Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 22(1): 333, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877492

RESUMO

In the realm of large-area trauma flap transplantation, averting ischaemic necrosis emerges as a pivotal concern. Several key mechanisms, including the promotion of angiogenesis, the inhibition of oxidative stress, the suppression of cell death, and the mitigation of inflammation, are crucial for enhancing skin flap survival. Apoptotic bodies (ABs), arising from cell apoptosis, have recently emerged as significant contributors to these functions. This study engineered three-dimensional (3D)-ABs using tissue-like mouse adipose-derived stem cells (mADSCs) cultured in a 3D environment to compare their superior biological effects against 2D-ABs in bolstering skin flap survival. The findings reveal that 3D-ABs (85.74 ± 4.51) % outperform 2D-ABs (76.48 ± 5.04) % in enhancing the survival rate of ischaemic skin flaps (60.45 ± 8.95) % (all p < 0.05). Mechanistically, they stimulated angiogenesis, mitigated oxidative stress, suppressed apoptosis, and facilitated the transition of macrophages from M1 to M2 polarization (all p < 0.05). A comparative analysis of microRNA (miRNA) profiles in 3D- and 2D-ABs identified several specific miRNAs (miR-423-5p-up, miR30b-5p-down, etc.) with pertinent roles. In summary, ABs derived from mADSCs cultured in a 3D spheroid-like arrangement exhibit heightened biological activity compared to those from 2D-cultured mADSCs and are more effective in promoting ischaemic skin flap survival. These effects are attributed to their influence on specific miRNAs.


Assuntos
Tecido Adiposo , Apoptose , Isquemia , MicroRNAs , Animais , Camundongos , Tecido Adiposo/citologia , MicroRNAs/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Estresse Oxidativo , Retalhos Cirúrgicos , Células Cultivadas , Camundongos Endogâmicos C57BL , Masculino , Sobrevivência Celular , Neovascularização Fisiológica , Técnicas de Cultura de Células em Três Dimensões/métodos
2.
Eur Spine J ; 33(3): 1069-1080, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38246903

RESUMO

PURPOSE: To compare the clinical outcomes and radiographic outcomes of cortical bone trajectory (CBT) and traditional trajectory (TT) pedicle screw fixation in patients treated with single-level transforaminal lumbar interbody fusion (TLIF). METHODS: This trial included a total of 224 patients with lumbar spine disease who required single-level TLIF surgery. Patients were randomly assigned to the CBT and TT groups at a 1:1 ratio. Demographics and clinical and radiographic data were collected to evaluate the efficacy and safety of CBT and TT screw fixation in TLIF. RESULTS: The baseline characteristic data were similar between the CBT and TT groups. Back and leg pain for both the CBT and TT groups improved significantly from baseline to 24 months postoperatively. The CBT group experienced less pain than the TT group at one week postoperatively. The postoperative radiographic results showed that the accuracy of screw placement was significantly increased in the CBT group compared with the TT group (P < 0.05). The CBT group had a significantly lower rate of FJV than the TT group (P < 0.05). In addition, the rate of fusion and the rate of screw loosening were similar between the CBT and TT groups according to screw loosening criteria. CONCLUSION: This prospective, randomized controlled analysis suggests that clinical outcomes and radiographic characteristics, including fusion rates and caudal screw loosening rates, were comparable between CBT and TT screw fixation. Compared with the TT group, the CBT group showed advantages in the accuracy of screw placement and the FJV rate. CLINICAL TRIALS REGISTRATION: This trial has been registered at the US National Institutes of Health Clinical Trials Registry: NCT03105167.


Assuntos
Parafusos Pediculares , Fusão Vertebral , Humanos , Parafusos Pediculares/efeitos adversos , Fusão Vertebral/métodos , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia , Estudos Prospectivos , Resultado do Tratamento , Osso Cortical/diagnóstico por imagem , Osso Cortical/cirurgia , Dor/etiologia
3.
J Neuroinflammation ; 20(1): 6, 2023 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-36609266

