Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
EMBO Rep ; 24(8): e56635, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37358015

ABSTRACT

Sepsis is a leading cause of in-hospital mortality resulting from a dysregulated response to infection. Novel immunomodulatory therapies targeting macrophage metabolism have emerged as an important focus for current sepsis research. However, understanding the mechanisms underlying macrophage metabolic reprogramming and how they impact immune response requires further investigation. Here, we identify macrophage-expressed Spinster homolog 2 (Spns2), a major transporter of sphingosine-1-phosphate (S1P), as a crucial metabolic mediator that regulates inflammation through the lactate-reactive oxygen species (ROS) axis. Spns2 deficiency in macrophages significantly enhances glycolysis, thereby increasing intracellular lactate production. As a key effector, intracellular lactate promotes pro-inflammatory response by increasing ROS generation. The overactivity of the lactate-ROS axis drives lethal hyperinflammation during the early phase of sepsis. Furthermore, diminished Spns2/S1P signaling impairs the ability of macrophages to sustain an antibacterial response, leading to significant innate immunosuppression in the late stage of infection. Notably, reinforcing Spns2/S1P signaling contributes to balancing the immune response during sepsis, preventing both early hyperinflammation and later immunosuppression, making it a promising therapeutic target for sepsis.


Subject(s)
Macrophages , Sepsis , Humans , Anion Transport Proteins/metabolism , Immunosuppression Therapy , Lactates , Macrophages/metabolism , Reactive Oxygen Species/metabolism
2.
J Cell Mol Med ; 26(10): 2908-2920, 2022 05.
Article in English | MEDLINE | ID: mdl-35384279

ABSTRACT

Spermatogenesis is a cyclical process in which different generations of spermatids undergo a series of developmental steps at a fixed time and finally produce spermatids. Here, we report that overexpression of PD-L1 (B7 homolog1) in the testis causes sperm developmental disorders and infertility in male mice, with severe malformation and sloughing during spermatid development, characterized by disorganized and collapsed seminiferous epithelium structure. PD-L1 needs to be simultaneously expressed on Sertoli cells and spermatogonia to cause spermatogenesis failure. After that, we excluded the influence of factors such as the PD-L1 receptor and humoral regulation, confirming that PD-L1 has an intrinsic function to interact with PD-L1. Studies have shown that PD-L1 not only serves as a ligand but also plays a receptor-like role in signal transduction. PD-L1 interacts with PD-L1 to affect the adhesive function of germ cells, causing malformation and spermatid sloughing. Taken together, these results indicate that PD-L1 can interact with PD-L1 to cause germ cell detachment and male infertility.


Subject(s)
B7-H1 Antigen , Seminiferous Tubules , Animals , B7-H1 Antigen/genetics , Male , Mice , Sertoli Cells , Spermatogenesis/genetics , Spermatogonia , Testis
3.
Glia ; 67(6): 1062-1075, 2019 06.
Article in English | MEDLINE | ID: mdl-30648289

ABSTRACT

Chronic pain is one of the most prevalent chronic diseases in the world. The plastic changes of sensory neurons in dorsal root ganglia (DRG) have been extensively studied as the underlying periphery mechanism. Recent studies revealed that satellite cells, the major glial cells in DRG, also played important roles in the development/modulation of chronic pain. Whether DRG satellite glial cells generate new neurons as their counterparts in enteric nerve ganglia and carotid body do under pathological conditions remains poorly investigated. Here, we report that chronic pain induces proliferation and upregulation of progenitor markers in the sex-determining region Y-box 2 (Sox2)- and platelet-derived growth factor receptor alpha (PDGFRα)-positive satellite glial cells. BrdU incorporation assay revealed the generation of IB4- and CGRP-positive neurons, but not NF200-positive neurons in DRG ipsilateral to injury. Genetic fate tracings showed that PDGFRα-positive cells did not generate neurons, whereas Sox2-positive cells produced both IB4- and CGRP-positive neurons. Interestingly, glial fibrillary acidic protein-positive cells, a subpopulation of Sox2-positive satellites, only gave birth to IB4-positive neurons. Local persistent delivery of tetrodotoxin to the sciatic nerve trunk significantly reduced the pain-induced neurogenesis. Furthermore, patch-clamp studies demonstrated that these glia-derived new neurons could fire action potentials and respond to capsaicin. Taken together, our data demonstrated a chronic pain-induced nociceptive neurogenesis in DRG from Sox2-positive satellite cells, indicating a possible contribution of DRG neurogenesis to the pathology of chronic pain.


Subject(s)
Chronic Pain/metabolism , Ganglia, Spinal/metabolism , Neurogenesis/physiology , SOXB1 Transcription Factors/biosynthesis , Satellite Cells, Perineuronal/metabolism , Animals , Chronic Pain/pathology , Ganglia, Spinal/chemistry , Ganglia, Spinal/pathology , Male , Mice , Mice, Inbred C57BL , SOXB1 Transcription Factors/analysis , Satellite Cells, Perineuronal/chemistry , Satellite Cells, Perineuronal/pathology
4.
FASEB J ; 32(7): 3597-3613, 2018 07.
Article in English | MEDLINE | ID: mdl-29452570

ABSTRACT

Spinster homolog 2 (SPNS2) is the membrane transporter of sphingosine-1-phosphate (S1P), and it participates in several physiologic processes by activating different S1P receptors (S1PRs). However, its functions in the nervous system remain largely unclear. We explored the important role of SPNS2 in the process of retinal morphogenesis using a spns2-deficient rat model. In the absence of the functional SPNS2 transporter, we observed progressively aggravating laminar disorganization of the epithelium at the postnatal stage of retinal development. Disrupted cell polarity, delayed cell-cycle exit of retinal progenitor cells, and insufficient migration of newborn neurons were proposed in this study as potential mechanisms accounting for this structural disorder. In addition, we analyzed the expression profiles of spns2 and s1prs, and proposed that SPNS2 regulated retinal morphogenesis by establishing the S1P level in the eye and activating S1PR3 signaling. These data indicate that SPNS2 is indispensable for normal retinal morphogenesis and provide new insights on the role of S1P in the developing retina using an established in vivo model.-Fang, C., Bian, G., Ren, P., Xiang, J., Song, J., Yu, C., Zhang, Q., Liu, L., Chen, K., Liu, F., Zhang, K., Wu, C., Sun, R., Hu, D., Ju, G., Wang, J. S1P transporter SPNS2 regulates proper postnatal retinal morphogenesis.


Subject(s)
Anion Transport Proteins/genetics , Neurogenesis , Retina/metabolism , Animals , Anion Transport Proteins/metabolism , Cells, Cultured , Lysophospholipids/metabolism , Rats , Rats, Sprague-Dawley , Retina/growth & development , Sphingosine/analogs & derivatives , Sphingosine/metabolism
5.
Mol Ther ; 23(6): 1077-1091, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25794051

ABSTRACT

Spinal cord injury (SCI) frequently provokes serious detrimental outcomes because neuronal regeneration is limited in the central nervous system (CNS). Thus, the creation of a permissive environment for transplantation therapy with neural stem/progenitor cells (NS/PCs) is a promising strategy to replace lost neuronal cells, promote repair, and stimulate functional plasticity after SCI. Macrophages are important SCI-associated inflammatory cells and a major source of secreted factors that modify the lesion milieu. Here, we used conditional medium (CM) from bone marrow-derived M1 or M2 polarized macrophages to culture murine NS/PCs. The NS/PCs showed enhanced astrocytic versus neuronal/oligodendrocytic differentiation in the presence of M1- versus M2-CM. Similarly, cotransplantation of NS/PCs with M1 and M2 macrophages into intact or injured murine spinal cord increased the number of engrafted NS/PC-derived astrocytes and neurons/oligodendrocytes, respectively. Furthermore, when cotransplantated with M2 macrophages, the NS/PC-derived neurons integrated into the local circuitry and enhanced locomotor recovery following SCI. Interesting, engrafted M1 macrophages promoted long-distance rostral migration of NS/PC-derived cells in a chemokine (C-X-C motif) receptor 4 (CXCR4)-dependent manner, while engrafted M2 macrophages resulted in limited cell migration of NS/PC-derived cells. Altogether, these findings suggest that the cotransplantation of NS/PCs together with polarized macrophages could constitute a promising therapeutic approach for SCI repair.


Subject(s)
Cell Differentiation , Cell Movement , Embryonic Stem Cells/transplantation , Macrophages/metabolism , Neural Stem Cells/transplantation , Spinal Cord/cytology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Central Nervous System/pathology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Male , Mice , Mice, Inbred C57BL , Oligodendroglia/cytology , Oligodendroglia/metabolism , Organ Transplantation/methods , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Spinal Cord Injuries/therapy
6.
Front Oncol ; 12: 740051, 2022.
Article in English | MEDLINE | ID: mdl-35127542

ABSTRACT

The tumor microenvironment (TME) is variable across tumor types and has diverse effects on malignant progression, based on the type and number of infiltrating stromal cells. In particular, TME effector genes and their competitive endogenous RNA (ceRNA) networks play a critical role in regulating malignant tumor progression. However, the core effector molecules involved in TME modulation of kidney renal papillary cell carcinoma (KIRP) are poorly understood. To address this question, a cohort containing 233 KIRP patients was derived from The Cancer Genome Atlas (TCGA) database, and the data were processed using the ESTIMATE algorithm. We further evaluated the relationship between immune scores (ISs) and stromal scores (SSs) and disease progression and found that high SSs were associated with a poor prognosis in KIRP. Differentially expressed genes (DEGs) were therefore screened based on SS scores, resulting in 2509 DEGs, including 1668 mRNAs, 783 long noncoding (lnc)RNAs, and 58 micro (mi)RNAs. DEGs were then filtered using the random variance and subjected to hierarchical clustering using EPCLUST. Weighted gene co-expression network analysis (WGCNA) was used to assess the prognostic capacity of these DEGs and identify target ceRNA networks, and lncRNA GUSBP11/miR-432-5p/CAMK2B in the turquoise module was selected as a promising ceRNA network. From this analysis CAMK2B was selected as the core gene predicted to be involved in stromal TMA regulation. We therefore explored the expression and function of CAMK2B in vitro and in vivo and provide evidence that this protein promotes stromal TME remodulation and inhibits proliferation in KIRP. Lastly, we show that vascular endothelial growth factor (VEGF), transforming growth factor (TGF)ß, and close homolog of L1 (CHL1) act as downstream effectors of CAMK2B in KIRP. Thus, in this study, we show that the TME determines prognosis of KIRP patients via the core effector molecule CAMK2B, which mediates both microenvironmental remodeling and tumor progression. Based on these findings, we propose that remodeling of the stromal microenvironment could represent an improved therapeutic approach relative to immunotherapy for KIRP.

7.
Mol Neurobiol ; 59(4): 2642-2655, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35142987

ABSTRACT

Abundant reactive gliosis and neuroinflammation are typical pathogenetic hallmarks of brains in Parkinson's disease (PD) patients, but regulation mechanisms are poorly understood. We are interested in role of programmed death-1 (PD-1) in glial reaction, neuroinflammation and neuronal injury in PD pathogenesis. Using PD mouse model and PD-1 knockout (KO) mice, we designed wild-type-control (WT-CON), WT-1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (WT-MPTP), PD-1-KO-control (KO-CON) and PD-1-KO-MPTP (KO-MPTP), and observed motor dysfunction of animal, morphological distribution of PD-1-positive cells, dopaminergic neuronal injury, glial activation and generation of inflammatory cytokines in midbrains by motor behavior detection, immunohistochemistry and western blot. WT-MPTP mouse model exhibited decrease of PD-1/Iba1-positive microglial cells in the substantia nigra compared with WT-CON mice. By comparison of four groups, PD-1 deficiency showed exacerbation in motor dysfunction of animals, decreased expression of TH protein and TH-positive neuronal protrusions. PD-1 deficiency enhanced microglial activation, production of proinflammatory cytokines like inducible nitric oxide synthase, tumor necrosis factor-α, interleukin-1ß and interleukin-6, and expression and phosphorylation of AKT and ERK1/2 in the substantia nigra of MPTP model. We concluded that PD-1 deficiency could aggravate motor dysfunction of MPTP mouse model by inducing microglial activation and neuroinflammation in midbrains, suggesting that PD-1 signaling abnormality might be possibly involved in PD pathogenesis.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neuroinflammatory Diseases , Parkinson Disease/pathology , Programmed Cell Death 1 Receptor/metabolism
8.
Neuroscience ; 440: 130-145, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32450294

ABSTRACT

Astrocytes are major glial cells critical in assisting the function of the central nervous system (CNS), but the functional changes and regulation mechanism of reactive astrocytes are still poorly understood in CNS diseases. In this study, mouse primary astrocytes were cultured, and inflammatory insult was performed to observe functional changes in astrocytes and the involvement of Notch-PI3K-AKT signaling activation through immunofluorescence, PCR, Western blot, CCK-8, and inhibition experiments. Notch downstream signal Hes-1 was clearly observed in the astrocytes, and Notch signal inhibitor GSI dose-dependently decreased the cleaved Notch-l level without an influence on cell viability. Inflammatory insult of lipopolysaccharide plus interferon-γ (LPS+IFNγ) induced an increase in pro-inflammatory cytokines, that is, iNOS, IL-1ß, IL-6, and TNF, at the protein and mRNA levels in activated astrocytes, which was reduced or blocked by GSI treatment. The cell viability of the astrocytes did not show significant differences among different groups. While an increase in MyD88, NF-кB, and phosphor-NF-кB was confirmed, upregulation of PI3K, AKT, and phosphor-AKT was observed in the activated astrocytes with LPS+IFNγ insult and was reduced by GSI treatment. Inhibitor experiments showed that inhibition of Notch-PI3K-AKT signaling activation reduced the pro-inflammatory cytokine production triggered by LPS+IFNγ inflammatory insult. This study showed that the reactive astrocytes displayed pro-inflammatory adaptability through Notch-PI3K-AKT signaling activation in response to inflammatory stimulation, suggesting that the Notch-PI3K-AKT pathway in reactive astrocytes may serve as a promising target against CNS inflammatory disorders.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Animals , Astrocytes/metabolism , Cells, Cultured , Central Nervous System/metabolism , Cytokines , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
9.
Mol Immunol ; 114: 289-298, 2019 10.
Article in English | MEDLINE | ID: mdl-31419705

ABSTRACT

Programmed cell death 1 ligand 1 (PD-L1) plays a critical role in mediating autoimmune diseases, including type I diabetes (T1D). B cells are important antigen-presenting cells (APCs) that make a major contribution to T1D development. However, B cells expressing low levels of PD-L1 that infiltrate insulitic islets in NOD mice may not inhibit effector T cells and prevent T1D. Here, we generated PD-L1 transgenic NOD (NOD.PD-L1Tg) mice, in which most immune cells overexpress PD-L1, to investigate the ability of B cells overexpressing PD-L1 to inhibit diabetic CD4+ T cells and prevent T1D. The severity of insulitis in NOD.PD-L1Tg mice was significantly lower than in NOD mice and none developed diabetes. In addition, there were no differences in expression of activity markers by APCs following LPS stimulation between two groups. In vitro studies revealed that B cells expressing high levels of PD-L1 inhibited proliferation of and cytokine secretion by pre-diabetic CD4+ T cells, whereas in vivo studies showed that NOD/SCID mice receiving diabetic CD4+ T cells mixed with B cells overexpressing PD-L1 became diabetic at a slower rate. Thus, we propose that B cells showing high expression of PD-L1 protect NOD mice against T1D and downregulate diabetogenic CD4+ T cells.


Subject(s)
B-Lymphocytes/metabolism , B7-H1 Antigen/metabolism , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred NOD/metabolism , Animals , Autoimmune Diseases/metabolism , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Down-Regulation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Prediabetic State/metabolism
10.
J Neurochem ; 105(1): 203-16, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18021294

ABSTRACT

Neurokinin peptides neurokinin-1 (NK1), neurokinin-3 (NK3), and related receptors are abundantly distributed in the substantia nigra (SN) and evidenced by their possible roles in the Parkinson's disease. Differential intervention roles of NK3 on kainic acid (KA)-induced neuronal injury in the SN of mice were thus in vitro and in vivo studied by Fluoro-Jade C (FJC) staining, immunohistochemistry to tyrosine hydroxylase (TH) or phospho-NMDA receptor, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. It revealed that (i) in contrast to protective effect of NK1 agonist septide that reduced FJC-positive degenerative neurons and lesion volume insulted by KA, NK3 agonist senktide significantly increased FJC-positive ones and lesion volume, and this effect was sufficiently reversed by NK3 antagonist SB218795; (ii) similarly, senktide reduced TH-positive neurons and this effect was antagonized by SB218795, but septide increased TH-positive ones; (iii) mechanistic observation showed differential influences of NK1 and NK3 agonists on phosphorylated-NMDA receptor subunit 1 (phospho-NMDAR1) and glial fibrillary acidic protein-expressing astrocytes, i.e. senktide enhanced of NMDA receptor phosphorylation and astrocyte activity, while septide reduced NMDA receptor phosphorylation and astrocytic response; (iv) cell culture further confirmed the exacerbating effect of NK3 agonist on KA-induced lesion of nigral cells or dopaminergic neurons, in which administration of senktide alone did not show significant cell toxicity. This study presents new evidence that neurokinin NK3 instead of NK1 synergistically exacerbate excitotoxic neuronal degeneration in the SN in a dose-dependent manner and possibly through modulation of NMDA receptor phosphorylation and astrocyte activity, suggesting their potential significance in novel pharmaceutical therapy against Parkinson's disease.


Subject(s)
Kainic Acid , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Receptors, Neurokinin-1/physiology , Receptors, Neurokinin-3/physiology , Substantia Nigra/pathology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Drug Synergism , Fluoresceins , Male , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/metabolism , Neurokinin-1 Receptor Antagonists , Neurons/drug effects , Neurons/metabolism , Organic Chemicals , Peptide Fragments/pharmacology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/pharmacology , Quinolines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neurokinin-3/antagonists & inhibitors , Substance P/analogs & derivatives , Substance P/pharmacology , Substantia Nigra/drug effects , Tetrazolium Salts , Thiazoles
11.
Neurochem Int ; 53(3-4): 56-62, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18639597

ABSTRACT

Parkinson's disease is a common and severe debilitating neurological disease that results from massive and progressive degenerative death of dopamine neurons in the substantia nigra, but the mechanisms of neuronal degeneration and disease progression remains largely obscure. We are interested in possible implications of low-affinity p75 neurotrophin receptor (p75NTR), which may mediate neuronal apoptosis in the central nervous system, in triggering cell death of the nigral dopamine neurons. The RT-PCR and immunohistochemistry were carried out to detect if p75NTR is expressed in these nigral neurons and up-regulated by kainic acid (KA) insult in adult rats. It revealed p75NTR-positive immunoreactivity in the substantia nigra, and co-localization of p75NTR and tyrosine hydroxylase (TH) was found in a large number of substantia nigra neurons beside confirmation of p75NTR in the choline acetyltransferase (ChAT)-positive forebrain neurons. Cell count data further indicated that about 47-100% of TH-positive nigral neurons and 98-100% of ChAT-positive forebrain neurons express p75NTR. More interestingly, significant increasing in both p75NTR mRNA and p75NTR-positive neurons occurred rapidly following KA insult in the substantia nigra of animal model. The present study has provided first evidence on p75NTR expression and KA-inducing p75NTR up-regulation in substantia nigra neurons in rodent animals. Taken together with previous data on p75NTR functions in neuronal apoptosis, this study also suggests that p75NTR may play important roles in neuronal cell survival or excitotoxic degeneration of dopamine neurons in the substantia nigra in pathogenesis of Parkinson's disease in human beings.


Subject(s)
Dopamine/metabolism , Neurons/metabolism , Parkinsonian Disorders/metabolism , Receptor, Nerve Growth Factor/genetics , Substantia Nigra/metabolism , Acetylcholine/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/physiology , Basal Nucleus of Meynert/cytology , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Count , Cell Survival/drug effects , Cell Survival/physiology , Choline O-Acetyltransferase/metabolism , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Parkinsonian Disorders/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/drug effects , Substantia Nigra/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Sci Signal ; 11(553)2018 10 23.
Article in English | MEDLINE | ID: mdl-30352949

ABSTRACT

In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. We identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal-regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)-c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Our results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.


Subject(s)
ErbB Receptors/physiology , Eyelids/embryology , Eyelids/physiology , Lysophospholipids/chemistry , Sphingosine/analogs & derivatives , ADAM10 Protein/physiology , ADAM17 Protein/physiology , Animals , Animals, Genetically Modified , Cell Movement , Fatty Acid Transport Proteins/genetics , Fatty Acid Transport Proteins/metabolism , Gene Expression Regulation, Developmental , Keratinocytes/cytology , Ligands , Phenotype , Rats , Signal Transduction , Sphingosine/chemistry , Transcriptional Activation
13.
Brain Res ; 1150: 55-61, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17397812

ABSTRACT

Fluoro-Jade C, a new-developed fluorescent dye, has been successfully applied for identification of neuronal degeneration in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-tetrahydro pyridine (MPTP)-treated mice in the present study. The animal model was first prepared by intraperitoneal injection of neurotoxicant MPTP that can specifically induce degeneration of dopamine neurons in the substantia nigra of C57BL/6 mice. Fluoro-Jade C was then utilized to stain the midbrain sections and semiquantitation analysis was carried out in comparison with controls. It revealed that Fluoro-Jade C-positive cells showed strong green color in neuronal profile and were observed in the substantia nigra of MPTP-treated mice whereas they were not detected in that of controls. The Fluoro-Jade C-positive cells were mostly shrunken or smaller-sized in their cell bodies in comparing with that of normal dopamine neurons of controls. In the midbrain of MPTP-treated mice, Fluoro-Jade C-positive neuronal cells were exclusively distributed in the substantia nigra pars compacta, but rarely seen in the ventral tegemental area where dopamine neurons were numerously distributed. Double-labeling experiments indicated that a population of Fluoro-Jade C-positive cells (23%) exhibited neuron-specific nuclear protein-immunoreactivity and none of them showed immunoreactivity to glial cell marker glial fibrillary acid protein. However, most of Fluoro-Jade C-positive degenerative neurons (98%) lost their immunoreactivity to dopaminergic marker tyrosine hydroxylase in the substantia nigra of MPTP-treated mice. Taken together with previous observations, this study has presented that Fluoro-Jade C can be sensitively and specifically utilized to identify the neuronal degeneration in the substantia nigra of rodent animals receiving MPTP insult.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Nerve Degeneration/diagnosis , Neurotoxins/pharmacology , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism , Animals , Cell Count/methods , Fluoresceins , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Neurons/drug effects , Neurons/metabolism , Organic Chemicals , Substantia Nigra/cytology
14.
Sci Rep ; 6: 26781, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27221778

ABSTRACT

A single microRNA (miRNA) can regulate expression of multiple proteins, and expression of an individual protein may be controlled by numerous miRNAs. This regulatory pattern strongly suggests that synergistic effects of miRNAs play critical roles in regulating biological processes. miR-9 and miR-124, two of the most abundant miRNAs in the mammalian nervous system, have important functions in neuronal development. In this study, we identified the small GTP-binding protein Rap2a as a common target of both miR-9 and miR-124. miR-9 and miR-124 together, but neither miRNA alone, strongly suppressed Rap2a, thereby promoting neuronal differentiation of neural stem cells (NSCs) and dendritic branching of differentiated neurons. Rap2a also diminished the dendritic complexity of mature neurons by decreasing the levels of pAKT and pGSK3ß. Our results reveal a novel pathway in which miR-9 and miR-124 synergistically repress expression of Rap2a to sustain homeostatic dendritic complexity during neuronal development and maturation.


Subject(s)
Gene Expression Regulation/genetics , MicroRNAs/genetics , Neurogenesis/genetics , rap GTP-Binding Proteins/antagonists & inhibitors , 3' Untranslated Regions/genetics , Animals , Dendrites/ultrastructure , Glycogen Synthase Kinase 3 beta/physiology , HEK293 Cells , Homeostasis , Humans , Mice , Neural Stem Cells/cytology , Neurons/ultrastructure , Proto-Oncogene Proteins c-akt/physiology , Recombinant Fusion Proteins/metabolism , Signal Transduction , rap GTP-Binding Proteins/genetics , rap GTP-Binding Proteins/physiology
15.
Mol Neurobiol ; 53(1): 662-676, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25520004

ABSTRACT

Inflammatory reactions are the most critical pathological processes occurring after spinal cord injury (SCI). Activated microglia/macrophages have either detrimental or beneficial effects on neural regeneration based on their functional polarized M1/M2 subsets. However, the mechanism of microglia/macrophage polarization to M1/M2 at the injured spinal cord environment remains unknown. In this study, wild-type (WT) or aldose reductase (AR)-knockout (KO) mice were subjected to SCI by a spinal crush injury model. The expression pattern of AR, behavior tests for locomotor activity, and lesion size were assessed at between 4 h and 28 days after SCI. We found that the expression of AR is upregulated in microglia/macrophages after SCI in WT mice. In AR KO mice, SCI led to smaller injury lesion areas compared to WT. AR deficiency-induced microglia/macrophages induce the M2 rather than the M1 response and promote locomotion recovery after SCI in mice. In the in vitro experiments, microglia cell lines (N9 or BV2) were treated with the AR inhibitor (ARI) fidarestat. AR inhibition caused 4-hydroxynonenal (HNE) accumulation, which induced the phosphorylation of the cAMP response element-binding protein (CREB) to promote Arg1 expression. KG501, the specific inhibitor of phosphorylated CREB, could cancel the upregulation of Arg1 by ARI or HNE stimulation. Our results suggest that AR works as a switch which can regulate microglia by polarizing cells to either the M1 or the M2 phenotype under M1 stimulation based on its states of activity. We suggest that inhibiting AR may be a promising therapeutic method for SCI in the future.


Subject(s)
Aldehyde Reductase/biosynthesis , Cell Polarity/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Macrophages/metabolism , Microglia/metabolism , Spinal Cord Injuries/metabolism , Aldehyde Reductase/deficiency , Animals , Cell Line , Cell Polarity/drug effects , Cells, Cultured , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Tumor Necrosis Factor-alpha/pharmacology
16.
Neural Regen Res ; 10(7): 1147-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26330841

ABSTRACT

MicroRNA-124 (miR-124) is abundantly expressed in neurons in the mammalian central nervous system, and plays critical roles in the regulation of gene expression during embryonic neurogenesis and postnatal neural differentiation. However, the expression profile of miR-124 after spinal cord injury and the underlying regulatory mechanisms are not well understood. In the present study, we examined the expression of miR-124 in mouse brain and spinal cord after spinal cord injury using in situ hybridization. Furthermore, the expression of miR-124 was examined with quantitative RT-PCR at 1, 3 and 7 days after spinal cord injury. The miR-124 expression in neurons at the site of injury was evaluated by in situ hybridization combined with NeuN immunohistochemical staining. The miR-124 was mainly expressed in neurons throughout the brain and spinal cord. The expression of miR-124 in neurons significantly decreased within 7 days after spinal cord injury. Some of the neurons in the peri-lesion area were NeuN(+)/miR-124(-). Moreover, the neurons distal to the peri-lesion site were NeuN(+)/miR-124(+). These findings indicate that miR-124 expression in neurons is reduced after spinal cord injury, and may reflect the severity of spinal cord injury.

17.
Neurotherapeutics ; 11(3): 636-50, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24853068

ABSTRACT

The inflammatory response following spinal cord injury (SCI) involves the activation of resident microglia and the infiltration of macrophages. Macrophages and microglia can be polarized into the classically activated proinflammatory M1 phenotype or the alternatively activated anti-inflammatory M2 phenotype. Programmed cell death 1 (PD-1) is a critical immune inhibitory receptor involved in innate and adaptive immune responses. However, whether PD-1 is involved in the modulation of macrophage/microglial polarization is unknown. In this study, the mRNA levels of pd1 gradually increased after SCI, and PD-1 protein was found in macrophages/microglia in injured spinal cord sections. PD-1 knockout (KO) mice showed poor locomotor recovery after spinal cord crushing compared with wild-type mice. M1-type macrophages/microglia accumulated in greater numbers in the injured spinal cord of PD-1-KO mice. Under polarized stimulation, induced expression of PD-1 occurred in cultured macrophages and microglia. PD-1 suppressed M1 polarization by reducing the phosphorylation of signal transducer and activator of transcription 1 (STAT1) and promoted M2 polarization by increasing STAT6 phosphorylation. In PD-1-KO mice, the M1 response was enhanced via the activation of STAT1 and nuclear factor-kappa B. Furthermore, PD-1 played various roles in phagocytosis in macrophages and microglia. Therefore, our results suggest that PD-1 signaling plays an important role in the regulation of macrophage/microglial polarization. Thus, deregulated PD-1 signaling may induce the polarization of macrophages/microglia toward the M1 phenotype. Overall, our results provide new insights into the modulatory mechanisms of macrophage/microglial polarization, thereby possibly facilitating the development of new therapies for SCI via the regulation of macrophage/microglial polarization through PD-1 signaling.


Subject(s)
Macrophages/cytology , Macrophages/metabolism , Microglia/cytology , Microglia/metabolism , Programmed Cell Death 1 Receptor/metabolism , Spinal Cord Injuries/metabolism , Animals , Cell Polarity , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phenotype , Primary Cell Culture , Programmed Cell Death 1 Receptor/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism
18.
Sci China Life Sci ; 55(11): 940-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23090063

ABSTRACT

The inhibitory co-receptor programmed death 1 (PD-1, encoded by pdcd1) and its two ligands PD-L1 and PD-L2 comprise an important immune inhibitory signaling pathway for defense against microbes and for self-tolerance. Unlike other members of the B7-CD28 family, expression of PD-L1 and PD-L2 is not limited to the immune system. In this study, we determined that a polyclonal antibody (pAb) (R&D Systems) against extracellular domains of mouse PD-L2 (mPD-L2) could recognize antigen(s) in diverse mouse tissues, including the anterior and intermediate pituitary gland, olfactory bulbs and olfactory epithelium, tongue epithelium, keratinized epithelial cells and skin and whisker hair follicles. These findings differed from previous reports of mPD-L2 localization. Reverse transcription PCR and Western blot analyses, however, were unable to detect any mPD-L2 transcripts or proteins of the 25-kD predicted molecular weight in RNA and protein extracts, respectively, from the above tissues, suggesting that the anti-mPD-L2 pAb cross-reacts with certain novel antigen(s). Developmental studies revealed that the earliest expression of mPD-L2-like antigen was in the olfactory epithelium at embryonic day 12.5 (E12.5). At E14.5, mPD-L2-like antigen was present in the skin, tongue and follicles of the skin and whiskers. The distribution patterns of mPD-L2-like antigen remained similar from E18.5 to adulthood. The results of bioinformatic analysis and other experiments suggested neural cell adhesion molecule and hemicentin-1 as candidate proteins with cross-reactivity to the anti-mPD-L2 pAb. These results demonstrate that care is required in interpreting staining patterns generated when anti-PD-L2 pAb is used to locate PD-L2-expressing cells in the central nervous system and epithelial tissues, such as the olfactory epithelium. In addition, this anti-PD-L2 pAb may be used as an alternative antibody for labeling the olfactory epithelium during embryonic development in mice.


Subject(s)
Antibodies/immunology , Cross Reactions , Programmed Cell Death 1 Receptor/immunology , Animals , Base Sequence , Blotting, Western , DNA Primers , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(4): 425-7, 2011 Apr.
Article in Zh | MEDLINE | ID: mdl-21481323

ABSTRACT

AIM: To detect and characterize of 6 monoclonal antibodies (mAbs) against different epitopes of rat Nogo-A molecule in immunohistochemistry to decide their applications in futrue. Four mAbs against Nogo66 fragment are named Nogo66-1, Nogo66-2, Nogo66-3 and Nogo66-4. The rest of 2 mAbs against N-termial 570-691aa fragment are named NogoN-1 and NogoN-2. METHODS: The immunofluorescence staining was used to detect the reactivity and specificity of those 6 mAbs in spinal tissue sections of rat. RESULTS: All 6 mAbs were double-labelled with commercial rabbit anti-Rat Nogo-A polyclonal antibody (PcAb) in spinal cord sections respecitvely. All 6 mAbs were colocalization with MBP respectively. However Nogo66-3 and NogoN-1 could also be double-staining with GFAP respectively. CONCLUSION: Nogo66-1, Nogo66-2, Nogo66-4 and NogoN-2 could recognize specifically in Nogo-A protein of tissues in immunohistochemical methods.


Subject(s)
Antibodies, Monoclonal/immunology , Myelin Proteins/immunology , Animals , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred BALB C , Myelin Proteins/analysis , Nogo Proteins , Rats
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 26(3): 231-4, 2010 Mar.
Article in Zh | MEDLINE | ID: mdl-20230685

ABSTRACT

AIM: To analysis the role of T lymphocytes in spinal cord regeneration by comparing the recovery of movement and the morphological changes of injury area between BALB/c and DO11.10 transgenic mice. METHODS: Producing a crush injury model of spinal cord with special forceps. Analyze the changes of spinal cord injury area with H&E and GFAP, CD11b and lymphocytic immunohistochemical staining. Evaluate the recovery of movement function with Basso-Beattie-Bresnahan (BBB) locomotion testing system at 0, 7, 14 and 21 day post-injury (dpi). RESULTS: There were thicker and fastened glial scar at 21 dpi in the BALB/c mice but not in DO11.10 mice. The number of macrophages/microglia infiltrated in spinal cord injury area were more in DO11.10 than that in BALB/c mice at 14 dpi. The numbers of T lymphocytes infiltrated in spinal cord injury area were less in DO11.10 than that in BALB/c mice at 21 dpi. In addition, compare to BALB/c mice, the locomotion movement recovery of DO11.10 mice were much more significant within 3 weeks after spinal cord injury by BBB scoring system. CONCLUSION: The infiltrated autoimmune activation T lymphocytes which specifically react to neural antigens are not beneficial to recovery of movement after spinal cord injury in mice.


Subject(s)
Locomotion , Spinal Cord Injuries/physiopathology , Animals , Disease Models, Animal , Humans , Leukemic Infiltration , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Spinal Cord Injuries/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL