Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Neurosci Lett ; 824: 137691, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38373630

RESUMO

Enhancing axonal regeneration is one of the most important processes in treating nerve injuries. Both magnetic and electrical stimulation have the effect of promoting nerve axon regeneration. But few study has investigated the effects of trans-spinal magnetic stimulation (TsMS) combined with electroacupuncture (EA) on nerve regeneration in rats with sciatic nerve injury. In this study, we compared the improvement of neurological function in rats with sciatic nerve crush injuries after 4 weeks of different interventions (EA, TsMS, or TsMS combined with EA). We further explored the morphological and molecular biological alterations following sciatic nerve injury by HE, Masson, RT-PCR, western blotting, immunofluorescence staining and small RNA transcriptome sequencing. The results showed that TsMS combined with EA treatment significantly promoted axonal regeneration, increased the survival rate of neurons, and suppressed denervation atrophy of the gastrocnemius muscle. Subsequent experiments suggested that the combination treatment may play an active role by mediating the miR-539-5p/Sema3A/PlexinA1 signaling axis.


Assuntos
Eletroacupuntura , MicroRNAs , Traumatismos dos Nervos Periféricos , Neuropatia Ciática , Ratos , Animais , Ratos Sprague-Dawley , Semaforina-3A/farmacologia , Axônios , Regeneração Nervosa/fisiologia , Nervo Isquiático/lesões , Neuropatia Ciática/terapia , Traumatismos dos Nervos Periféricos/terapia , MicroRNAs/genética , MicroRNAs/farmacologia
2.
Int J Oral Sci ; 16(1): 5, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38238300

RESUMO

Bone formation and deposition are initiated by sensory nerve infiltration in adaptive bone remodeling. Here, we focused on the role of Semaphorin 3A (Sema3A), expressed by sensory nerves, in mechanical loads-induced bone formation and nerve withdrawal using orthodontic tooth movement (OTM) model. Firstly, bone formation was activated after the 3rd day of OTM, coinciding with a decrease in sensory nerves and an increase in pain threshold. Sema3A, rather than nerve growth factor (NGF), highly expressed in both trigeminal ganglion and the axons of periodontal ligament following the 3rd day of OTM. Moreover, in vitro mechanical loads upregulated Sema3A in neurons instead of in human periodontal ligament cells (hPDLCs) within 24 hours. Furthermore, exogenous Sema3A restored the suppressed alveolar bone formation and the osteogenic differentiation of hPDLCs induced by mechanical overload. Mechanistically, Sema3A prevented overstretching of F-actin induced by mechanical overload through ROCK2 pathway, maintaining mitochondrial dynamics as mitochondrial fusion. Therefore, Sema3A exhibits dual therapeutic effects in mechanical loads-induced bone formation, both as a pain-sensitive analgesic and a positive regulator for bone formation.


Assuntos
Osteogênese , Semaforina-3A , Humanos , Remodelação Óssea , Diferenciação Celular , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Gânglio Trigeminal/metabolismo
3.
Arch Gerontol Geriatr ; 117: 105260, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37979338

RESUMO

OBJECTIVES: Exercise training plays a significant role in preventing the destruction of central nerve neurons and muscle atrophy. The purpose of the present study was to investigate the effect of a period of swimming training on the expression of Neural cell adhesion molecule (NCAM), Semaphorin 3A (SEMA3A), and Profilin-1 (PFN1) proteins in the gastrocnemius muscle of Alzheimer-like phenotype rats. METHODS & MATERIALS: 32 Wistar males were (6 weeks of age) divided into four groups: Healthy Control (HC), Alzheimer-like phenotype's Control (AC), Healthy Training (HT), and Alzheimer-like phenotype's Training (AT). Alzheimer-like phenotypes were induced by beta-amyloid injection in the hippocampus. The training program consisted of 20 swimming sessions. Gastrocnemius muscle was removed after the intervention, and NCAM, SEMA3A, and PFN1 proteins were measured by the immunohistoflorescent method. RESULTS: The results showed that SEMA3A was increased (p = 0.001), and NCAM (p = 0.001), and PFN1 (p = 0.001) were decreased in AC compared to the HC group. Also, the results showed that NCAM (p = 0.001) and Pfn1 (p = 0.002) increased in the HT group compared to HC, and the NCAM (p = 0.001) and Pfn1 (p = 0.002) in AT group compared to AC (p = 0.001) increased significantly, while SEMA3A was reduced in the HT group compared to HC (p = 0.001) and AT group compared to AC (p = 0.001) CONCLUSION: Swimming effectively improves axon regeneration and neuronal formation in motor neurons and, therefore, can be an effective intervention to prevent and control the complications of Alzheimer-like phenotype.


Assuntos
Doença de Alzheimer , Natação , Masculino , Humanos , Ratos , Animais , Ratos Wistar , Natação/fisiologia , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Axônios/metabolismo , Regeneração Nervosa , Músculo Esquelético/metabolismo , Moléculas de Adesão de Célula Nervosa/genética , Moléculas de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/farmacologia , Profilinas/farmacologia
4.
JCI Insight ; 8(21)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37788099

RESUMO

Glioblastoma (GBM) is the most lethal brain cancer with a dismal prognosis. Stem-like GBM cells (GSCs) are a major driver of GBM propagation and recurrence; thus, understanding the molecular mechanisms that promote GSCs may lead to effective therapeutic approaches. Through in vitro clonogenic growth-based assays, we determined mitogenic activities of the ligand molecules that are implicated in neural development. We have identified that semaphorin 3A (Sema3A), originally known as an axon guidance molecule in the CNS, promotes clonogenic growth of GBM cells but not normal neural progenitor cells (NPCs). Mechanistically, Sema3A binds to its receptor neuropilin-1 (NRP1) and facilitates an interaction between NRP1 and TGF-ß receptor 1 (TGF-ßR1), which in turn leads to activation of canonical TGF-ß signaling in both GSCs and NPCs. TGF-ß signaling enhances self-renewal and survival of GBM tumors through induction of key stem cell factors, but it evokes cytostatic responses in NPCs. Blockage of the Sema3A/NRP1 axis via shRNA-mediated knockdown of Sema3A or NRP1 impeded clonogenic growth and TGF-ß pathway activity in GSCs and inhibited tumor growth in vivo. Taken together, these findings suggest that the Sema3A/NRP1/TGF-ßR1 signaling axis is a critical regulator of GSC propagation and a potential therapeutic target for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Glioblastoma/patologia , Neuropilina-1/genética , Neoplasias Encefálicas/patologia , Fator de Crescimento Transformador beta
5.
Cell Biochem Biophys ; 81(3): 543-552, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37421591

RESUMO

Genetically engineered stem cells, not only acting as vector delivering growth factors or cytokines but also exhibiting improved cell properties, are promising cells for periodontal tissue regeneration. Sema3A is a power secretory osteoprotective factor. In this study, we aimed to construct Sema3A modified periodontal ligament stem cells (PDLSCs) and evaluated their osteogenic capability and crosstalk with pre-osteoblasts MC3T3-E1. First, Sema3A modified PDLSCs was constructed using lentivirus infection system carrying Sema3A gene and the transduction efficiency was analyzed. The osteogenic differentiation and proliferation of Sema3A-PDLSCs was evaluated. Then, MC3T3-E1 was directly co-cultured with Sema3A-PDLSCs or cultured in condition medium of Sema3A-PDLSCs and the osteogenic ability of MC3T3-E1 was assessed. The results showed that Sema3A-PDLSCs expressed and secreted upregulated Sema3A protein, which confirmed successful construction of Sema3A modified PDLSCs. After osteogenic induction, Sema3A-PDLSCs expressed upregulated ALP, OCN, RUNX2, and SP7 mRNA, expressed higher ALP activity, and produced more mineralization nodes, compared with Vector-PDLSCs. Whereas, there was no obvious differences in proliferation between Sema3A-PDLSCs and Vector-PDLSCs. MC3T3-E1 expressed upregulated mRNA of ALP, OCN, RUNX2, and SP7 when directly co-cultured with Sema3A-PDLSCs than Vector-PDLSCs. MC3T3-E1 also expressed upregulated osteogenic markers, showed higher ALP activity, and produced more mineralization nodes when cultured using condition medium of Sema3A-PDLSCs instead of Vector-PDLSCs. In conclusion, our results indicated that Sema3A modified PDLSCs showed enhanced osteogenic capability, and also facilitated differentiation of pre-osteoblasts.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Osteogênese , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Ligamento Periodontal , RNA Mensageiro/metabolismo , Semaforina-3A/genética , Semaforina-3A/farmacologia , Semaforina-3A/metabolismo , Células-Tronco/metabolismo , Animais , Camundongos
6.
J Bone Miner Res ; 38(8): 1175-1191, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37221130

RESUMO

miR-196b-5p plays a role in various malignancies. We have recently reported its function in regulating adipogenesis. However, it remains to be clarified whether and how miR-196b-5p affects bone cells and bone homeostasis. In this study, in vitro functional experiments showed an inhibitory effect of miR-196b-5p on osteoblast differentiation. Mechanistic explorations revealed that miR-196b-5p directly targeted semaphorin 3a (Sema3a) and inhibited Wnt/ß-catenin signaling. SEMA3A attenuated the impaired osteogenesis induced by miR-196b-5p. Osteoblast-specific miR-196b transgenic mice showed significant reduction of bone mass. Trabecular osteoblasts were reduced and bone formation was suppressed, whereas osteoclasts, marrow adipocytes, and serum levels of bone resorption markers were increased in the transgenic mice. The osteoblastic progenitor cells from the transgenic mice had decreased SEMA3A levels and exhibited retarded osteogenic differentiation, whereas those marrow osteoclastic progenitors exhibited enhanced osteoclastogenic differentiation. miR-196b-5p and SEMA3A oppositely regulated the expression of receptor activator of nuclear factor-κB ligand and osteoprotegerin. The calvarial osteoblastic cells expressing the transgene promoted osteoclastogenesis, whereas the osteoblasts overexpressing Sema3a inhibited it. Finally, in vivo transfection of miR-196b-5p inhibitor to the marrow reduced ovariectomy-induced bone loss in mice. Our study has identified that miR-196b-5p plays a key role in osteoblast and osteoclast differentiation and regulates bone homeostasis. Inhibition of miR-196b-5p may be beneficial for amelioration of osteoporosis. © 2023 American Society for Bone and Mineral Research (ASBMR).


Assuntos
MicroRNAs , Osteoclastos , Animais , Feminino , Camundongos , Diferenciação Celular , Homeostase , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia
7.
Inflammation ; 46(3): 876-891, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36598593

RESUMO

Macrophages actively participate in immunomodulatory processes throughout periodontal inflammation. Regulation of M1/M2 polarization affects macrophage chemokine and cytokine secretion, resulting in a distinct immunological status that influences prognosis. Semaphorin 3A (Sema3A), a neurite growth factor, exerts anti-inflammatory effects. In this study, we investigated the immunomodulation of Sema3A on macrophage-related immune responses in vivo and in vitro. Topical medications of Sema3A in mice with periodontitis alleviated inflammatory cell infiltration into gingival tissue and reduced areas with positive IL-6 and TNFα expression. We observed that the positive area with the M2 macrophage marker CD206 increased and that of the M1 macrophage marker iNOS decreased in Sema3A-treated mice. It has been postulated that Sema3A alleviates periodontitis by regulating alternative macrophage activation. To understand the mechanism underlying Sema3A modulation of macrophage polarization, an in vitro macrophage research model was established with RAW264.7 cells, and we demonstrated that Sema3A promotes LPS/IFNγ-induced M1 macrophages to polarize into M2 macrophages and activates the PI3K/AKT/mTOR signaling pathways. Inhibition of the PI3K signaling pathway activation might reduce anti-inflammatory activity and boost the expression of the inflammatory cytokines, iNOS, IL-12, TNFα, and IL-6. This study indicated that Sema3A might be a feasible drug to regulate alternative macrophage activation in the inflammatory response and thus alleviate periodontitis.


Assuntos
Periodontite , Semaforina-3A , Camundongos , Animais , Semaforina-3A/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Ativação de Macrófagos , Interleucina-6/farmacologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Anti-Inflamatórios/farmacologia , Periodontite/tratamento farmacológico
8.
Curr Eye Res ; 48(1): 70-79, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36271834

RESUMO

PURPOSE: This study aimed to explore the effect of the Semaphorin3A (Sema3A)/Neuropilin-1 (Nrp-1) pathway on Müller cell activities and endoplasmic reticulum (ER) stress induced by high glucose (HG) in vitro. METHODS: The primary Müller cells of C57BL/6J mice were isolated and cultured in normal or high glucose medium. The expression of endogenous Sema3A and its coreceptor Nrp-1 was measured by Western blot. Müller cells were incubated with exogenous recombinant Sema3A protein or transfected with lentiviral vectors expressing small hairpin RNA (shRNA) to knock down the expression of endogenous Sema3A. The proliferation of Müller cells was detected by CCK-8 assay and EdU staining. The migratory ability was detected by the Transwell migration assay. The level of endoplasmic reticulum (ER) stress was analyzed through the detection of GRP78/BiP, IRE1α, phosphorylated IRE1αS724 (p-IRE1αS724), and the splicing rate of XBP1 (XBP1s/XBP1) by using immunofluorescence, Western blot or quantitative polymerase chain reaction (qPCR). RESULTS: HG induced the upregulation of endogenous Sema3A and Nrp-1 receptors in Müller cells. The expression of GRP78/BiP and IRE1α was upregulated by HG, with an increased splicing rate of XBP1. Exogenous Sema3A inhibited HG-induced Müller cell proliferation, migration, and GRP78/BiP-IRE1α-XBP1 axis activation. Knockdown of Sema3A promoted proliferation, migration, and ER stress induced by high glucose in Müller cells. CONCLUSION: Sema3A inhibited the increased proliferative and migratory activities induced by high glucose by attenuating ER stress in Müller cells.


Assuntos
Proteínas Serina-Treonina Quinases , Semaforina-3A , Animais , Camundongos , Apoptose , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Endorribonucleases/metabolismo , Células Ependimogliais/metabolismo , Glucose/farmacologia , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Semaforina-3A/farmacologia
9.
J Diabetes Investig ; 14(2): 193-204, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36583231

RESUMO

AIMS/INTRODUCTION: Long non-coding RNAs (lncRNAs) exert essential functions in the pathogenesis of diabetic nephropathy (DN). LncRNA T-cell factor 7 (TCF7) and semaphorin-3A (SEMA3A) have been found to be involved in the progression of diabetic nephropathy. However, whether the effect of TCF7 on the pathogenesis of diabetic nephropathy is mediated by SEMA3A remains unclear. MATERIALS AND METHODS: TCF7, miR-16-5p, and SEMA3A were quantified by a qRT-PCR or immunoblotting method. A CCK-8 assay gauged the cell viability. Measurement of cell apoptosis was done using flow cytometry. RNA immunoprecipitation (RIP), dual-luciferase reporter, and RNA pull-down assays were utilized to assay the targeted interactions among the variables. RESULTS: The TCF7 and SEMA3A levels were elevated in serum from patients with diabetic nephropathy. TCF7 silencing or SEMA3A depletion ameliorated high glucose (HG)-induced podocyte injury. Moreover, TCF7 silencing protected against HG-induced podocyte injury by down-regulating SEMA3A. TCF7 targeted miR-16-5p, and miR-16-5p targeted SEMA3A. Furthermore, TCF7 affected the expression of SEMA3A by competing specifically for shared miR-16-5p. CONCLUSIONS: These findings suggested that TCF7 silencing attenuated high glucose-induced podocyte damage partially through the miR-16-5p/SEMA3A regulation cascade.


Assuntos
Nefropatias Diabéticas , MicroRNAs , Podócitos , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Nefropatias Diabéticas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Glucose/toxicidade , Glucose/metabolismo , Apoptose , Fator 1 de Transcrição de Linfócitos T/metabolismo
10.
Acta Biomater ; 157: 236-251, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36435442

RESUMO

Semaphorin 3A (sema3A) is an osteoprotective factor that enhances bone formation while inhibiting osteoclast bone resorption. It is produced by rat calvarial osteoblasts cultured on grit-blasted/acid-etched microtextured (SLA) titanium surfaces at higher levels than on tissue culture polystyrene, suggesting that it may improve performance of titanium implants in vivo, particularly in conditions characterized by compromised bone quality. To test this, we established a clinically relevant type 2 diabetes mellitus (T2DM) rat model and used a non-toxic click hydrogel that rapidly polymerizes in situ (GEL) to provide localized controlled delivery of sema3A. In vitro studies confirmed that sema3A released from GEL was biologically active, increasing osteoblast differentiation of a pre-osteoblast cell-line. Whereas increased sema3A production was not observed in T2DM calvarial osteoblasts cultured on SLA, exogenous sema3A enhanced surface-induced osteoblast differentiation, indicating that it would be a viable candidate for in vivo use. Delivery of sema3A either by GEL or by local injection to bone defects enhanced osseointegration of SLA implants in the T2DM rats. Trabecular bone mass and bone-to-implant contact were decreased in T2DM rats compared to normal rats; sema3A delivered locally improved both parameters. These findings suggest that reduced trabecular bone contributes to poor osseointegration in T2DM patients and support GEL as a promising treatment option for sustained release of therapeutic doses of sema3A. Moreover, using this clinically translatable T2DM model and developing a biocompatible, Cu-free click chemistry hydrogel platform for the non-invasive delivery of therapeutics has major implications for regenerative medicine as a whole. STATEMENT OF SIGNIFICANCE: Osseointegration is compromised in patients with poor bone quality due to conditions like type 2 diabetes mellitus (T2DM). Previously, we showed that semaphorin 3A (sema3A) production is increased when human bone marrow stromal cells are cultured on titanium substrates that support osseointegration in vivo, suggesting it may enhance peri-implant osteogenesis in diabetes. Here we established a spontaneously developing T2DM rat model with clinical translatability and used it to assess sema3A effectiveness. Sema3A was delivered to the implant site via a novel copper-free click hydrogel, which has minimal swelling behavior and superior rheological properties. Osseointegration was successfully restored, and enhanced compared to burst release through injections. This study provides scientific evidence for using sema3A to treat impaired osseointegration in T2DM patients.


Assuntos
Diabetes Mellitus Tipo 2 , Semaforina-3A , Ratos , Humanos , Animais , Semaforina-3A/farmacologia , Osseointegração , Titânio/farmacologia , Hidrogéis , Osteogênese , Osteoblastos , Propriedades de Superfície
11.
Transl Vis Sci Technol ; 11(6): 17, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35727188

RESUMO

Purpose: Semaphorin 3A (Sema3A) is an axonal guidance molecule that inhibits angiogenesis by vasorepulsion and blocks revascularization in the ischemic retina. BI-X is an intravitreal anti-Sema3A agent under clinical investigation in patients with proliferative diabetic retinopathy (PDR) and diabetic macular ischemia (DMI). Methods: Surface plasmon resonance was used to determine binding affinity of BI-X to human and murine Sema3A. In vitro, human retinal microvascular endothelial cells (HRMECs) were used to assess effects of BI-X on cell permeability and cytoskeletal collapse induced by Sema3A. In vivo, intravitreal BI-X or an anti-trinitrophenol control antibody was administered in both eyes in mice with oxygen-induced retinopathy (OIR). Retinal flat mounts were prepared, and avascular area and tip cell density were determined using confocal laser-scanning microscopy. Results: Dissociation constants for BI-X binding to human and murine Sema3A were 29 pM and 27 pM, respectively. In vitro, BI-X prevented HRMEC permeability and cytoskeletal collapse induced by Sema3A. In vivo, BI-X increased tip cell density by 33% (P < 0.001) and reduced avascular area by 12% (not significant). A significant negative correlation was evident between avascular area and tip cell density (r2 = 0.4205, P < 0.0001). Conclusions: BI-X binds to human Sema3A with picomolar affinity and prevents cell permeability and cytoskeletal collapse in HRMECs. BI-X also enhances revascularization in mice with OIR. Translational Relevance: BI-X is a potent inhibitor of human Sema3A that improves revascularization in a murine model of OIR; BI-X is currently being investigated in patients with laser-treated PDR and DMI.


Assuntos
Citoesqueleto , Retinopatia Diabética , Doenças Retinianas , Animais , Contagem de Células , Permeabilidade da Membrana Celular , Retinopatia Diabética/tratamento farmacológico , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Oxigênio/toxicidade , Permeabilidade , Retina , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia
12.
Cell Rep ; 38(11): 110483, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35294878

RESUMO

The development of the apical dendrite from the leading process of the bipolar pyramidal neuron might be directed by spatially organized extrinsic cues acting on localized intrinsic determinants. The extracellular cues regulating apical dendrite polarization remain elusive. We show that leading process and apical dendrite development are directed by class III Semaphorins and mediated by a localized cGMP-synthesizing complex. The scaffolding protein Scribble that associates with the cGMP-synthesizing enzyme soluble guanylate cyclase (sGC) also associates with the Semaphorin3A (Sema3A) co-receptor PlexinA3. Deletion or knockdown of PlexinA3 and Sema3A or disruption of PlexinA3-Scribble association prevents Sema3A-mediated cGMP increase and causes defects in apical dendrite development. These manipulations also impair bipolar polarity and leading process establishment. Local cGMP elevation or sGC expression rescues the effects of PlexinA3 knockdown or PlexinA3-Scribble complex disruption. During neuronal polarization, leading process and apical dendrite development are directed by a scaffold that links Semaphorin cue to cGMP increase.


Assuntos
Semaforina-3A , Semaforinas , Células Cultivadas , GMP Cíclico/metabolismo , Dendritos/metabolismo , Neurogênese , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Semaforinas/metabolismo
13.
Cell Death Differ ; 29(10): 1941-1954, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35347234

RESUMO

Semaphorin 3A (Sema3A) has been recognized as a crucial regulator of morphogenesis and homeostasis over a wide range of organ systems. However, its function in cutaneous wound healing is poorly understood. In our study, we demonstrated that Sema3A adenovirus plasmids transfection limited keratinocyte proliferation and decreased migrative capacity as assessed by in vitro wound healing assay. Sema3A transduction inhibited TGF-ß1-mediated keratinocyte migration and EMT process. Besides, we applied mice with K14-Cre-mediated deletion of Sema3A and found that Sema3A depletion postponed wound closure with decreased re-epithelialization and matrix growth. Contrary to the results obtained with full-length Sema3A plasmids transfection, increased keratinocyte migration with recombinant Sema3A proteins resulted in quicker closure of the wounding area after a scratch. Further, exogenously applied recombinant Sema3A worked with EGF to maintain the activation of EGFR by interacting with NRP1 and thereby regulated the internalization of the EGFR-NRP1 complex. Taken together, these results indicated a paradoxical role of autonomous and non-autonomous Sema3A expression during wound healing. Combined administration of recombinant EGF and Sema3A proteins could accelerate the process of wound repair, thus providing promising treatment prospects in the future.


Assuntos
Semaforina-3A , Fator de Crescimento Transformador beta1 , Animais , Fator de Crescimento Epidérmico , Receptores ErbB , Camundongos , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Cicatrização
14.
Eur J Orthod ; 44(4): 420-426, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35290462

RESUMO

BACKGROUND AND OBJECTIVES: Cementoblasts can communicate with osteoclasts by synthesis and secretion of cytokines, such as RANKL, OPG, and M-CSF. Previously, we reported that irisin promotes the differentiation of cementoblasts, while the effect of irisin on cementoblast-mediated osteoclastogenesis remains inconclusive. This study aimed to explore the effect of irisin on the expression of osteoclastogenesis-related cytokines in cementoblasts. MATERIAL AND METHODS: An immortalized murine cementoblast cell line OCCM-30 was used. Immunofluorescence and Western Blot were performed to identify the expression of irisin receptor integrin alphaV and the activation of its downstream signals in OCCM-30 cells. Cells were treated with irisin (100 ng/ml) for various time lengths ranging from 0 to 72 hours, and then qRT-PCR was used to detect the expression of osteoclastogenesis-related genes, including RANKL, IL-6, M-CSF, OPG, Wnt5A, Sema3A. Cells were also incubated with irisin in a series of concentrations (0-200 ng/ml) for 24 hours, and then qRT-PCR and ELISA were performed to examine the above osteoclastogenesis-related cytokines. RESULTS: Irisin receptor integrin alphaV was expressed in OCCM-30 cells and its downstream signaling pathways were markedly activated by irisin. Both qRT-PCR and ELISA results revealed that RANKL and IL-6 were up-regulated by irisin while M-CSF, OPG, Wnt5A, Sema3A remained unaffected. CONCLUSIONS: OCCM-30 cells were responsive to the stimulation of irisin. The expression of RANKL and IL-6 was significantly enhanced by irisin, suggesting a possible promotive effect on cementoblast-mediated osteoclastogenesis.


Assuntos
Cemento Dentário , Osteoclastos , Animais , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Diferenciação Celular , Fibronectinas/metabolismo , Fibronectinas/farmacologia , Integrina alfaV/metabolismo , Integrina alfaV/farmacologia , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia
15.
J Biomater Appl ; 36(10): 1775-1785, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35225049

RESUMO

Recently, semaphorin 3A (Sema3A) has been identified as a critical gene for osteogenic differentiation of mesenchymal stem cells and increases osteoblastic bone formation. However, in current research studies, there is a lack of focus on whether Sema3a can affect the osseointegration of titanium rods in diabetes and through what biological mechanisms. Therefore, the present work was aimed to evaluate the effect of local administration with Sema3A on hydroxyapatite coated titanium rod osseointegration in diabetic rat model and preliminary exploration of possible mechanisms. The MC3T3-E1 cells were co-cultured with Sema3A and high glucose and induced to osteogenesis, and the cell viability, osteogenic activity was observed by Cell Counting Kit-8(CCK-8), Alkaline Phosphatase staining, Alizarin Red Staining, and Western Blot. In vitro experiments, CCK-8, ALP, and ARS staining results show that the mineralization and osteogenic activity of MC3T3-E1increased significantly after intervention by Sema3A, as well as a higher levels of protein expressions including Runt-Related Transcription Factor 2, silent mating type information regulation 2 homolog-1(SIRT1), catalase (CAT), superoxide dismutase 1 (SOD1), and superoxide dismutase 2 (SOD2). In vivo experiment, a better stability and osseointegration of the titanium rod were observed after treatment with Sema3A, as well as a higher SOD1, SOD2, CAT, and SIRT1 gene expression. The present study indicates that local treatment with Sema3A was associated with increased osseointegration of titanium rod by reducing the oxidative stress of osteoblasts and enhancing the function of osteoblasts in a diabetic rat.


Assuntos
Diabetes Mellitus Experimental , Osseointegração , Animais , Diferenciação Celular , Diabetes Mellitus Experimental/terapia , Durapatita/farmacologia , Osteoblastos , Osteogênese , Ratos , Semaforina-3A/farmacologia , Semaforina-3A/uso terapêutico , Titânio/farmacologia
16.
Clin Exp Nephrol ; 26(5): 385-397, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34997887

RESUMO

BACKGROUND: Diabetic nephropathy (DN), a diabetic complication, is the leading cause of end-stage renal disease. KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1), a long non-coding RNA, has been unmasked to participate in the pathogenesis of DN. However, the specific mechanism by which KCNQ1OT1 regulates podocyte injury remains unclear. METHODS: Relative expression of KCNQ1OT1 was measured with quantitative real-time polymerase chain reaction (qRT-PCR). The levels of inflammatory cytokines were analyzed by enzyme linked immunosorbent assay (ELISA). The viability, proliferation, and apoptosis of high glucose (HG)-treated podocyte were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. Protein levels were analyzed by western blotting. The regulatory mechanism of KCNQ1OT1 was surveyed by bioinformatics analysis, dual-luciferase reporter, and RNA immunoprecipitation (RIP) assays. RESULTS: We observed an apparent upregulation in KCNQ1OT1 expression in serums of DN patients and HG-treated podocytes. Furthermore, KCNQ1OT1 downregulation alleviated HG-induced inflammation, proliferation repression, and apoptosis in podocytes. Notably, KCNQ1OT1 was identified as a miR-23b-3p sponge, and miR-23b-3p directly targeted Semaphorin-3A (Sema3A). Moreover, miR-23b-3p silencing reversed KCNQ1OT1 knockdown-mediated effects on inflammation, proliferation, and apoptosis of HG-induced podocytes. Also, Sema3A overexpression reversed the effects of miR-23b-3p mimic on inflammation, proliferation, and apoptosis of HG-induced podocytes. Importantly, KCNQ1OT1 regulated Sema3A expression by sponging miR-23b-3p. CONCLUSIONS: HG-induced KCNQ1OT1 promoted inflammation, proliferation repression, and apoptosis of podocytes via increasing Sema3A expression through sponging miR-23b-3p. This study provided evidence to support the involvement of KCNQ1OT1 in the pathogenesis of DN.


Assuntos
Nefropatias Diabéticas , MicroRNAs , Podócitos , Apoptose , Nefropatias Diabéticas/patologia , Feminino , Glucose/metabolismo , Glucose/toxicidade , Humanos , Inflamação , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Podócitos/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia
17.
J Periodontal Res ; 57(2): 425-433, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35037251

RESUMO

OBJECTIVE AND BACKGROUND: The occurrence and development of periodontitis are closely related to hypoxia of the periodontal microenvironment. Periodontal ligament stem cells (PDLSCs) are considered to have potential to regenerate periodontal tissues. Semaphorin 3A (Sema3A) plays an essential role in promoting osteogenesis. However, the effect of Sema3A on osteogenesis of PDLSCs under hypoxia remains unclear. The aim of this study was to investigate the effect of Sema3A on osteogenesis of PDLSCs under hypoxia. METHODS: Isolated PDLSCs were identified using flow cytometry. Adipogenic differentiation potential was identified by oil red O staining. Osteogenesis was measured using Alizarin Red S staining and ALP staining. Intracellular hypoxia was induced using cobalt chloride (CoCl2 ). The expression level of hypoxia-inducible factor-1α (HIF-1α) was detected via ELISA. Expression of osteogenic markers and Sema3A was analyzed using western blot and real-time PCR. RESULTS: The proliferation and osteogenesis of PDLSCs were markedly inhibited with increased concentrations of CoCl2 . Under the treatment with a low concentration of CoCl2 , expression of related osteogenic markers and Sema3A decreased in a time-dependent manner. ARS and ALP staining results also showed that osteogenic calcification decreased under hypoxia. Apigenin, an inhibitor of HIF-1α, effectively up-regulated expression of Sema3A and osteogenic markers with CoCl2 treatment. Moreover, exogenous Sema3A significantly increased the expression of osteogenesis-related markers and mineralization of PDLSCs according to ALP and ARS staining with CoCl2 treatment. CONCLUSIONS: Hypoxia markedly inhibited osteogenesis of PDLSCs. Sema3A explicitly attenuated the hypoxia suppression of osteogenesis in PDLSCs.


Assuntos
Osteogênese , Ligamento Periodontal , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Hipóxia/metabolismo , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Células-Tronco/metabolismo
18.
Immunol Res ; 70(2): 135-142, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35031951

RESUMO

Semaphorin-3A (Sema-3A), a secreted member of the semaphorin family, is well known for playing regulatory functions at all stages of the immune response. Sema-3A transduces signals by binding to its cognate receptors, namely, class A plexins (Plxns A1 to A4) and neuropilin-1 (Nrp-1). The downstream diverse signaling pathways induced by connecting Sema-3A to its receptors were found to be involved in the pathogenesis of different immunological disorders, ranging from cancer to autoimmunity and allergies. Recent studies have demonstrated that Sema-3A expression is diminished in the murine models and patients with allergic rhinitis (AR; a chronic inflammatory disorder of the nasal mucosa), suggesting the involvement of Sema-3A in AR pathogenesis. Investigations also revealed that treatment of these mice with exogenous Sema-3A protein alleviates the clinical symptom scores of AR, thereby compensating for the reduced expression of Sema-3A in AR. Indeed, Sema-3A treatment could suppress allergic responses in AR via inhibiting Th2/Th17 responses and boosting Th1/Treg responses. Also, Sema-3A could diminish dendritic cell (DC) maturation and T cell proliferation. Since it is implicated in the pathogenesis of AR; thus, Sema-3A turns to be a promising tool of therapy to be studied and utilized in this disease. This review intends to highlight the recent evidence on the role of Sema-3A in AR pathogenesis and summarizes the recent findings regarding the expression status of Sema-3A, as well as its therapeutic potential for treating this disease. HIGHLIGHTS: Sema-3A plays regulatory functions at all stages of the immune response. Sema-3A receptors are the class A plexins (A1-A4) and neuropilin-1 (Nrp-1). Sema-3A expression is reduced in murine models and patients with allergic rhinitis. Connecting Sema-3A to Nrp-1 increases Foxp3 expression in Treg cells. Injecting Sema-3A protein exerts therapeutic effects in mouse models of allergic diseases. Sema-3A shows promise as a therapeutic tool for the treatment of allergic rhinitis.


Assuntos
Rinite Alérgica , Semaforina-3A , Animais , Humanos , Camundongos , Neuropilina-1 , Rinite Alérgica/tratamento farmacológico , Semaforina-3A/metabolismo , Semaforina-3A/farmacologia , Transdução de Sinais , Linfócitos T Reguladores
19.
Connect Tissue Res ; 63(4): 382-392, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34486459

RESUMO

Distraction osteogenesis (DO) is a bone regeneration technique used to treat maxillofacial disorders, fracture nonunion, and large bone defects. It is well known for its amazing regenerative potential, but an extended consolidation period limits its clinical use. The interaction between the nervous system and bone regeneration has attracted great attention in recent years. Sema3A is a key axonal chemorepellent which has been proved to have bone-protective effects. In this article, we try to improve DO by local administration of Sema3A and explore the possible mechanisms. Forty wildtype, male, adult mice were divided into two groups after tibia osteotomy surgery. Sema3A or Saline was daily injected transcutaneous into the center of the distraction zone during the consolidation period. Micro-CT images were taken at 4, 6,8 and 10 weeks post-surgery; vascular density and biomechanical testing were performed at 10 weeks post-surgery. We also set up in vitro vessel growth assay to evaluate the effect of Sema3A on angiogenesis. Compared with the Saline group, Sema3A treatment significantly accelerated bone regeneration, improved angiogenesis and callus' biomechanical strength. At 10 weeks post-surgery, compared with the Saline group, the BV/TV, BMD, TMD increased by about 23%, 22%, 18% respectively, vascular density increased by about 49% in the Sema3A group. Histological images and western-blot showed decreased expression of VEGF-A and increased expression of Ang-1 at 4 weeks post-surgery in the Sema3A group. In vitro, Sema3A suppressed VEGF-induced angiogenesis but had little effect on Ang-induced angiogenesis. Conclusion: Sema3A could accelerate bone regeneration and improve angiogenesis during DO.


Assuntos
Regeneração Óssea , Osteogênese por Distração , Semaforina-3A , Animais , Masculino , Camundongos , Osteogênese , Osteogênese por Distração/métodos , Semaforina-3A/farmacologia
20.
PLoS One ; 15(11): e0234950, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33232327

RESUMO

Following injury to the peripheral and central nervous systems, tissue levels of transforming growth factor (TGF)-ß1 often increase, which is key for wound healing and scarring. However, active wound regions and scars appear to inhibit process outgrowth by regenerating neurons. We recently showed that corneal wound myofibroblasts block corneal nerve regeneration in vivo, and sensory neurite outgrowth in vitro in a manner that relies critically on TGF-ß1. In turn, delayed, abnormal re-innervation contributes to long-term sensory dysfunctions of the ocular surface. Here, we exposed morphologically and biochemically-differentiated sensory neurons from the ND7/23 cell line to TGF-ß1 to identify the intracellular signals regulating these anti-neuritogenic effects, contrasting them with those of Semaphorin(Sema)3A, a known inhibitor of neurite outgrowth. Neuronal morphology was quantified using phase-contrast imaging. Western blotting and specific inhibitors were then used to identify key molecular mediators. Differentiated ND7/23 cells expressed neuron-specific markers, including those involved in neurite extension and polarization. TGF-ß1 increased phosphorylation of collapsin response mediator protein-2 (CRMP2), a molecule that is key for neurite extension. We now show that both glycogen synthase kinase (GSK)-3ß and Smad3 modulate phosphorylation of CRMP2 after treatment with TGF-ß1. GSK-3ß appeared to exert a particularly strong effect, which could be explained by its ability to phosphorylate not only CRMP2, but also Smad3. In conclusion, TGF-ß1's inhibition of neurite outgrowth in sensory neurons appears to be regulated through a highly-conserved signaling pathway, which involves the GSK-3ß/CRMP-2 loop via both canonical and non-canonical mechanisms. It is hoped that by defining the signaling pathways that control neurite outgrowth in wound environments, it will become possible to identify optimal molecular targets to promote re-innervation following injury.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/efeitos adversos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Camundongos , Microscopia de Contraste de Fase , Fator de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Semaforina-3A/farmacologia , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...