RESUMO

Spinal cord injury (SCI) is a devastating injury that may result in permanent motor impairment. The active ingredients of medications are unable to reach the affected area due to the blood‒brain barrier. Elamipretide (SS-31) is a new and innovative aromatic cationic peptide. Because of its alternating aromatic and cationic groups, it freely crosses the blood‒brain barrier. It is also believed to decrease inflammation and protect against a variety of neurological illnesses. This study explored the therapeutic value of SS-31 in functional recovery after SCI and its possible underlying mechanism. A spinal cord contusion injury model as well as the Basso Mouse Scale, footprint assessment, and inclined plane test were employed to assess how well individuals could function following SCI. The area of glial scarring, the number of dendrites, and the number of synapses after SCI were confirmed by HE, Masson, MAP2, and Syn staining. Western blotting, immunofluorescence, and enzyme-linked immunosorbent assays were employed to examine the expression levels of pyroptosis-, autophagy-, lysosomal membrane permeabilization (LMP)- and MAPK signalling-related proteins. The outcomes showed that SS-31 inhibited pyroptosis, enhanced autophagy and attenuated LMP in SCI. Mechanistically, we applied AAV vectors to upregulate Pla2g4A in vivo and found that SS-31 enhanced autophagy and attenuated pyroptosis and LMP by inhibiting phosphorylation of cPLA2. Ultimately, we applied asiatic acid (a p38-MAPK agonist) to test whether SS-31 regulated cPLA2 partially through the MAPK-P38 signalling pathway. Our group is the first to suggest that SS-31 promotes functional recovery partially by inhibiting cPLA2-mediated autophagy impairment and preventing LMP and pyroptosis after SCI, which may have potential clinical application value.


Assuntos
Piroptose , Traumatismos da Medula Espinal , Camundongos , Animais , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Lisossomos/metabolismo , Fosfolipases A2 Citosólicas/metabolismo
4.
J Neuroinflammation ; 19(1): 242, 2022 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-36195926

RESUMO

Stimulator of interferons genes (STING), which is crucial for the secretion of type I interferons and proinflammatory cytokines in response to cytosolic nucleic acids, plays a key role in the innate immune system. Studies have revealed the participation of the STING pathway in unregulated inflammatory processes, traumatic brain injury (TBI), spinal cord injury (SCI), subarachnoid haemorrhage (SAH) and hypoxic-ischaemic encephalopathy (HIE). STING signalling is markedly increased in CNS injury, and STING agonists might facilitate the pathogenesis of CNS injury. However, the effects of STING-regulated signalling activation in CNS injury are not well understood. Aberrant activation of STING increases inflammatory events, type I interferon responses, and cell death. cGAS is the primary pathway that induces STING activation. Herein, we provide a comprehensive review of the latest findings related to STING signalling and the cGAS-STING pathway and highlight the control mechanisms and their functions in CNS injury. Furthermore, we summarize and explore the most recent advances toward obtaining an understanding of the involvement of STING signalling in programmed cell death (autophagy, necroptosis, ferroptosis and pyroptosis) during CNS injury. We also review potential therapeutic agents that are capable of regulating the cGAS-STING signalling pathway, which facilitates our understanding of cGAS-STING signalling functions in CNS injury and the potential value of this signalling pathway as a treatment target.


Assuntos
Doenças do Sistema Nervoso Central , Ferroptose , Interferon Tipo I , Ácidos Nucleicos , Autofagia/fisiologia , Citocinas/metabolismo , Humanos , Inflamação , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Necroptose , Nucleotidiltransferases/metabolismo , Piroptose
5.
J Cell Physiol ; 236(5): 3641-3659, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33044023

RESUMO

Random-pattern skin flaps are widely applied to rebuild and restore soft-tissue damage in reconstructive surgery; however, ischemia and subsequent ischemia-reperfusion injury lead to flap necrosis and are major complications. Exenatide, a glucagon-like peptide-1 analog, exerts therapeutic benefits for diabetic wounds, cardiac injury, and nonalcoholic fatty liver disease. Furthermore, Exenatide is a known activator of autophagy, which is a complex process of subcellular degradation that may enhance the viability of random skin flaps. In this study, we explored whether exenatide can improve skin flap survival. Our results showed that exenatide augments autophagy, increases flap viability, enhances angiogenesis, reduces oxidative stress, and alleviates pyroptosis. Coadministration of exenatide with 3-methyladenine and chloroquine, potent inhibitors of autophagy, reversed the beneficial effects, suggesting that the therapeutic benefits of exenatide for skin flaps are due largely to autophagy activation. Mechanistically, we identified that exenatide enhanced activation and nuclear translocation of TFE3, which leads to autophagy activation. Furthermore, we found that exenatide activates the AMPK-SKP2-CARM1 and AMPK-mTOR signaling pathways, which likely lead to exenatide's effects on activating TFE3. Overall, our findings suggest that exenatide may be a potent therapy to prevent flap necrosis, and we also reveal novel mechanistic insight into exenatide's effect on flap survival.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Exenatida/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Pele , Pele/irrigação sanguínea , Adenina/análogos & derivados , Adenina/farmacologia , Adenilato Quinase/metabolismo , Animais , Autofagia/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Regulação para Baixo/efeitos dos fármacos , Edema/patologia , Masculino , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteína-Arginina N-Metiltransferases/metabolismo , Piroptose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Cell Biol Toxicol ; 37(3): 401-419, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32740777

