Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Glia ; 71(7): 1715-1728, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36971019

RESUMO

Our previous studies indicated that RhoA knockdown or inhibition could alleviate the proliferation, migration, and differentiation of Schwann cells. However, the role of RhoA in Schwann cells during nerve injury and repair is still unknown. Herein, we developed two lines of Schwann cells conditional RhoA knockout (cKO) mice by breeding RhoAflox / flox mice with PlpCre -ERT2 or DhhCre mice. Our results indicate that RhoA cKO in Schwann cells accelerates axonal regrowth and remyelination after sciatic nerve injury, which enhances the recovery of nerve conduction and hindlimb gait, and alleviates the amyotrophy in gastrocnemius muscle. Mechanistic studies in both in vivo and in vitro models revealed that RhoA cKO could facilitate Schwann cell dedifferentiation via JNK pathway. Schwann cell dedifferentiation subsequently promotes Wallerian degeneration by enhancing phagocytosis and myelinophagy, as well as stimulating the production of neurotrophins (NT-3, NGF, BDNF, and GDNF). These findings shed light on the role of RhoA in Schwann cells during nerve injury and repair, indicating that cell type-specific RhoA targeting could serve as a promising molecular therapeutic strategy for peripheral nerve injury.


Assuntos
Traumatismos dos Nervos Periféricos , Neuropatia Ciática , Camundongos , Animais , Desdiferenciação Celular , Nervo Isquiático/metabolismo , Células de Schwann/metabolismo , Neuropatia Ciática/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/metabolismo
2.
Phytother Res ; 37(10): 4607-4620, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37380363

RESUMO

Atractylenolide-III (AT-III) is well known as its role in antioxidant and anti-inflammatory. Present study was aimed to figure out its effects on osteoarthritis and potential mechanisms. Rat model, human osteoarthritis cartilage explants as well as rat/human chondrocyte cultures were prepared to test AT-III's effects on osteoarthritis progression and chondrocyte senescence. Potential targeted molecules of AT-III were predicted using network pharmacology and molecular docking, assessed by Western blotting and then verified with rescue experiments. AT-III treatment alleviated osteoarthritis severity (shown by OARSI grading score and micro-CT) and chondrocyte senescence (indexed by levels of SA-ß-gal, P16, P53, MMP13, ROS and ratio of healthy/collapsed mitochondrial membrane potentials). Network pharmacology and molecular docking suggested that AT-III might play role through NF-κB pathway. Further experiments revealed that AT-III reduced phosphorylation of IKKα/ß, IκBα and P65 in NF-κB pathway. As well as nuclear translocation of p65. Both in vivo and in vitro experiments indicated that AT-III's effects on osteoarthritis and anti-senescence were reversed by an NF-κB agonist. AT-III could alleviate osteoarthritis by inhibiting chondrocyte senescence through NF-κB pathway, which indicated that AT-III is a prospective drug for osteoarthritis treatment.

3.
J Neuroinflammation ; 18(1): 234, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34654444

RESUMO

BACKGROUND: Plenty of macrophages are recruited to the injured nerve to play key roles in the immunoreaction and engulf the debris of degenerated axons and myelin during Wallerian degeneration, thus creating a conducive microenvironment for nerve regeneration. Recently, drugs targeting the RhoA pathway have been widely used to promote peripheral axonal regeneration. However, the role of RhoA in macrophage during Wallerian degeneration and nerve regeneration after peripheral nerve injury is still unknown. Herein, we come up with the hypothesis that RhoA might influence Wallerian degeneration and nerve regeneration by affecting the migration and phagocytosis of macrophages after peripheral nerve injury. METHODS: Immunohistochemistry, Western blotting, H&E staining, and electrophysiology were performed to access the Wallerian degeneration and axonal regeneration after sciatic nerve transection and crush injury in the LyzCre+/-; RhoAflox/flox (cKO) mice or Lyz2Cre+/- (Cre) mice, regardless of sex. Macrophages' migration and phagocytosis were detected in the injured nerves and the cultured macrophages. Moreover, the expression and potential roles of ROCK and MLCK were also evaluated in the cultured macrophages. RESULTS: 1. RhoA was specifically knocked out in macrophages of the cKO mice; 2. The segmentation of axons and myelin, the axonal regeneration, and nerve conduction in the injured nerve were significantly impeded while the myoatrophy was more severe in the cKO mice compared with those in Cre mice; 3. RhoA knockout attenuated the migration and phagocytosis of macrophages in vivo and in vitro; 4. ROCK and MLCK were downregulated in the cKO macrophages while inhibition of ROCK and MLCK could weaken the migration and phagocytosis of macrophages. CONCLUSIONS: Our findings suggest that RhoA depletion in macrophages exerts a detrimental effect on Wallerian degeneration and nerve regeneration, which is most likely due to the impaired migration and phagocytosis of macrophages resulted from disrupted RhoA/ROCK/MLCK pathway. Since previous research has proved RhoA inhibition in neurons was favoring for axonal regeneration, the present study reminds us of that the cellular specificity of RhoA-targeted drugs is needed to be considered in the future application for treating peripheral nerve injury.


Assuntos
Macrófagos/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Degeneração Walleriana/metabolismo , Degeneração Walleriana/prevenção & controle , Proteína rhoA de Ligação ao GTP/deficiência , Animais , Movimento Celular/fisiologia , Células Cultivadas , Feminino , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismos dos Nervos Periféricos/patologia , Degeneração Walleriana/patologia , Proteína rhoA de Ligação ao GTP/genética
4.
Connect Tissue Res ; 60(3): 240-253, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29916278

RESUMO

In physiological and pathological environments, the concentration of oxygen around osteoblasts varies widely. No studies have systematically evaluated the effects of different oxygen concentrations on the proliferation, survival, migration, and osteogenic differentiation of osteoblasts. In this study, we cultured the osteoblast precursor cell line MC3T3-E1 in small individual chambers with oxygen concentrations of 1%, 3%, 6%, 9%, and 21%. Cell proliferation was evaluated by the proliferation index test and EdU staining. To test cell survival, a live/dead assay was performed. A tablet scratch assay was performed to detect the migratory ability of the cells. Bone nodule formation experiments and immunofluorescence and Western blotting analyses of osteogenic-related proteins were performed to assess the osteogenic differentiation of the cells. We found that the proliferation and osteogenic differentiation ability of MC3T3-E1 cells in different oxygen concentrations were both approximately bell-shaped curves and that the optimal oxygen concentrations were approximately 6% and 9%, respectively. The live/dead assay showed that the survival of MC3T3-E1 cells in different oxygen concentrations was affected by the amount of serum. The tablet scratch experiment showed that there was greater cell migration with oxygen concentrations of 1%, 3%, and 21% than with oxygen concentrations of 6% and 9%. Our results have significant reference value for the intervention of the pathological processes involving osteoblasts, such as fracture, osteoporosis, and some vascular diseases. These results also have an important guiding role for the new scientific idea that osteoblasts can function as treatment cells to repair bone defects.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Oxigênio/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Camundongos , Osteocalcina/metabolismo
5.
Connect Tissue Res ; 60(6): 583-596, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31035811

RESUMO

Purpose: Mesenchymal stem cells (MSCs) seeded on biocompatible scaffolds have therapeutic potential for bone defect repair. However, MSCs can be affected by hypoxia and nutritional deficiency due to a lack of blood vessels in the scaffolds. Here, we explored the effects of hypoxia on MSC differentiation to clarify these mechanisms. Methods: Peripheral blood mesenchymal stem cells (PBMSCs) were cultured in small individual chambers with oxygen concentrations of 1%, 9%, and 21%. Cell proliferation was evaluated by Cell Counting Kit 8 assays, and cell survival was determined using live/dead assays. Scratch assays were performed to evaluate cell migration. Ca2+ deposition/mineralization experiments, reverse transcription quantitative real-time polymerase chain reaction, and Western blotting were performed to assess the osteogenic differentiation of cells. Notch1 expression was downregulated by lentivirus-transfected PBMSCs to observe the effects of Notch1 knockdown on osteogenic gene and protein expression. Results: PBMSCs exposed to hypoxia (1% O2) demonstrated accelerated proliferation, increased migration, and reduced survival in the absence of serum. Although 9% oxygen promoted osteogenic differentiation, the osteogenic differentiation of PBMSCs was significantly reduced by 1% O2, and this effect was associated with increased Notch1 expression. Reducing Notch1 expression using small interfering RNA significantly restored the osteogenic differentiation of PBMSCs. Conclusions: Hypoxia accelerated proliferation, increased migration, and reduced PBMSC differentiation into osteoblasts by increasing Notch1 expression. These findings may contribute to the development of appropriate cell culture or in vivo transplantation conditions to maintain the full osteogenic potential of PBMSCs.


Assuntos
Células Sanguíneas/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Receptor Notch1/biossíntese , Regulação para Cima , Animais , Células Sanguíneas/citologia , Hipóxia Celular , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Sprague-Dawley
6.
Neural Plast ; 2018: 8561704, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29849572

RESUMO

Accumulating evidences suggest that peripheral nerve injury (PNI) may initiate astrocytic responses in the central nervous system (CNS). However, the response of astrocytes in the spinal ventral horn and its potential role in nerve regeneration after PNI remain unclear. Herein, we firstly illustrated that astrocytes in the spinal ventral horn were dramatically activated in the early stage following sciatic nerve injury, and these profiles were eliminated in the chronic stage. Additionally, we found that the expression of neurotrophins, including brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and neurotrophin-3 (NT-3), also accompanied with astrocyte activation. In comparison with the irreversible transected subjects, astrocyte activation and the neurotrophic upregulation in the early stage were more drastic in case the transected nerve was rebridged immediately after injury. Furthermore, administering fluorocitrate to inhibit astrocyte activation resulted in decreased neurotrophin expression in the spinal ventral horn and delayed axonal regeneration in the nerve as well as motor function recovery. Overall, the present study indicates that peripheral nerve injury can initiate astrocyte activation accompanied with neurotrophin upregulation in the spinal ventral horn. The above responses mainly occur in the early stage of PNI and may contribute to nerve regeneration and motor function recovery.


Assuntos
Astrócitos/metabolismo , Regeneração Nervosa , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Corno Ventral da Medula Espinal/metabolismo , Animais , Feminino , Fatores de Crescimento Neural/metabolismo , Traumatismos dos Nervos Periféricos/complicações , Ratos Sprague-Dawley , Nervo Isquiático/lesões , Traumatismos da Medula Espinal/complicações , Corno Ventral da Medula Espinal/fisiopatologia
7.
Molecules ; 23(2)2018 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-29473886

RESUMO

The study tried to explore the role of sugar-residues and mechanisms of phenolic phenylpropanoid antioxidants. Acteoside, along with its apioside forsythoside B and rhamnoside poliumoside, were comparatively investigated using various antioxidant assays. In three electron-transfer (ET)-based assays (FRAP, CUPRAC, PTIO•-scavenging at pH 4.5), the relative antioxidant levels roughly ruled as: acteoside >forsythoside B > poliumoside. Such order was also observed in H⁺-transfer-involved PTIO•-scavenging assay at pH 7.4, and in three multiple-pathway-involved radical-scavenging assays, i.e., ABTS⁺•-scavenging, DPPH•-scavenging, and •O2--scavenging. In UV-vis spectra, each of them displayed a red-shift at 335→364 nm and two weak peaks (480 and 719 nm), when mixed with Fe2+; however, acteoside gave the weakest absorption. In Ultra-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight tandem mass spectrometry (UPLC-ESI-Q-TOF-MS/MS) analysis, no radical-adduct-formation (RAF) peak was found. MTT assay revealed that poliumoside exhibited the highest viability of oxidative-stressed bone marrow-derived mesenchymal stem cells. In conclusion, acteoside, forsythoside B, and poliumoside may be involved in multiple-pathways to exert the antioxidant action, including ET, H⁺-transfer, or Fe2+-chelating, but not RAF. The ET and H⁺-transfer may be hindered by rhamnosyl and apiosyl moieties; however, the Fe2+-chelating potential can be enhanced by two sugar-residues (especially rhamnosyl moiety). The general effect of rhamnosyl and apiosyl moieties is to improve the antioxidant or cytoprotective effects.


Assuntos
Antioxidantes/química , Antioxidantes/farmacologia , Citoproteção , Glucosídeos/química , Glucosídeos/farmacologia , Fenóis/química , Fenóis/farmacologia , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Fenômenos Mecânicos , Metais/química , Estrutura Molecular , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta , Espectrometria de Massas em Tandem
8.
Ann Neurol ; 75(2): 255-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24339129

RESUMO

OBJECTIVE: The peripheral myelin protein-22 (PMP22) gene is associated with the most common types of inherited neuropathies, including hereditary neuropathy with liability to pressure palsies (HNPP) caused by PMP22 deficiency. However, the function of PMP22 has yet to be defined. Our previous study has shown that PMP22 deficiency causes an impaired propagation of nerve action potentials in the absence of demyelination. In the present study, we tested an alternative mechanism relating to myelin permeability. METHODS: Utilizing Pmp22(+) (/) (-) mice as a model of HNPP, we evaluated myelin junctions and their permeability using morphological, electrophysiological, and biochemical approaches. RESULTS: We show disruption of multiple types of cell junction complexes in peripheral nerve, resulting in increased permeability of myelin and impaired action potential propagation. We further demonstrate that PMP22 interacts with immunoglobulin domain-containing proteins known to regulate tight/adherens junctions and/or transmembrane adhesions, including junctional adhesion molecule-C (JAM-C) and myelin-associated glycoprotein (MAG). Deletion of Jam-c or Mag in mice recapitulates pathology in HNPP. INTERPRETATION: Our study reveals a novel mechanism by which PMP22 deficiency affects nerve conduction not through removal of myelin, but through disruption of myelin junctions.


Assuntos
Artrogripose/genética , Artrogripose/metabolismo , Neuropatia Hereditária Motora e Sensorial/genética , Neuropatia Hereditária Motora e Sensorial/metabolismo , Proteínas da Mielina/deficiência , Bainha de Mielina/metabolismo , Junções Íntimas/patologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Moléculas de Adesão Juncional/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/genética , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Potássio/farmacologia , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/genética , Junções Íntimas/metabolismo
9.
J Anat ; 224(5): 575-82, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24506149

RESUMO

c-Jun activation has been implicated not only in neuronal degeneration, but also in survival and regeneration. Here, we investigated c-Jun activation in injured motoneurons by using a nerve crush model in neonatal rats. We identified two distinct subpopulations of motoneurons: about 60% underwent degeneration following injury whereas the remaining 40% survived and induced a regeneration response at 3 weeks post injury. However, all motoneurons examined expressed phosphorylated-c-Jun-immunoreactivity (p-c-Jun-IR) at the early stage of 3 days following injury. These results suggest that active c-Jun was induced in all neonatal motoneurons following nerve crush injury, regardless of whether they were destined to degenerate or undergo successful regeneration at a later stage. Our findings therefore support the hypothesis that active c-Jun is involved in both neuronal degeneration and regeneration.


Assuntos
Axônios/metabolismo , Plexo Braquial/lesões , Plexo Braquial/fisiologia , Morte Celular/fisiologia , Neurônios Motores/fisiologia , Regeneração Nervosa/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Medula Espinal/metabolismo , Animais , Plexo Braquial/citologia , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Fosforilação , Ratos , Ratos Sprague-Dawley
10.
Nat Med ; 13(10): 1228-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17906634

RESUMO

Demyelinating diseases, such as multiple sclerosis, are characterized by the loss of the myelin sheath around neurons, owing to inflammation and gliosis in the central nervous system (CNS). Current treatments therefore target anti-inflammatory mechanisms to impede or slow disease progression. The identification of a means to enhance axon myelination would present new therapeutic approaches to inhibit and possibly reverse disease progression. Previously, LRR and Ig domain-containing, Nogo receptor-interacting protein (LINGO-1) has been identified as an in vitro and in vivo negative regulator of oligodendrocyte differentiation and myelination. Here we show that loss of LINGO-1 function by Lingo1 gene knockout or by treatment with an antibody antagonist of LINGO-1 function leads to functional recovery from experimental autoimmune encephalomyelitis. This is reflected biologically by improved axonal integrity, as confirmed by magnetic resonance diffusion tensor imaging, and by newly formed myelin sheaths, as determined by electron microscopy. Antagonism of LINGO-1 or its pathway is therefore a promising approach for the treatment of demyelinating diseases of the CNS.


Assuntos
Axônios/fisiologia , Encefalomielite Autoimune Experimental/induzido quimicamente , Proteínas de Membrana/antagonistas & inibidores , Bainha de Mielina/fisiologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Traumatismos da Medula Espinal/terapia , Animais , Axônios/diagnóstico por imagem , Axônios/ultraestrutura , Encefalomielite Autoimune Experimental/patologia , Injeções Espinhais , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Proteínas da Mielina , Bainha de Mielina/ultraestrutura , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Associada a Mielina/farmacologia , Glicoproteína Mielina-Oligodendrócito , Proteínas do Tecido Nervoso/administração & dosagem , Proteínas do Tecido Nervoso/fisiologia , Ratos , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Tomografia Computadorizada por Raios X
11.
J Peripher Nerv Syst ; 18(2): 162-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23781963

RESUMO

Skin biopsies have primarily been used to study the non-myelinated nerve fibers of the epidermis in a variety of neuropathies. In this study, we have expanded the skin biopsy technique to glabrous, non-hairy skin to evaluate myelinated nerve fibers in the most highly prevalent peripheral nerve disease, diabetic polyneuropathy (DPN). Twenty patients with DPN (Type I, n = 9; Type II, n = 11) and 16 age-matched healthy controls (age 29-73) underwent skin biopsy of the index finger, nerve conduction studies (NCS), and composite neuropathy scoring. In patients with DPN, we found a statistically significant reduction of both mechanoreceptive Meissner corpuscles (MCs) and their afferent myelinated nerve fibers (p = 0.01). This myelinated nerve fiber loss was correlated with the decreased amplitudes of sensory/motor responses in NCS. This study supports the utilization of skin biopsy to quantitatively evaluate axonal loss of myelinated nerve fibers in patients with DPN.


Assuntos
Neuropatias Diabéticas/patologia , Fibras Nervosas Mielinizadas/patologia , Adulto , Idoso , Biópsia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Feminino , Dedos/inervação , Dedos/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Condução Nervosa , Pele/inervação , Pele/patologia
12.
Nanomedicine ; 9(3): 305-15, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22960189

RESUMO

Peripheral nerve injury still remains a refractory challenge for both clinical and basic researchers. A novel nanofiber conduit made of blood vessel and filled with amphiphilic hydrogel of self-assembling nanofiber scaffold (SAPNS) was implanted to repair a 10 mm nerve gap after sciatic nerve transection. Empty blood vessel conduit was implanted serving as control. Results showed that this novel nanofiber conduit enabled the peripheral axons to regenerate across and beyond the 10 mm gap. Motoneuron protection, axonal regeneration and remyelination were significantly enhanced with SAPNS scaffold treatments. The target reinnervation and functional recovery induced by the regenerative nerve conduit suggest that SAPNS-based conduit is highly promising application in the treatment of peripheral nerve defect. FROM THE CLINICAL EDITOR: In this paper by Zhan et al, a novel self-assembling nanofiber scaffold is reported to promote regeneration of peripheral nerves in a sciatic nerve injury model. The promising results and the obvious medical need raises hope for a clinical translation of this approach hopefully in the near future.


Assuntos
Nanofibras/química , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/fisiopatologia , Alicerces Teciduais/química , Animais , Axônios/metabolismo , Movimento Celular , Feminino , Implantes Experimentais , Inflamação/complicações , Inflamação/patologia , Inflamação/fisiopatologia , Atividade Motora , Neurônios Motores/patologia , Músculos/inervação , Músculos/patologia , Músculos/fisiopatologia , Músculos/ultraestrutura , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , Nanofibras/ultraestrutura , Fibras Nervosas/patologia , Fibras Nervosas/ultraestrutura , Tamanho do Órgão , Traumatismos dos Nervos Periféricos/complicações , Traumatismos dos Nervos Periféricos/patologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Células de Schwann/metabolismo , Células de Schwann/patologia , Coloração e Rotulagem
13.
Mol Neurobiol ; 60(1): 51-67, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36216996

RESUMO

Spinal cord injury (SCI) can lead to the destruction of the blood-spinal cord barrier (BSCB), causing various inflammatory cytokines, neutrophils, and macrophages to infiltrate the lesion area, resulting in secondary injury. Basement membranes (BMs) are maintained by all types of cells in the BSCB and contribute to BSCB maintenance. Perlecan is an important constituent of vascular BMs, maintaining vascular integrity and neuroprotection. However, it is not clear whether Perlecan is involved in BSCB repair after SCI. In this study, we found that Perlecan was specifically expressed in the BMs in the spinal cord and underwent degradation/remodeling after SCI. Subsequently, a CRISPR/Cas9-based SAM system was used to overexpress Perlecan in the injured spinal cord, resulting in significantly enhanced locomotor recovery and neural regeneration. Overexpression of Perlecan reduced BSCB permeability along with the neuroinflammatory response. Interestingly, Perlecan inhibited stress fiber formation by interacting with integrin ß1 and inhibiting downstream ROCK/MLC signaling, resulting in reduced tight junctions (TJs) disassembly and improved BSCB integrity. Furthermore, the integrin receptor antagonist GRGDSP abolished the effects of Perlecan overexpression on BSCB permeability and TJs integrity. Overall, our findings suggest that Perlecan reduces BSCB permeability and the neuroinflammatory response by interacting with integrin ß1 and inhibiting the downstream ROCK/MLC pathway to promote neurological recovery after SCI.


Assuntos
Integrina beta1 , Traumatismos da Medula Espinal , Animais , Barreira Hematoencefálica/patologia , Proteínas da Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Medula Espinal/patologia , Traumatismos da Medula Espinal/patologia , Camundongos
14.
ACS Biomater Sci Eng ; 9(12): 6821-6834, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38011305

RESUMO

In recent years, conductive biomaterials have been widely used to enhance peripheral nerve regeneration. However, most biomaterials use electronic conductors to increase the conductivity of materials. As information carriers, electronic conductors always transmit discontinuous electrical signals, while biological systems essentially transmit continuous signals through ions. Herein, an ion-based conductive hydrogel was fabricated by simple copolymerization of the zwitterionic monomer sulfobetin methacrylate and hydroxyethyl methacrylate. Benefiting from the excellent mechanical stability, suitable electrical conductivity, and good cytocompatibility of the zwitterionic hydrogel, the Schwann cells cultured on the hydrogel could grow and proliferate better, and dorsal root ganglian had an increased neurite length. The zwitterionic hydrogel-based nerve guidance conduits were then implanted into a 10 mm sciatic nerve defect model in rats. Morphological analysis and electrophysiological data showed that the grafts achieved a regeneration effect close to that of the autologous nerve. Overall, our developed zwitterionic hydrogel facilitates efficient and efficacious peripheral nerve regeneration by mimicking the electrical and mechanical properties of the extracellular matrix and creating a suitable regeneration microenvironment, providing a new material reserve for the repair of peripheral nerve injury.


Assuntos
Materiais Biocompatíveis , Hidrogéis , Ratos , Animais , Hidrogéis/farmacologia , Materiais Biocompatíveis/farmacologia , Nervo Isquiático/fisiologia , Alicerces Teciduais , Regeneração Nervosa/fisiologia
15.
Neural Regen Res ; 18(12): 2757-2761, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37449641

RESUMO

Dendrites play irreplaceable roles in the nerve conduction pathway and are vulnerable to various insults. Peripheral axotomy of motor neurons results in the retraction of dendritic arbors, and the dendritic arbor can be re-expanded when reinnervation is allowed. RhoA is a target that regulates the cytoskeleton and promotes neuronal survival and axon regeneration. However, the role of RhoA in dendrite degeneration and regeneration is unknown. In this study, we explored the potential role of RhoA in dendrites. A line of motor neuronal RhoA conditional knockout mice was developed by crossbreeding HB9Cre+ mice with RhoAflox/flox mice. We established two models for assaying dendrite degeneration and regeneration, in which the brachial plexus was transection or crush injured, respectively. We found that at 28 days after brachial plexus transection, the density, complexity, and structural integrity of dendrites in the ventral horn of the spinal cord of RhoA conditional knockout mice were slightly decreased compared with that in Cre mice. Dendrites underwent degeneration at 7 and 14 days after brachial plexus transection and recovered at 28-56 days. The density, complexity, and structural integrity of dendrites in the ventral horn of the spinal cord of RhoA conditional knockout mice recovered compared with results in Cre mice. These findings suggest that RhoA knockout in motor neurons attenuates dendrite degeneration and promotes dendrite regeneration after peripheral nerve injury.

16.
Tissue Eng Part A ; 28(3-4): 111-124, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34157886

RESUMO

Bone defects caused by infection, tumor, trauma, and so on remain difficult to treat clinically. Bone tissue engineering (BTE) has great application prospect in promoting bone defect repair. Polycaprolactone (PCL) is a commonly used material for creating BTE scaffolds. In addition, self-assembling peptides (SAPs) can function as the extracellular matrix and promote osteogenesis and angiogenesis. In the work, a PCL scaffold was constructed by 3D printing, then integrated with bone marrow mesenchymal stem cells (BMSCs) and SAPs. The research aimed to assess the bone repair ability of PCL/BMSC/SAP implants. BMSC proliferation in PCL/SAP scaffolds was assessed via Cell Counting Kit-8. In vitro osteogenesis of BMSCs cultured in PCL/SAP scaffolds was assessed by alkaline phosphatase staining and activity assays. Enzyme-linked immunosorbent assays were also performed to detect the levels of osteogenic factors. The effects of BMSC-conditioned medium from 3D culture systems on the migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) were assessed by scratch, transwell, and tube formation assays. After 8 weeks of in vivo transplantation, radiography and histology were used to evaluate bone regeneration, and immunohistochemistry staining was utilized to detect neovascularization. In vitro results demonstrated that PCL/SAP scaffolds promoted BMSC proliferation and osteogenesis compared to PCL scaffolds, and the PCL/BMSC/SAP conditional medium (CM) enhanced HUVEC migration and angiogenesis compared to the PCL/BMSC CM. In vivo results showed that, compared to the blank control, PCL, and PCL/BMSC groups, the PCL/BMSC/SAP group had significantly increased bone and blood vessel formation. Thus, the combination of BMSC-seeded 3D-printed PCL and SAPs can be an effective approach for treating bone defects. Impact statement Both polycaprolactone (PCL) and self-assembling peptides (SAPs) have been broadly applied in bone defect repair. However, the poor osteoinductivity of PCL and weak mechanical strength of SAPs have limited their clinical application. Here, a 3D-printed PCL scaffold was fabricated for seeding bone marrow mesenchymal stem cells (BMSCs), then combined with SAPs to construct a composite PCL/BMSC/SAP implant for treating the calvarial defect. We showed that transplantation of PCL/BMSC/SAP composite implants clearly promoted bone regeneration and neovascularization. To our knowledge, this is the first study to treat bone defects by combination of BMSC-seeded 3D-printed PCL and SAPs.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Regeneração Óssea , Diferenciação Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos/farmacologia , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais
17.
Mol Neurobiol ; 59(1): 429-444, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34708329

RESUMO

Silent information regulator 6 (SIRT6) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Previous studies have been reported a pro-regenerative role of SIRT6 in central nervous system injury. However, the role of SIRT6 in peripheral nerve injury is still unknown. Given the importance and necessity of Schwann cell dedifferentiation response to peripheral nerve injury, we aim to investigate the molecular mechanism of SIRT6 steering Schwann cell dedifferentiation during Wallerian degeneration in injured peripheral nerve. Herein, we first examined the expression pattern of SIRT6 after peripheral nerve injury. Using the explants of sciatic nerve, an ex vivo model of nerve degeneration, we provided evidences indicating that SIRT6 inhibitor accelerates Schwann cell dedifferentiation as well as axonal and myelin degeneration, while SIRT6 activator attenuates this process. Moreover, in an in vitro Schwann cell dedifferentiation model, we found SIRT6 inhibitor promotes Schwann cell dedifferentiation through upregulating the expression of c-Jun. In addition, downregulation of c-Jun reverse the effects of SIRT6 inhibition on the Schwann cells dedifferentiation and axonal and myelin degeneration. In summary, we first described SIRT6 acts as a negative regulator for Schwann cells dedifferentiation during Wallerian degeneration and c-Jun worked as a direct downstream partner of SIRT6 in injured peripheral nerve.


Assuntos
Desdiferenciação Celular/fisiologia , Traumatismos dos Nervos Periféricos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Células de Schwann/metabolismo , Sirtuínas/metabolismo , Degeneração Walleriana/metabolismo , Animais , Desdiferenciação Celular/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/patologia , Ratos , Células de Schwann/efeitos dos fármacos , Sirtuínas/antagonistas & inibidores , Degeneração Walleriana/patologia
18.
Biomed Mater ; 17(6)2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36170855

RESUMO

Wound healing of skin defects is complex. For the treatment of large and deep wounds, it is a good alternative to accept artificial dermis grafting at the first stage surgery, and autologous split-thickness skin grafting 2-3 weeks later at the second stage surgery. In addition, the effectiveness of numerous cytokines such as fibroblast growth factor (FGF) on wounds healing has been widely researched. The traditional view is that direct external application orin vivoinjection of exogenous FGFs may not achieve the desired therapeutic effect as the effective concentration cannot be maintained for a long time. Therefore, some researchers have tried to integrate various cytokines into skin substitutes for combined application. However, we believe that considering the current situation, it is still difficult to achieve mass production of these types of artificial dermis. Here, we manufactured a collagen-chondroitin sulfate scaffold material by imitating the marketed artificial dermis materials. Then, we combined it with recombinant human acidic FGF in a single full dose during the first-stage artificial dermis transplantation, which is simple and completely feasible but always controversial in the current clinical work, to explore whether this combinatorial therapy could serve as an efficient way wound healing in the Balb/c-nu mice full-thickness skin defect model.


Assuntos
Transplante de Pele , Pele Artificial , Animais , Sulfatos de Condroitina , Colágeno , Citocinas , Fator 1 de Crescimento de Fibroblastos , Humanos , Camundongos , Camundongos Nus
19.
Commun Biol ; 5(1): 495, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614315

RESUMO

The chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) has been used in the treatment and repair of cartilage defects; however, the in-depth regulatory mechanisms by which RNA modifications are involved in this process are still poorly understood. Here, we found that Sox9, a critical transcription factor that mediates chondrogenic differentiation, exhibited enhanced translation by ribosome sequencing in chondrogenic pellets, which was accompanied by increased 5-methylcytosine (m5C) and N6-methyladenosine (m6A) levels. Nsun4-mediated m5C and Mettl3-mediated m6A modifications were required for Sox9-regulated chondrogenic differentiation. Interestingly, we showed that in the 3'UTR of Sox9 mRNA, Nsun4 catalyzed the m5C modification and Mettl3 catalyzed the m6A modification. Furthermore, we found that Nsun4 and Mettl3 co-regulated the translational reprogramming of Sox9 via the formation of a complex. Surface plasmon resonance (SPR) assays showed that this complex was assembled along with the recruitment of Ythdf2 and eEF1α-1. Moreover, BMSCs overexpressing Mettl3 and Nsun4 can promote the repair of cartilage defects in vivo. Taken together, our study demonstrates that m5C and m6A co-regulate the translation of Sox9 during the chondrogenic differentiation of BMSCs, which provides a therapeutic target for clinical implications.


Assuntos
Condrogênese , Células-Tronco Mesenquimais , Adenosina , Diferenciação Celular/genética , Condrogênese/genética , RNA Mensageiro
20.
Tissue Eng Part A ; 28(15-16): 700-711, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35152730

RESUMO

The combination of three-dimensional (3D) printed scaffold materials and various cytokines can achieve the purpose of tissue reconstruction more efficiently. In this study, we prepared platelet-rich plasma (PRP)/gelatin microspheres combined with 3D printed polycaprolactone/ß-tricalcium phosphate scaffolds to solve the key problem that PRP cannot be released under control and the release time is too short, and thus better promote bone repair. Consequently, the composite scaffold displayed a good mechanical property and sustained cytokine release for ∼3 weeks. Increased survival, proliferation, migration, and osteogenic and angiogenic differentiation of bone marrow mesenchymal stem cells were observed compared with the control groups. The in vivo study demonstrated that the composite scaffold with PRP/gelatin microspheres led to greater positive effects in promoting large bone defect repair. In conclusion, in this study, a new type of PRP long-term sustained-release composite scaffold material was constructed that effectively improved the survival, proliferation, and differentiation of cells in the transplanted area, thereby better promoting the repair of large bone defects. Impact statement Reconstruction of bone tissue and blood vessels at bone defects takes time. Platelet-rich plasma (PRP) has been widely used in bone defect repair because it contains a variety of cytokine that can promote local osteogenesis and angiogenesis. In this study, we constructed a new type of polycaprolactone/ß-tricalcium phosphate/PRP/gelatin scaffold to solve the predicament of short cytokine release time in PRP-related materials. We proved that this scaffold can not only achieve long-term PRP-related cytokine release (more than 3 weeks) but also promote osteogenesis and bone defect repair. We believe that this is a novel concept of developing the sustained PRP-related cytokine releasing bioscaffold for treating large bone defect.


Assuntos
Citocinas , Plasma Rico em Plaquetas , Regeneração Óssea , Gelatina , Osteogênese , Impressão Tridimensional , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA