Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
ACS Biomater Sci Eng ; 2024 Jun 17.
Article En | MEDLINE | ID: mdl-38885017

Osteoarthritis (OA) is a chronic joint disease characterized by cartilage imbalance and disruption of cartilage extracellular matrix secretion. Identifying key genes that regulate cartilage differentiation and developing effective therapeutic strategies to restore their expression is crucial. In a previous study, we observed a significant correlation between the expression of the gene encoding casein kinase-2 interacting protein-1 (CKIP-1) in the cartilage of OA patients and OA severity scores, suggesting its potential involvement in OA development. To test this hypothesis, we synthesized a chondrocyte affinity plasmid, liposomes CKIP-1, to enhance CKIP-1 expression in chondrocytes. Our results demonstrated that injection of CAP-Lipos-CKIP-1 plasmid significantly improved OA joint destruction and restored joint motor function by enhancing cartilage extracellular matrix (ECM) secretion. Histological and cytological analyses confirmed that CKIP-1 maintains altered the phosphorylation of the signal transduction molecule SMAD2/3 of the transforming growth factor-ß (TGF-ß) pathway by promoting the phosphorylation of the 8T, 416S sit. Taken together, this work highlights a novel approach for the precise modulation of chondrocyte phenotype from an inflammatory to a noninflammatory state for the treatment of OA and may be broadly applicable to patients suffering from other arthritic diseases.

2.
Brain Behav Immun ; 119: 431-453, 2024 Jul.
Article En | MEDLINE | ID: mdl-38636566

Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.


ATP Binding Cassette Transporter 1 , Macrophages , Spinal Cord Injuries , Animals , ATP Binding Cassette Transporter 1/metabolism , Spinal Cord Injuries/metabolism , Mice , Male , Macrophages/metabolism , Foam Cells/metabolism , Mice, Inbred C57BL , Spinal Cord/metabolism , Mice, Knockout , Disease Models, Animal
4.
Curr Med Sci ; 43(4): 749-758, 2023 Aug.
Article En | MEDLINE | ID: mdl-37558864

OBJECTIVE: This study aims to investigate the effects of hydralazine on inflammation induced by spinal cord injury (SCI) in the central nervous system (CNS) and its mechanism in promoting the structural and functional recovery of the injured CNS. METHODS: A compressive SCI mouse model was utilized for this investigation. Immunofluorescence and quantitative real-time polymerase chain reaction were employed to examine the levels of acrolein, acrolein-induced inflammation-related factors, and macrophages at the injury site and within the CNS. Western blotting was used to evaluate the activity of the phosphoinositide 3-kinase (PI3K)/AKT pathway to study macrophage regulation. The neuropathic pain and motor function recovery were evaluated by glutamic acid decarboxylase 65/67 (GAD65/67), vesicular glutamate transporter 1 (VGLUT1), paw withdrawal response, and Basso Mouse Scale score. Nissl staining and Luxol Fast Blue (LFB) staining were performed to investigate the structural recovery of the injured CNS. RESULTS: Hydralazine downregulated the levels of acrolein, IL-1ß, and TNF-α in the spinal cord. The downregulation of acrolein induced by hydralazine promoted the activation of the PI3K/AKT pathway, leading to M2 macrophage polarization, which protected neurons against SCI-induced inflammation. Additionally, hydralazine promoted the structural recovery of the injured spinal cord area. Mitigating inflammation and oxidative stress by hydralazine in the animal model alleviated neuropathic pain and altered neurotransmitter expression. Furthermore, hydralazine facilitated motor function recovery following SCI. Nissl staining and LFB staining indicated that hydralazine promoted the structural recovery of the injured CNS. CONCLUSION: Hydralazine, an acrolein scavenger, significantly mitigated SCI-induced inflammation and oxidative stress in vivo, modulated macrophage activation, and consequently promoted the structural and functional recovery of the injured CNS.


Neuralgia , Spinal Cord Injuries , Rats , Mice , Animals , Phosphatidylinositol 3-Kinases/metabolism , Acrolein/metabolism , Acrolein/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Hydralazine/pharmacology , Neuralgia/drug therapy , Inflammation/drug therapy , Inflammation/metabolism , Oxidative Stress , Macrophages/metabolism
5.
Acta Physiol (Oxf) ; 239(1): e14018, 2023 09.
Article En | MEDLINE | ID: mdl-37401731

AIM: Under hypobaric hypoxia (HH), the heart triggers various defense mechanisms including metabolic remodeling against lack of oxygen. Mitofusin 2 (MFN2), located at the mitochondrial outer membrane, is closely involved in the regulation of mitochondrial fusion and cell metabolism. To date, however, the role of MFN2 in cardiac response to HH has not been explored. METHODS: Loss- and gain-of-function approaches were used to investigate the role of MFN2 in cardiac response to HH. In vitro, the function of MFN2 in the contraction of primary neonatal rat cardiomyocytes under hypoxia was examined. Non-targeted metabolomics and mitochondrial respiration analyses, as well as functional experiments were performed to explore underlying molecular mechanisms. RESULTS: Our data demonstrated that, following 4 weeks of HH, cardiac-specific MFN2 knockout (MFN2 cKO) mice exhibited significantly better cardiac function than control mice. Moreover, restoring the expression of MFN2 clearly inhibited the cardiac response to HH in MFN2 cKO mice. Importantly, MFN2 knockout significantly improved cardiac metabolic reprogramming during HH, resulting in reduced capacity for fatty acid oxidation (FAO) and oxidative phosphorylation, and increased glycolysis and ATP production. In vitro data showed that down-regulation of MFN2 promoted cardiomyocyte contractility under hypoxia. Interestingly, increased FAO through palmitate treatment decreased contractility of cardiomyocyte with MFN2 knockdown under hypoxia. Furthermore, treatment with mdivi-1, an inhibitor of mitochondrial fission, disrupted HH-induced metabolic reprogramming and subsequently promoted cardiac dysfunction in MFN2-knockout hearts. CONCLUSION: Our findings provide the first evidence that down-regulation of MFN2 preserves cardiac function in chronic HH by promoting cardiac metabolic reprogramming.


Mitochondria , Myocytes, Cardiac , Animals , Mice , Rats , Hydrolases/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Myocytes, Cardiac/metabolism
6.
Exp Neurol ; 363: 114367, 2023 05.
Article En | MEDLINE | ID: mdl-36858281

Spinal cord injury (SCI) results in severe motor and sensory dysfunction with no effective therapy. Spinal cord debris (sp) from injured spinal cord evokes secondary SCI continuously. We and other researchers have previously clarified that it is mainly bone marrow derived macrophages (BMDMs) infiltrating in the lesion epicenter to clear sp, rather than local microglia. Unfortunately, the pro-inflammatory phenotype of these infiltrating BMDMs is predominant which impairs wound healing. Hydralazine, as a potent vasodilator and scavenger of acrolein, has protective effects in many diseases. Hydralazine is also confirmed to promote motor function and hypersensitivity in SCI rats through scavenging acrolein. However, few studies have explored the effects of hydralazine on immunomodulation, as well as spontaneous pain and emotional response, the important syndromes in clinical patients. It remains unclear whether hydralazine affects infiltrating BMDMs after SCI. In this study, we targeted BMDMs to explore the influence of hydralazine on immune cells in a mouse model of SCI, and also investigated the contribution of polarized BMDMs to hydralazine-induced neurological function recovery after SCI in male mice. The adult male mice underwent T10 spinal cord compression. The results showed that in addition to improving motor function and hypersensitivity, hydralazine relieved SCI-induced spontaneous pain and emotional response, which is a newly discovered function of hydralazine. Hydralazine inhibited the recruitments of pro-inflammatory BMDMs and educated infiltrated BMDMs to a more reparative phenotype involving in multiple biological processes associated with SCI pathology, including immune/inflammation response, neurogenesis, lipid metabolism, oxidative stress, fibrosis formation, and angiogenesis, etc. As an overall effect, hydralazine-treated BMDMs loaden with sp partially rescued neurological function after SCI. It is concluded that hydralazine plays an immunomodulation role of educating pro-inflammatory BMDMs to a more reparative phenotype; and hydralazine-educated BMDMs contribute to hydralazine-induced improvement of neurological function in SCI mice, which provides support for drug and cell treatment options for SCI therapy.


Acrolein , Spinal Cord Injuries , Rats , Mice , Male , Animals , Acrolein/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Macrophages/metabolism , Hydralazine/pharmacology , Hydralazine/therapeutic use , Hydralazine/metabolism , Spinal Cord/pathology , Pain/metabolism
7.
J Comp Physiol B ; 193(3): 329-350, 2023 06.
Article En | MEDLINE | ID: mdl-36988658

Loss of bone mass can occur in mammals after prolonged disuse but the situation for hibernators that are in a state of torpor for many months of the year is not yet fully understood. The present study assesses the bone remodeling mechanisms present in Daurian ground squirrels (Spermophilus dauricus) during hibernation as compared with a model of hindlimb disuse. Differences in microstructure, mechanical properties, bone remodeling-related proteins (Runx2, OCN, ALP, RANKL, CTK and MMP-9) and key proteins of Wnt/ß-catenin signaling pathway (GSK-3ß and phospho-ß-catenin) were evaluated in ground squirrels under 3 conditions: summer active (SA) vs. hibernation (HIB) vs. hindlimb unloaded (HLU). The results indicated that the body weight in HLU ground squirrels was lower than the SA group, and the middle tibia diameter in the HLU group was lower than that in SA and HIB groups. The thickness of cortical and trabecular bone in femurs from HLU ground squirrels was lower than in SA and HIB groups. Most parameters of the tibia in the HLU group were lower than those in SA and HIB groups, which indicated cortical bone loss in ground squirrels. Moreover, our data showed that the changes in microscopic parameters in the femur were more obvious than those in the tibia in HLU and HIB ground squirrels. The levels of Runx2 and ALP were lower in HLU ground squirrels than SA and HIB groups. The protein levels of OCN were unchanged in the three groups, but the protein levels of ALP were lower in the HLU group than in SA and HIB groups. RANKL, CTK and MMP-9 protein levels were significantly decreased in tibia of HLU ground squirrels as compared with SA and HIB groups. In addition, the protein expression levels of RANKL, CTK and MMP-9 showed no statistical difference between SA and HIB ground squirrels. Thus, the mechanisms involved in the balance between bone formation and resorption in hibernating and hindlimb unloading ground squirrels may be different. The present study showed that in femur, the Wnt signaling pathway was inhibited, the protein level of GSK-3ß was increased, and the protein expression of phospho-ß-catenin was decreased in the HIB group as compared with the SA group, which indicates that the Wnt signaling pathway has a great influence on the femur of the HIB group. In conclusion, the natural anti-osteoporosis properties of Daurian ground squirrels are seasonal. The squirrels do not experience bone loss when they are inactive for a long time during hibernation, but the mechanisms of anti-osteoporosis did not work in HLU summer active squirrels.


Core Binding Factor Alpha 1 Subunit , Hibernation , Animals , Core Binding Factor Alpha 1 Subunit/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Matrix Metalloproteinase 9/metabolism , beta Catenin/metabolism , Sciuridae/physiology , Hindlimb Suspension , Bone Remodeling , Hindlimb/physiology , Hibernation/physiology
8.
NPJ Regen Med ; 7(1): 42, 2022 Sep 02.
Article En | MEDLINE | ID: mdl-36056026

Reactive astrogliosis usually bears some properties of neural progenitors. How injury triggers astrocyte dedifferentiation remains largely unclear. Here, we report that ischemia induces rapid up-regulation of Wnt2 protein in apoptotic neurons and activation of canonical Wnt signaling in reactive astrocytes in mice, primates and human. Local delivery of Wnt2 shRNA abolished the dedifferentiation of astrocytes while over-expressing Wnt2 promoted progenitor marker expression and neurogenesis. Both the activation of Wnt signaling and dedifferentiation of astrocytes was compromised in ischemic caspase-3-/- cortex. Over-expressing stabilized ß-catenin not only facilitated neurogenesis but also promoted functional recovery in ischemic caspase-3-/- mice. Further analysis showed that apoptotic neurons up-regulated Wnt2 protein via internal ribosome entry site (IRES)-mediated translation. Knocking down death associated protein 5 (DAP5), a key protein in IRES-mediated protein translation, significantly diminished Wnt activation and astrocyte dedifferentiation. Our data demonstrated an apoptosis-initiated Wnt-activating mechanism which triggers astrocytic dedifferentiation and facilitates neuronal regeneration.

9.
J Cell Mol Med ; 26(10): 2908-2920, 2022 05.
Article En | MEDLINE | ID: mdl-35384279

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.


B7-H1 Antigen , Seminiferous Tubules , Animals , B7-H1 Antigen/genetics , Male , Mice , Sertoli Cells , Spermatogenesis/genetics , Spermatogonia , Testis
10.
Transl Res ; 235: 102-114, 2021 09.
Article En | MEDLINE | ID: mdl-33798765

Traumatic brain injury (TBI) is one of the leading causes of disability and paralysis around the world. Secondary injury, characterized by progressive neuronal loss and astrogliosis, plays important roles in the post-TBI cognitive impairment and mood disorder. Unfortunately, there still lacks effective treatments, particularly surgery interferences for it. Recent findings of intercellular mitochondria transfer implies a potential therapeutic value of mitochondria transplantation for TBI, which has not been tested yet. In the present study, we demonstrated a quick dysfunction of mitochondria, up-regulation of Tom20 in the injured cortex and subsequent cognitive and mood impairment. Our data demonstrated that mitochondria derived from allogeneic liver or autogeneic muscle stimulated similar microglial activation in brain parenchyma. In vitro experiments showed that exogenous mitochondria could be easily internalized by neurons, astrocytes, and microglia, except for oligodendrocytes. Mitochondria transplantation effectively rescued neuronal apoptosis, restored the expression of Tom20 and the phosphorylation of JNK. Further analysis revealed that mitochondria transplantation in injured cortex induced a significant up-regulation of BDNF in reactive astrocytes, improved animals' spatial memory and alleviated anxiety. In together, our data indicate that mitochondria transplantation may has the potential of clinical translation for TBI treatment, in combination with surgery.


Astrocytes/metabolism , Brain Injuries, Traumatic/therapy , Brain-Derived Neurotrophic Factor/biosynthesis , Mitochondria/transplantation , Neurons/physiology , Animals , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/psychology , Cell Survival , Cells, Cultured , Endocytosis , Male , Mice , Mice, Inbred C57BL , Mitochondria/physiology
11.
Mol Immunol ; 114: 289-298, 2019 10.
Article En | MEDLINE | ID: mdl-31419705

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.


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
12.
Sci Signal ; 11(553)2018 10 23.
Article En | MEDLINE | ID: mdl-30352949

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.


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.
FASEB J ; 32(7): 3597-3613, 2018 07.
Article En | MEDLINE | ID: mdl-29452570

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.


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
14.
Sci Rep ; 6: 26781, 2016 05 25.
Article En | MEDLINE | ID: mdl-27221778

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.


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 En | MEDLINE | ID: mdl-25520004

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.


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 En | MEDLINE | ID: mdl-26330841

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.
Mol Ther ; 23(6): 1077-1091, 2015 Jun.
Article En | MEDLINE | ID: mdl-25794051

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.


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
18.
Neurotherapeutics ; 11(3): 636-50, 2014 Jul.
Article En | MEDLINE | ID: mdl-24853068

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.


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
19.
PLoS One ; 9(2): e88554, 2014.
Article En | MEDLINE | ID: mdl-24533107

In a previous study, we generated two monoclonal antibodies (mAbs) in mice, aNogoA-N and aNogo-66 mAb, which were raised against recombinant N-terminal fragments of rat NogoA and Nogo-66, respectively. When compared with the commercial rabbit anti-rat NogoA polyclonal antibody (pAb), which can specifically recognise NogoA, the two mAbs were also specific for the NogoA antigen in immunofluorescence histochemical (IHC) staining and Western blot (WB) analysis. Serial truncations of NogoA covering the N-terminal region of NogoA (aa 570-691) and Nogo-66 (aa 1026-1091) were expressed in E. coli. The epitopes recognised by aNogoA-N and aNogo-66 are located in the aa 634-668 and aa 1026-1055 regions of NogoA, respectively. Both mAbs remarkably enhanced the axon growth and branching of cultured hippocampal neurons in vitro. These results suggest that the antibodies that bind to aa 634-668 and aa 1026-1055 of NogoA may have stimulatory effects on axon growth and branching. Additionally, the two mAbs that we generated are specific for NogoA and significantly block NogoA function. In conclusion, two sites in NogoA located within aa 634-668 and aa 1026-1055 are recognised by our two antibodies and are novel and potentially promising targets for repair after central nervous system (CNS) injury.


Antibodies, Monoclonal/chemistry , Axons/physiology , Myelin Proteins/chemistry , Neurons/cytology , Animals , Central Nervous System/injuries , Epitope Mapping , Epitopes/chemistry , GAP-43 Protein/chemistry , Hippocampus/cytology , Immunohistochemistry , Male , Microscopy, Fluorescence , Myelin Sheath/chemistry , Nogo Proteins , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry
20.
Sci China Life Sci ; 55(11): 940-7, 2012 Nov.
Article En | MEDLINE | ID: mdl-23090063

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.


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
...