RESUMO

Spinal cord injury (SCI) is a devastating neurological trauma that causes losses of motor and sensory function. Sestrin2, also known as hypoxia inducible gene 95, is emerging as a critical determinant of cell homeostasis in response to cellular stress. However, the role of sestrin2 in the neuronal response to endoplasmic reticulum (ER) stress and the potential mechanism remain undefined. In this study, we investigated the effects of sestrin2 on ER stress and delineated an underlying molecular mechanism after SCI. Here, we found that elevated sestrin2 is a protective process in neurons against chemical ER stress induced by tunicamycin (TM) or traumatic invasion, while treatment with PERK inhibitor or knockdown of ATF4 reduces sestrin2 expression upon ER stress. In addition, we demonstrated that overexpression of sestrin2 limits ER stress, promoting neuronal survival and improving functional recovery after SCI, which is associated with activation of autophagy and restoration of autophagic flux mediated by sestrin2. Moreover, we also found that sestrin2 activates autophagy dependent on the AMPK-mTOR signaling pathway. Consistently, inhibition of AMPK abrogates the effect of sestrin2 on the activation of autophagy, and blockage of autophagic flux abolishes the effect of sestrin2 on limiting ER stress and neural death. Together, our data reveal that upregulation of sestrin2 is an important resistance mechanism of neurons to ER stress and the potential role of sestrin2 as a therapeutic target for SCI. Graphical abstract.


Assuntos
Fator 4 Ativador da Transcrição/genética , Autofagia/genética , Peroxidases/genética , Traumatismos da Medula Espinal/genética , Quinases Proteína-Quinases Ativadas por AMP/genética , Animais , Apoptose/genética , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais/genética , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Serina-Treonina Quinases TOR/genética , Tunicamicina/farmacologia
7.
Scand J Immunol ; 91(2): e12843, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31657484

RESUMO

Inflammatory bowel disease (IBD) is a chronic, non-specific, inflammatory gastrointestinal disease that mainly consists of Crohn's disease and ulcerative colitis. However, the aetiology and pathogenesis of IBD are still unclear. B10 (IL-10 producing regulatory B) cells, a subset of regulatory B cells, are known to contribute to intestinal homeostasis and the aberrant frequency of B10 cells is associated with IBD. We have recently reported that B10 cells can be induced by ManLAM (mannose-capped lipoarabinomannan), a major cell-wall lipoglycan of M tb (Mycobacterium tuberculosis). In the current study, the ManLAM-induced B10 cells were adoptively transferred into IL(interleukin)-10-/- mice and the roles of ManLAM-induced B10 cells were investigated in DSS (dextran sodium sulphate)-induced IBD model. ManLAM-induced B10 cells decrease colitis severity in the mice. The B10 cells downregulate Th1 polarization in spleen and MLNs (mesenteric lymph nodes) of DSS-treated mice. These results suggest that IL-10 production by ManLAM-treated B cells contributes to keeping the balance between CD4+ T cell subsets and protect mice from DSS-induced IBD.


Assuntos
Linfócitos B Reguladores/imunologia , Doenças Inflamatórias Intestinais/imunologia , Interleucina-10/metabolismo , Lipopolissacarídeos/metabolismo , Manose/metabolismo , Mycobacterium tuberculosis/metabolismo , Células Th1/imunologia , Animais , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Tolerância Imunológica , Doenças Inflamatórias Intestinais/induzido quimicamente , Lipopolissacarídeos/imunologia , Manose/imunologia , Camundongos , Camundongos Knockout
8.
Z Gerontol Geriatr ; 53(2): 163-170, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31950363

RESUMO

Osteoporotic bones heal more slowly and ineffectively than normal bones. A combination of antibodies against sclerosing protein (Scl-Ab), and parathyroid hormone 1-34 (PTH 1-34) may improve healing. A standard osteoporotic rat model was established 12 weeks after bilateral ovarian resection (OVX). Bone defects were created in the right femora of 80 rats, which were randomly divided into 4 groups: control, Scl-Ab (25 mg/kg twice weekly), PTH (60 µg/kg of PTH 1-34 three times a week) and PTH plus Scl-Ab. After 12 weeks of treatment the rats were sacrificed and blood and the distal femora were harvested for biochemical evaluation, histology, microcomputed tomography and biomechanical testing. Compared to the control group, monotherapy and combination therapy with PTH and/or Scl-Ab promoted the formation of new bone, enhanced maximum femoral loading and increased the levels of procollagen type I N­terminal propeptide (PINP) and osteocalcin. The administration of PTH + Scl-Ab maximally enhanced bone defect healing. Combination treatment was better than either treatment alone, indicating a synergistic effect.


Assuntos
Anticorpos/administração & dosagem , Proteínas Morfogenéticas Ósseas/imunologia , Remodelação Óssea/fisiologia , Consolidação da Fratura/efeitos dos fármacos , Hormônio Paratireóideo/uso terapêutico , Animais , Densidade Óssea/efeitos dos fármacos , Calo Ósseo/efeitos dos fármacos , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Humanos , Ovariectomia , Hormônio Paratireóideo/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X/métodos
9.
Int Orthop ; 43(9): 2141-2149, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30903254

RESUMO

PURPOSE: Displaced intra-articular calcaneus fractures Sanders type IV(DIACFS IV) can result in an unsatisfactory prognosis and a high complication rate. Our investigation intends to compare the outcomes of DIACFS IV treated by open reduction and internal fixation (ORIF) via sinus tarsi approach (STA) with these via extensile lateral approach (ELA). METHODS: Sixty-nine patients (82 ft) with DIACFS IV who were treated with ORIF (29 in STA group and 40 in ELA group) were retrospectively assessed. Median follow-up was 50 months in two groups. Radiographic results were reviewed pre-operatively and post-operatively, and relative complications were collected. Clinical outcomes were evaluated using the American Orthopaedic Foot and Ankle Society (AOFAS) score and visual analog scale (VAS). RESULTS: The wound-healing complication rate was 14.28% in STA group and 34.04% in ELA group (p = .043), and overall complication rate was 54% and 77% (p = .056), respectively. Seven cases of sural nerve injury only occurred in ELA group. The post-operative radiographs of the calcaneus (Böhler's angle, height, width, and length) were significantly different from those measured pre-operatively in each group. And these data were parallel between the two groups. In STA and ELA groups, the average AOFAS was 75.45 versus 72.44 (p = .496), and the mean VAS was 23.95 versus 30.93 (p = .088), respectively. CONCLUSION: Similar clinical and radiographic outcomes are achieved between STA and ELA. And STA has a lower incidence of wound healing complication and sural nerve injury. Therefore, ORIF via STA can be a considerable management for DIACFS IV.


Assuntos
Calcâneo/lesões , Calcâneo/cirurgia , Traumatismos do Pé/cirurgia , Fixação Interna de Fraturas/métodos , Fraturas Intra-Articulares/cirurgia , Adulto , Calcâneo/diagnóstico por imagem , Feminino , Traumatismos do Pé/diagnóstico por imagem , Fixação Interna de Fraturas/efeitos adversos , Humanos , Fraturas Intra-Articulares/classificação , Fraturas Intra-Articulares/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Redução Aberta/métodos , Prognóstico , Estudos Retrospectivos , Adulto Jovem
10.
Z Gerontol Geriatr ; 51(8): 875-881, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29043444

RESUMO

The effects of both alendronate (ALN) and Drynaria rhizome extracts (DRE) alone could promote bone healing in osteoporotic fractures but there are no reports about the combined use of ALN and DRE for promotion of bone healing of fractures in osteoporotic settings. This study investigated the effects of ALN plus DRE on fractures in osteopenic rats. Osteopenic rats underwent unilateral transverse osteotomy on the femur fixed by a sterilized Kirschner wire 2 weeks after intragastric administration of retinoic acid (80 mg/kg body weight/day). Subsequently, the animals were randomly divided into four groups: control, ALN, DRE and ALN + DRE. All rats from groups ALN, DRE and ALN + DRE received ALN (40 mg/kg, weekly), DRE (90 mg/kg/day), or both for 2, 4 and 6 weeks. The results of our study indicated that all treatment promoted fracture healing and callus formation compared to controls but ALN + DRE treatment showed significantly stronger effects than ALN or DRE alone in histological, X­ray and biomechanical tests. These results seem to indicate that combined treatment with ALN and DRE has an additive effect on fracture healing and callus formation in osteoporotic rats.


Assuntos
Conservadores da Densidade Óssea , Fraturas por Osteoporose , Extratos Vegetais , Polypodiaceae , Alendronato , Animais , Conservadores da Densidade Óssea/uso terapêutico , Feminino , Consolidação da Fratura , Fraturas por Osteoporose/tratamento farmacológico , Ovariectomia , Extratos Vegetais/uso terapêutico , Polypodiaceae/química , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Rizoma
11.
Int J Mol Sci ; 18(2)2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28230791

RESUMO

Previous studies have indicated that autophagy plays a critical role in spinal cord injury (SCI), including traumatic spinal cord injury (TSCI) and ischemia-reperfusion spinal cord injury (IRSCI). However, while the understanding of mechanisms underlying autophagy in SCI has progressed, there remain several controversial points: (1) temporal pattern results of autophagic activation after SCI are not consistent across studies; (2) effect of accumulation of autophagosomes due to the blockade or enhancement of autophagic flux is uncertain; (3) overall effect of enhanced autophagy remains undefined, with both beneficial and detrimental outcomes reported in SCI literature. In this review, the temporal pattern of autophagic activation, autophagic flux, autophagic cell death, relationship between autophagy and apoptosis, and pharmacological intervention of autophagy in TSCI (contusion injury, compression injury and hemisection injury) and IRSCI are discussed. Types of SCI and severity appear to contribute to differences in outcomes regarding temporal pattern, flux, and function of autophagy. With future development of specific strategies on autophagy intervention, autophagy may play an important role in improving functional recovery in patients with SCI.


Assuntos
Autofagia , Traumatismos da Medula Espinal/metabolismo , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Biomarcadores , Morte Celular/efeitos dos fármacos , Lisossomos/metabolismo , Fagossomos/metabolismo , Traumatismos da Medula Espinal/diagnóstico , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/etiologia
12.
J Adv Res ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38704090

RESUMO

BACKGROUND: Central nervous system (CNS) injury causes severe organ damage due to both damage resulting from the injury and subsequent cell death. However, there are currently no effective treatments for countering the irreversible loss of cell function. Parthanatos is a poly (ADP-ribose) polymerase 1 (PARP-1)-dependent form of programmed cell death that is partly responsible for neural cell death. Consequently, the mechanism by which parthanatos promotes CNS injury has attracted significant scientific interest. AIM OF REVIEW: Our review aims to summarize the potential role of parthanatos in CNS injury and its molecular and pathophysiological mechanisms. Understanding the role of parthanatos and related molecules in CNS injury is crucial for developing effective treatment strategies and identifying important directions for future in-depth research. KEY SCIENTIFIC CONCEPTS OF REVIEW: Parthanatos (from Thanatos, the personification of death according to Greek mythology) is a type of programmed cell death that is initiated by the overactivation of PARP-1. This process triggers a cascade of reactions, including the accumulation of poly(ADP-ribose) (PAR), the nuclear translocation of apoptosis-inducing factor (AIF) after its release from mitochondria, and subsequent massive DNA fragmentation caused by migration inhibitory factor (MIF) forming a complex with AIF. Secondary molecular mechanisms, such as excitotoxicity and oxidative stress-induced overactivation of PARP-1, significantly exacerbate neuronal damage following initial mechanical injury to the CNS. Furthermore, parthanatos is not only associated with neuronal damage but also interacts with various other types of cell death. This review focuses on the latest research concerning the parthanatos cell death pathway, particularly considering its regulatory mechanisms and functions in CNS damage. We highlight the associations between parthanatos and different cell types involved in CNS damage and discuss potential therapeutic agents targeting the parthanatos pathway.

13.
Mol Neurobiol ; 61(1): 55-73, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37581847

RESUMO

Spinal cord injury (SCI) is a severe medical condition with lasting effects. The efficacy of numerous clinical treatments is hampered by the intricate pathophysiological mechanism of SCI. Fibroblast growth factor 18 (FGF-18) has been found to exert neuroprotective effects after brain ischaemia, but its effect after SCI has not been well explored. The aim of the present study was to explore the therapeutic effect of FGF-18 on SCI and the related mechanism. In the present study, a mouse model of SCI was used, and the results showed that FGF-18 may significantly affect functional recovery. The present findings demonstrated that FGF-18 directly promoted functional recovery by increasing autophagy and decreasing pyroptosis. In addition, FGF-18 increased autophagy, and the well-known autophagy inhibitor 3-methyladenine (3MA) reversed the therapeutic benefits of FGF-18 after SCI, suggesting that autophagy mediates the therapeutic effects of FGF-18 on SCI. A mechanistic study revealed that after stimulation of the protein kinase B (AKT)-transient receptor potential mucolipin 1 (TRPML1)-calcineurin signalling pathway, the FGF-18-induced increase in autophagy was mediated by the dephosphorylation and nuclear translocation of transcription factor E3 (TFE3). Together, these findings indicated that FGF-18 is a robust autophagy modulator capable of accelerating functional recovery after SCI, suggesting that it may be a promising treatment for SCI in the clinic.


Assuntos
Fatores de Crescimento de Fibroblastos , Proteínas Proto-Oncogênicas c-akt , Traumatismos da Medula Espinal , Ratos , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piroptose , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/metabolismo , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Autofagia
14.
Br J Pharmacol ; 181(7): 1068-1090, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37850255

RESUMO

BACKGROUND AND PURPOSE: Ischaemia-reperfusion (I/R) injury is a major contributor to skin flap necrosis, which presents a challenge in achieving satisfactory therapeutic outcomes. Previous studies showed that cathelicidin-BF (BF-30) protects tissues from I/R injury. In this investigation, BF-30 was synthesized and its role and mechanism in promoting survival of I/R-injured skin flaps explored. EXPERIMENTAL APPROACH: Survival rate analysis and laser Doppler blood flow analysis were used to evaluate I/R-injured flap viability. Western blotting, immunofluorescence, TdT-mediated dUTP nick end labelling (TUNEL) and dihydroethidium were utilized to examine the levels of apoptosis, pyroptosis, oxidative stress, transcription factor EB (TFEB)-mediated autophagy and molecules related to the adenosine 5'-monophosphate-activated protein kinase (AMPK)-transient receptor potential mucolipin 1 (TRPML1)-calcineurin signalling pathway. KEY RESULTS: The outcomes revealed that BF-30 enhanced I/R-injured island skin flap viability. Autophagy, oxidative stress, pyroptosis and apoptosis were related to the BF-30 capability to enhance I/R-injured flap survival. Improved autophagy flux and tolerance to oxidative stress promoted the inhibition of apoptosis and pyroptosis in vascular endothelial cells. Activation of TFEB increased autophagy and inhibited endothelial cell oxidative stress in I/R-injured flaps. A reduction in TFEB level led to a loss of the protective effect of BF-30, by reducing autophagy flux and increasing the accumulation of reactive oxygen species (ROS) in endothelial cells. Additionally, BF-30 modulated TFEB activity via the AMPK-TRPML1-calcineurin signalling pathway. CONCLUSION AND IMPLICATIONS: BF-30 promotes I/R-injured skin flap survival by TFEB-mediated up-regulation of autophagy and inhibition of oxidative stress, which may have possible clinical applications.


Assuntos
Piroptose , Traumatismo por Reperfusão , Humanos , Espécies Reativas de Oxigênio/metabolismo , Catelicidinas/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Células Endoteliais/metabolismo , Calcineurina/farmacologia , Autofagia , Traumatismo por Reperfusão/metabolismo , Fatores de Transcrição
15.
Br J Pharmacol ; 181(5): 712-734, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37766498

RESUMO

BACKGROUND AND PURPOSE: Autophagy is a protective factor for controlling neuronal damage, while necroptosis promotes neuroinflammation after spinal cord injury (SCI). DADLE (D-Ala2 , D-Leu5 ]-enkephalin) is a selective agonist for delta (δ) opioid receptor and has been identified as a promising drug for neuroprotection. The aim of this study was to investigate the mechanism/s by which DADLE causes locomotor recovery following SCI. EXPERIMENTAL APPROACH: Spinal cord contusion model was used and DADLE was given by i.p. (16 mg·kg-1 ) in mice for following experiments. Motor function was assessed by footprint and Basso mouse scale (BMS) score analysis. Western blotting used to evaluate related protein expression. Immunofluorescence showed the protein expression in each cell and its distribution. Network pharmacology analysis was used to find the related signalling pathways. KEY RESULTS: DADLE promoted functional recovery after SCI. In SCI model of mice, DADLE significantly increased autophagic flux and inhibited necroptosis. Concurrently, DADLE restored autophagic flux by decreasing lysosomal membrane permeabilization (LMP). Additionally, chloroquine administration reversed the protective effect of DADLE to inhibit necroptosis. Further analysis showed that DADLE decreased phosphorylated cPLA2 , overexpression of cPLA2 partially reversed DADLE inhibitory effect on LMP and necroptosis, as well as the promotion autophagy. Finally, AMPK/SIRT1/p38 pathway regulating cPLA2 is involved in the action DADLE on SCI and naltrindole inhibited DADLE action on δ receptor and on AMPK signalling pathway. CONCLUSION AND IMPLICATION: DADLE causes its neuroprotective effects on SCI by promoting autophagic flux and inhibiting necroptosis by decreasing LMP via activating δ receptor/AMPK/SIRT1/p38/cPLA2 pathway.


Assuntos
Leucina Encefalina-2-Alanina , Traumatismos da Medula Espinal , Animais , Camundongos , Proteínas Quinases Ativadas por AMP/metabolismo , Leucina Encefalina-2-Alanina/metabolismo , Leucina Encefalina-2-Alanina/farmacologia , Lisossomos/metabolismo , Fosfolipases/metabolismo , Receptores Opioides delta/metabolismo , Recuperação de Função Fisiológica , Sirtuína 1/metabolismo , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo
16.
Adv Healthc Mater ; 13(12): e2303462, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38243745

RESUMO

Oxidative stress (OS) is one of the crucial molecular events of secondary spinal cord injury (SCI). Basic fibroblast growth factor (bFGF) is a multipotent cell growth factor with an anti-oxidant effect. However, bFGF has a short half-life in vivo, which limits its therapeutic application. Biodegradable polymers with excellent biocompatibility have been recently applied in SCI. The negative aspect is that polymers cannot provide a significant therapeutic effect. Betulinic acid (BA), a natural anti-inflammatory compound, has been polymerized into poly (betulinic acid) (PBA) to serve as a drug carrier for bFGF. This study explores the therapeutic effects and underlying molecular mechanisms of PBA nanoparticles (NPs) loaded with bFGF (PBA-bFGF NPs) in SCI. Results show that PBA-bFGF NPs produce remarkable biocompatibility in vivo and in vitro. The results also demonstrate that local delivery of PBA-bFGF NPs enhances motor function recovery, inhibits OS, mitigates neuroinflammation, and alleviates neuronal apoptosis following SCI. Furthermore, the results indicate that local delivery of PBA-bFGF NPs activates the nuclear factor erythroid 2-related factor 2 (Nrf-2) signaling pathway following SCI. In summary, results suggest that local delivery of PBA-bFGF NPs delivers potential therapeutic advantages in the treatment and management of SCI.


Assuntos
Ácido Betulínico , Fator 2 de Crescimento de Fibroblastos , Nanopartículas , Traumatismos da Medula Espinal , Animais , Masculino , Ratos , Apoptose/efeitos dos fármacos , Ácido Betulínico/química , Portadores de Fármacos/química , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/química , Fator 2 de Crescimento de Fibroblastos/farmacologia , Nanopartículas/química , Nanopartículas/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Polímeros/química , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico
17.
J Agric Food Chem ; 72(26): 14727-14746, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38907713

RESUMO

Background: Following spinal cord injury (SCI), autophagy plays a positive role in neuronal protection, whereas pyroptosis triggers an inflammatory response. Ginsenoside-Rh2 (GRh2), known for its neuroprotective effects, is considered a promising drug. However, the exact molecular mechanisms underlying these protective effects remain unclear. Aim of the Study: Explore the therapeutic value of GRh2 in SCI and its potential mechanisms of action. Materials and Methods: An SCI mouse model was established, followed by random grouping and drug treatments under different conditions. Subsequently, the functional recovery of SCI mice after GRh2 treatment was assessed using hematoxylin and eosin, Masson's trichrome, and Nissl staining, footprint analysis, Basso Mouse Scale scoring, and inclined plane tests. The expression levels of relevant indicators in the mice were detected using Western blotting, immunofluorescence, and a quantitative polymerase chain reaction. Network pharmacology analysis was used to identify the relevant signaling pathways through which GRh2 exerts its therapeutic effects. Results: GRh2 promoted functional recovery after SCI. GRh2 significantly inhibits pyroptosis by enhancing autophagy in SCI mice. Simultaneously, the neuroprotective effect of GRh2, achieved through the inhibition of pyroptosis, is partially reversed by 3-methyladenine, an autophagy inhibitor. Additionally, the increase in autophagy induced by GRh2 is mediated by the promotion of transcription factor EB (TFEB) nuclear translocation and dephosphorylation. Partial attenuation of the protective effects of GRh2 was observed after TFEB knockdown. Additionally, GRh2 can modulate the activity of TFEB in mice post-SCI through the EGFR-MAPK signaling pathway, and NSC228155 (an EGFR activator) can partially reverse the effect of GRh2 on the EGFR-MAPK signaling pathway. Conclusions: GRh2 improves functional recovery after SCI by upregulating TFEB-mediated autophagic flux and inhibiting pyroptosis, indicating its potential clinical applicability.


Assuntos
Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Ginsenosídeos , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/genética , Ginsenosídeos/farmacologia , Ginsenosídeos/administração & dosagem , Autofagia/efeitos dos fármacos , Camundongos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/administração & dosagem , Masculino , Modelos Animais de Doenças
18.
Adv Sci (Weinh) ; 11(24): e2307238, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38639443

RESUMO

Preventing and treating avascular necrosis at the distal end of the flaps are critical to surgery success, but current treatments are not ideal. A recent study shows that apoptotic bodies (ABs) generated near the site of apoptosis can be taken up and promote cell proliferation. The study reveals that ABs derived from fibroblast-like cells in the subcutaneous connective tissue (FSCT cells) of skin flaps promoted ischaemic flap survival. It is also found that ABs inhibited cell death and oxidative stress and promoted M1-to-M2 polarization in macrophages. Transcriptome sequencing and protein level testing demonstrated that ABs promoted ischaemic flap survival in endothelial cells and macrophages by inhibiting ferroptosis via the KEAP1-Nrf2 axis. Furthermore, microRNA (miR) sequencing data and in vitro and in vivo experiments demonstrated that ABs inhibited KEAP1 by delivering miR-339-5p to exert therapeutic effects. In conclusion, FSCT cell-derived ABs inhibited ferroptosis, promoted the macrophage M1-to-M2 transition via the miR-339-5p/KEAP1/Nrf2 axis and promoted ischaemic flap survival. These results provide a potential therapeutic strategy to promote ischaemic flap survival by administering ABs.


Assuntos
Ferroptose , Fibroblastos , Proteína 1 Associada a ECH Semelhante a Kelch , MicroRNAs , Fator 2 Relacionado a NF-E2 , Retalhos Cirúrgicos , Animais , Camundongos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Ferroptose/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fibroblastos/metabolismo , Modelos Animais de Doenças , Isquemia/metabolismo , Isquemia/genética , Masculino , Apoptose/genética , Tecido Conjuntivo/metabolismo , Transdução de Sinais/genética
19.
Mol Neurobiol ; 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38888836

RESUMO

Lysosomes play a crucial role in various intracellular pathways as their final destination. Various stressors, whether mild or severe, can induce lysosomal membrane permeabilization (LMP), resulting in the release of lysosomal enzymes into the cytoplasm. LMP not only plays a pivotal role in various cellular events but also significantly contributes to programmed cell death (PCD). Previous research has demonstrated the participation of LMP in central nervous system (CNS) injuries, including traumatic brain injury (TBI), spinal cord injury (SCI), subarachnoid hemorrhage (SAH), and hypoxic-ischemic encephalopathy (HIE). However, the mechanisms underlying LMP in CNS injuries are poorly understood. The occurrence of LMP leads to the activation of inflammatory pathways, increased levels of oxidative stress, and PCD. Herein, we present a comprehensive overview of the latest findings regarding LMP and highlight its functions in cellular events and PCDs (lysosome-dependent cell death, apoptosis, pyroptosis, ferroptosis, and autophagy). In addition, we consolidate the most recent insights into LMP in CNS injury by summarizing and exploring the latest advances. We also review potential therapeutic strategies that aim to preserve LMP or inhibit the release of enzymes from lysosomes to alleviate the consequences of LMP in CNS injury. A better understanding of the role that LMP plays in CNS injury may facilitate the development of strategic treatment options for CNS injury.

20.
Burns Trauma ; 12: tkae035, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855574

RESUMO

Background: Ensuring the survival of the distal end of a random flap during hypoperfusion (ischaemia) is difficult in clinical practice. Effective prevention of programmed cell death is a potential strategy for inhibiting ischaemic flap necrosis. The activation of stimulator of interferon genes (STING) pathway promotes inflammation and leads to cell death. The epidermal growth factor family member neuregulin-1 (NRG1) reduces cell death by activating the protein kinase B (AKT) signalling pathway. Moreover, AKT signalling negatively regulates STING activity. We aimed to verify the efficacy of NRG1 injection in protecting against flap necrosis. Additionally, we investigated whether NRG1 effectively enhances ischemic flap survival by inhibiting pyroptosis and necroptosis through STING suppression. Methods: A random-pattern skin flap model was generated on the backs of C57BL/6 mice. The skin flap survival area was determined. The blood supply and vascular network of the flap was assessed by laser Doppler blood flow analysis. Cluster of differentiation 34 immunohistochemistry (IHC) and haematoxylin and eosin (H&E) staining of the flap sections revealed microvessels. Transcriptome sequencing analysis revealed the mechanism by which NRG1 promotes the survival of ischaemic flaps. The levels of angiogenesis, oxidative stress, necroptosis, pyroptosis and indicators associated with signalling pathways in flaps were examined by IHC, immunofluorescence and Western blotting. Packaging adeno-associated virus (AAV) was used to activate STING in flaps. Results: NRG1 promoted the survival of ischaemic flaps. An increased subcutaneous vascular network and neovascularization were found in ischaemic flaps after the application of NRG1. Transcriptomic gene ontology enrichment analysis and protein level detection indicated that necroptosis, pyroptosis and STING activity were reduced in the NRG1 group. The phosphorylation of AKT and forkhead box O3a (FOXO3a) were increased after NRG1 treatment. The increased expression of STING in flaps induced by AAV reversed the therapeutic effect of NRG1. The ability of NRG1 to phosphorylate AKT-FOXO3a, inhibit STING and promote flap survival was abolished after the application of the AKT inhibitor MK2206. Conclusions: NRG1 inhibits pyroptosis and necroptosis by activating the AKT-FOXO3a signalling pathway to suppress STING activation and promote ischaemic flap survival.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA