-30c-5p/SDC2 axis. This study provides a possible therapeutic approach to AS.


Subject(s)
Atherosclerosis , Intracranial Arteriosclerosis , MicroRNAs , RNA, Long Noncoding/genetics , Atherosclerosis/pathology , Cell Movement , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation , Humans , Intracranial Arteriosclerosis/metabolism , Intracranial Arteriosclerosis/pathology , Lipoproteins, LDL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Syndecan-2/genetics , Syndecan-2/metabolism , Syndecan-2/pharmacology
3.
Sci Adv ; 7(36): eabh0609, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34516909

ABSTRACT

Microglia have been implicated in neuroinflammatory diseases, including multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). We demonstrate that microglia mediate EAE disease progression via a mechanism relying on the noncanonical nuclear factor kB (NF-κB) pathway. Microglia-specific deletion of the noncanonical NF-κB-inducing kinase (NIK) impairs EAE disease progression. Although microglial NIK is dispensable for the initial phase of T cell infiltration into the central nervous system (CNS) and EAE disease onset, it is critical for the subsequent CNS recruitment of inflammatory T cells and monocytes. Our data suggest that following their initial CNS infiltration, T cells activate the microglial noncanonical NF-κB pathway, which synergizes with the T cell-derived cytokine granulocyte-macrophage colony-stimulating factor to induce expression of chemokines involved in the second-wave of T cell recruitment and disease progression. These findings highlight a mechanism of microglial function that is dependent on NIK signaling and required for EAE disease progression.

4.
iScience ; 24(3): 102186, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33718838

ABSTRACT

Microglia are ubiquitous central nervous system (CNS)-resident macrophages that maintain homeostasis of neural tissues and protect them from pathogen attacks. Yet, their differentiation in different compartments remains elusive. We performed single-cell RNA-seq to compare microglial subtypes in the cortex and the spinal cord. A multi-way comparative analysis was carried out on samples from C57/BL and HIV gp120 transgenic mice at two, four, and eight months of age. The results revealed overlapping but distinct microglial populations in the cortex and the spinal cord. The differential heterogeneity of microglia in these CNS regions was further suggested by their disparity of plasticity in response to life span progression and HIV-1 pathogenic protein gp120. Our findings indicate that microglia in different CNS compartments are adapted to their local environments to fulfill region-specific biological functions.

5.
J Neuroinflammation ; 17(1): 249, 2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32843067

ABSTRACT

BACKGROUND: The Zika virus (ZIKV) outbreak that occurred in multiple countries was linked to increased risk of nervous system injuries and congenital defects. However, host immunity- and immune-mediated pathogenesis in ZIKV infection are not well understood. Interleukin-22 (IL-22) is a crucial cytokine for regulating host immunity in infectious diseases. Whether IL-22 plays, a role in ZIKV infection is unknown. METHODS: The cellular source of IL-22 was identified in IFNAR-/- mice and wild-type (WT) neonatal mice during ZIKV infection. To determine the role of IL-22, we challenged 1-day-old WT and IL-22-/- mice with ZIKV and monitored clinical manifestations. Glial cell activation in the brain was assessed by confocal imaging. ZIKV-specific CD8+ T cell responses in both the spleen and brain were analyzed by flow cytometry. In addition, glial cells were cultured in vitro and infected with ZIKV in the presence of IL-22, followed by the evaluation of cell proliferation, cytokine expression, and viral loads. RESULTS: We found that γδ T cells were the main source of IL-22 during ZIKV infection in both the spleen and brain. WT mice began to exhibit weight loss, staggered steps, bilateral hind limb paralysis, and weakness at 10 days post-infection (dpi) and ultimately succumbed to infection at 16-19 dpi. IL-22 deficiency lessened weight loss, moderated the systemic inflammatory response, and greatly improved clinical signs of neurological disease and mortality. ZIKV infection also induced the activation of microglia and astrocytes in vitro. Additional analysis demonstrated that the absence of IL-22 resulted in reduced activation of microglia and astrocytes in the cortex. Although IL-22 displayed a negligible effect on glial cells in vitro, IL-22-/- mice mounted more vigorous ZIKV-specific CD8+ T cell responses, which led to a more effective control of ZIKV in the brain. CONCLUSIONS: Our data revealed a pathogenic role of IL-22 in ZIKV encephalitis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukins/metabolism , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Interleukins/genetics , Mice , Mice, Knockout , Neuroglia/metabolism , Neuroglia/virology , Zika Virus Infection/metabolism , Interleukin-22
6.
Sci Rep ; 10(1): 5137, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198485

ABSTRACT

While astrocytes have been traditionally described as passive supportive cells, studies during the last decade have shown they are active players in many aspects of CNS physiology and function both in normal and disease states. However, the precise mechanisms regulating astrocytes function and interactions within the CNS are still poorly understood. This knowledge gap is due in large part to the limitations of current image analysis tools that cannot process astrocyte images efficiently and to the lack of methods capable of quantifying their complex morphological characteristics. To provide an unbiased and accurate framework for the quantitative analysis of fluorescent images of astrocytes, we introduce a new automated image processing pipeline whose main novelties include an innovative module for cell detection based on multiscale directional filters and a segmentation routine that leverages deep learning and sparse representations to reduce the need of training data and improve performance. Extensive numerical tests show that our method performs very competitively with respect to state-of-the-art methods also in challenging images where astrocytes are clustered together. Our code is released open source and freely available to the scientific community.


Subject(s)
Astrocytes/physiology , Brain/cytology , Brain/physiology , Deep Learning , Image Processing, Computer-Assisted/methods , Algorithms , Humans , Neural Networks, Computer
7.
J Neurosci ; 39(42): 8408-8421, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31471472

ABSTRACT

HIV-1 infection of the nervous system causes various neurological diseases, and synaptic degeneration is likely a critical step in the neuropathogenesis. Our prior studies revealed a significant decrease of synaptic protein, specifically in the spinal dorsal horn of patients with HIV-1 in whom pain developed, suggesting a potential contribution of synaptic degeneration to the pathogenesis of HIV-associated pain. However, the mechanism by which HIV-1 causes the spinal synaptic degeneration is unclear. Here, we identified a critical role of microglia in the synaptic degeneration. In primary cortical cultures (day in vitro 14) and spinal cords of 3- to 5-month-old mice (both sexes), microglial ablation inhibited gp120-induced synapse decrease. Fractalkine (FKN), a microglia activation chemokine specifically expressed in neurons, was upregulated by gp120, and knockout of the FKN receptor CX3CR1, which is predominantly expressed in microglia, protected synapses from gp120-induced toxicity. These results indicate that the neuron-to-microglia intercellular FKN/CX3CR1 signaling plays a role in gp120-induced synaptic degeneration. To elucidate the mechanism controlling this intercellular signaling, we tested the role of the Wnt/ß-catenin pathway in regulating FKN expression. Inhibition of Wnt/ß-catenin signaling blocked both gp120-induced FKN upregulation and synaptic degeneration, and gp120 stimulated Wnt/ß-catenin-regulated FKN expression via NMDA receptors (NMDARs). Furthermore, NMDAR antagonist APV, Wnt/ß-catenin signaling suppressor DKK1, or knockout of CX3CR1 alleviated gp120-induced mechanical allodynia in mice, suggesting a critical contribution of the Wnt/ß-catenin/FKN/CX3R1 pathway to gp120-induced pain. These findings collectively suggest that HIV-1 gp120 induces synaptic degeneration in the spinal pain neural circuit by activating microglia via Wnt3a/ß-catenin-regulated FKN expression in neurons.SIGNIFICANCE STATEMENT Synaptic degeneration develops in the spinal cord dorsal horn of HIV patients with chronic pain, but the patients without the pain disorder do not show this neuropathology, indicating a pathogenic contribution of the synaptic degeneration to the development of HIV-associated pain. However, the mechanism underlying the synaptic degeneration is unclear. We report here that HIV-1 gp120, a neurotoxic protein that is specifically associated with the manifestation of pain in HIV patients, induces synapse loss via microglia. Further studies elucidate that gp120 activates microglia by stimulating Wnt/ß-catenin-regulated fractalkine in neuron. The results demonstrate a critical role of microglia in the pathogenesis of HIV-associated synaptic degeneration in the spinal pain neural circuit.


Subject(s)
HIV Envelope Protein gp120/pharmacology , Microglia/drug effects , Nerve Degeneration/metabolism , Spinal Cord/drug effects , Synapses/drug effects , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Mice , Mice, Knockout , Microglia/metabolism , Neuralgia/metabolism , Spinal Cord/metabolism , Synapses/metabolism , Up-Regulation/drug effects
8.
J Clin Invest ; 128(11): 4980-4991, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30247157

ABSTRACT

The E3 ubiquitin ligase Pellino 1 (Peli1) is a microglia-specific mediator of autoimmune encephalomyelitis. Its role in neurotropic flavivirus infection is largely unknown. Here, we report that mice deficient in Peli1 (Peli1-/-) were more resistant to lethal West Nile virus (WNV) infection and exhibited reduced viral loads in tissues and attenuated brain inflammation. Peli1 mediates chemokine and proinflammatory cytokine production in microglia and promotes T cell and macrophage infiltration into the CNS. Unexpectedly, Peli1 was required for WNV entry and replication in mouse macrophages and mouse and human neurons and microglia. It was also highly expressed on WNV-infected neurons and adjacent inflammatory cells from postmortem patients who died of acute WNV encephalitis. WNV passaged in Peli1-/- macrophages or neurons induced a lower viral load and impaired activation in WT microglia and thereby reduced lethality in mice. Smaducin-6, which blocks interactions between Peli1 and IRAK1, RIP1, and IKKε, did not inhibit WNV-triggered microglia activation. Collectively, our findings suggest a nonimmune regulatory role for Peli1 in promoting microglia activation during WNV infection and identify a potentially novel host factor for flavivirus cell entry and replication.


Subject(s)
Neurons/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , West Nile Fever/metabolism , West Nile virus/physiology , Animals , Chemokines/genetics , Chemokines/metabolism , Chlorocebus aethiops , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Macrophages/metabolism , Macrophages/pathology , Macrophages/virology , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Microglia/virology , Neurons/pathology , Neurons/virology , Nuclear Proteins/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Ubiquitin-Protein Ligases/genetics , Vero Cells , Viral Load , West Nile Fever/genetics , West Nile Fever/pathology
9.
Arch Insect Biochem Physiol ; 98(1): e21449, 2018 May.
Article in English | MEDLINE | ID: mdl-29377233

ABSTRACT

Previous study showed that exogenously applied recombinant thymosin from Bombyx mori (BmTHY) reduces B. mori nucleopolyhedrovirus (BmNPV) proliferation in silkworm. Which stands to reason that BmTHY in B. mori is crucial for the defense against BmNPV. However, little is known about the effect of endogenously overexpressed or repressed BmTHY on B. mori resistance to virus infection. To study this issue, we constructed an overexpression and inhibited expression systems of BmTHY in BmN cells. The viral titer and the analysis from the quantitative real-time polymerase chain reaction (PCR) revealed that overexpression of BmTHY decreased the copies of BmNPV gene gp41, which goes over to inhibit the proliferation of BmNPV in BmN cells, while the inhibited expression of BmTHY significantly enhanced viral proliferation in infected BmN cells. These results indicated that endogenous BmTHY can inhibit BmNPV proliferation and replication in infected BmN cells. Furthermore, Co-IP showed that BmTHY could bind to actin in BmN cells. Also, the overexpression or inhibited expression of BmTHY shifted the ratio of F/G-actin in infected BmN cells. Lastly, the BmTHY, an actin-interacting protein, might be one of the key host factors against BmNPV, which inhibits viral proliferation and replication in BmN cells.


Subject(s)
Bombyx/virology , Insect Proteins/metabolism , Nucleopolyhedroviruses/physiology , Thymosin/metabolism , Actins/metabolism , Animals , Bombyx/genetics , Bombyx/metabolism , Cell Line , Insect Proteins/genetics , Nucleopolyhedroviruses/genetics , Thymosin/genetics , Virus Replication
10.
Mol Brain ; 10(1): 40, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851400

ABSTRACT

Human immunodeficiency virus (HIV) infection induces neuronal injuries, with almost 50% of infected individuals developing HIV-associated neurocognitive disorders (HAND). Although highly activate antiretroviral therapy (HAART) has significantly reduced the incidence of severe dementia, the overall prevalence of HAND remains high. Synaptic degeneration is emerging as one of the most relevant neuropathologies associate with HAND. Previous studies have reported critical roles of viral proteins and inflammatory responses in this pathogenesis. Infected cells, including macrophages, microglia and astrocytes, may release viral proteins and other neurotoxins to stimulate neurons and cause excessive calcium influx, overproduction of free radicals and disruption of neurotransmitter hemostasis. The dysregulation of neural circuits likely leads to synaptic damage and loss. Identification of the specific mechanism of the synaptic degeneration may facilitate the development of effective therapeutic approaches to treat HAND.


Subject(s)
AIDS Dementia Complex/pathology , Nerve Degeneration/pathology , Synapses/pathology , Animals , Disease Models, Animal , Humans , Models, Biological , Phagocytosis
11.
J Neuroimmune Pharmacol ; 11(1): 182-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26582091

ABSTRACT

HIV-1 envelope glycoprotein gp120 (gp120) is a major virulence protein implicated in the pathogenesis of HIV-associated neurocognitive disorders (HAND). Although gp120 has been suggested to cause synaptic and neuronal injuries by disrupting NMDA receptor (NMDAR) function, the underlying mechanism is unclear. Here, we show that gp120Bal down-regulates the phosphorylation of the NMDAR subunit1 NR1 (at Ser896 and Ser897), which is essential for NMDAR function. This effect of gp120Bal is blocked by specific antagonists of both NMDA and AMPA receptors, indicating a critical role of synaptic activation. Furthermore, AMD3100 and maraviroc, antagonists of CCR5 and CXCR4 chemokine receptors, respectively, inhibit the effect of gp120Bal on NR1, suggesting that CXCR4 and CCR5 activation are involved. These findings may provide mechanistic insights into the synaptopathogenesis caused by HIV-1 infection.


Subject(s)
Cerebral Cortex/metabolism , HIV Envelope Protein gp120/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Cerebral Cortex/virology , Down-Regulation , Fluorescent Antibody Technique , Glutamic Acid/metabolism , HIV-1/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism
12.
J Mol Neurosci ; 47(2): 340-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492229

ABSTRACT

Mammalian target of rapamycin (mTOR) signaling plays a critical role in the regulation of activity-dependent protein synthesis in neurons. It is well established that the GTPase-activating protein tuberous sclerosis complex proteins (2TSC2) is an upstream inhibitor of mTOR. In this study, we show that glutamate stimulation down-regulates TSC2 protein in cortical cultures via NMDA receptor (NMDAR) activation. Interestingly, the mTOR-specific inhibitor rapamycin blocks the glutamate-induced TSC2 down-regulation. This finding suggests that NMDAR activation evokes an mTOR-mediated negative regulation of TSC2. In addition, we also show that the glutamate-induced down-regulation of TSC2 protein is blocked by proteasome inhibitor MG132, indicating the involvement of proteasome-mediated protein degradation. We propose that the NMDAR activation stimulates an mTOR-proteasome pathway to degrade TSC2 protein.


Subject(s)
Down-Regulation/physiology , Glutamic Acid/physiology , Neurons/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cerebral Cortex/physiology , Down-Regulation/drug effects , Female , Gene Expression Regulation, Enzymologic/physiology , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Neurons/drug effects , Neurons/enzymology , Primary Cell Culture , Proteasome Endopeptidase Complex/physiology , Tuberous Sclerosis Complex 2 Protein
SELECTION OF CITATIONS
SEARCH DETAIL