Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
J Biol Chem ; 299(6): 104689, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37044216

RESUMO

The basal chordate amphioxus is a model for tracing the origin and evolution of vertebrate immunity. To explore the evolution of immunoreceptor signaling pathways, we searched the associated receptors of the amphioxus Branchiostoma belcheri (Bb) homolog of immunoreceptor signaling adaptor protein Grb2. Mass-spectrum analysis of BbGrb2 immunoprecipitates from B. belcheri intestine lysates revealed a folate receptor (FR) domain- and leucine-rich repeat (LRR)-containing protein (FrLRR). Sequence and structural analysis showed that FrLRR is a membrane protein with a predicted curved solenoid structure. The N-terminal Fr domain contains very few folate-binding sites; the following LRR region is a Slit2-type LRR, and a GPI-anchored site was predicted at the C-terminus. RT-PCR analysis showed FrLRR is a transcription-mediated fusion gene of BbFR-like and BbSlit2-N-like genes. Genomic DNA structure analysis implied the B. belcheri FrLRR gene locus and the corresponding locus in Branchiostoma floridae might be generated by exon shuffling of a Slit2-N-like gene into an FR gene. RT-qPCR, immunostaining, and immunoblot results showed that FrLRR was primarily distributed in B. belcheri intestinal tissue. We further demonstrated that FrLRR localized to the cell membrane and lysosomes. Functionally, FrLRR mediated and promoted bacteria-binding and phagocytosis, and FrLRR antibody blocking or Grb2 knockdown inhibited FrLRR-mediated phagocytosis. Interestingly, we found that human Slit2-N (hSlit2-N) also mediated direct bacteria-binding and phagocytosis which was inhibited by Slit2-N antibody blocking or Grb2 knockdown. Together, these results indicate FrLRR and hSlit2-N may function as phagocytotic-receptors to promote phagocytosis through Grb2, implying the Slit2-N-type-LRR-containing proteins play a role in bacterial binding and elimination.


Assuntos
Anfioxos , Animais , Humanos , Anfioxos/genética , Leucina , Sítios de Ligação , Transdução de Sinais , Fagocitose , Filogenia
2.
Mol Cancer ; 23(1): 198, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39272149

RESUMO

Tumor cells remodel the phenotype and function of tumor microenvironment (TME) cells to favor tumor progression. Previous studies have shown that neutrophils in TME are polarized to N2 tumor-associated neutrophils (TANs) by tumor derived factors, thus promoting tumor growth and metastasis, angiogenesis, therapy resistance, and immunosuppression. Exosomes act as critical intercellular messengers in human health and diseases including cancer. So far, the biological roles of exosomes from N2 TANs in gastric cancer have not been well characterized. Herein, we represented the first report that exosomes from N2 TANs promoted gastric cancer metastasis in vitro and in vivo. We found that exosomes from N2 TANs transferred miR-4745-5p/3911 to gastric cancer cells to downregulate SLIT2 (slit guidance ligand 2) gene expression. Adenovirus-mediated overexpression of SLIT2 reversed the promotion of gastric cancer metastasis by N2 TANs derived exosomes. We further revealed that gastric cancer cells induced glucose metabolic reprogramming in neutrophils through exosomal HMGB1 (high mobility group protein B1)/NF-κB pathway, which mediated neutrophil N2 polarization and miR-4745-5p/3911 upregulation. We further employed ddPCR (droplet digital PCR) to detect the expression of miR-4745-5p/3911 in N2 TANs exosomes from human serum samples and found their increased levels in gastric cancer patients compared to healthy controls and benign gastric disease patients. Conclusively, our results indicate that N2 TANs facilitate cancer metastasis via regulation of SLIT2 in gastric cancer cells by exosomal miR-4745-5p/3911, which provides a new insight into the roles of TME cells derived exosomes in gastric cancer metastasis and offers a potential biomarker for gastric cancer diagnosis.


Assuntos
Exossomos , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , MicroRNAs , Proteínas do Tecido Nervoso , Neutrófilos , Neoplasias Gástricas , Microambiente Tumoral , Neoplasias Gástricas/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Exossomos/metabolismo , Exossomos/genética , Humanos , Neutrófilos/metabolismo , Neutrófilos/patologia , MicroRNAs/genética , Animais , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Linhagem Celular Tumoral , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Microambiente Tumoral/genética , Metástase Neoplásica , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Masculino
3.
Mol Cell Probes ; 73: 101947, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38122948

RESUMO

Airway fibrosis is among the pathological manifestations of benign central airway obstruction noted in the absence of effective treatments and requires new drug targets to be developed. Slit guidance ligand 2-roundabout guidance receptor 1 (Slit2-Robo1) is involved in fibrosis and organ development. However, its significance in airway fibrosis has not yet been reported. The study explored how the recombinant protein Slit2 functions in transforming growth factor-ß1 (TGF-ß1)-mediated airway fibrosis in vivo and in vitro. In this study, Slit2 expression initially increased in the tracheal granulation tissues of patients with tracheobronchial stenosis but decreased in the fibrotic tissue. In primary rat tracheal fibroblasts (RTFs), recombinant Slit2 inhibited the expression of extracellular matrices such as Timp1, α-SMA, and COL1A2, whereas recombinant TGF-ß1 promoted the expression of Robo1, α-SMA, and COL1A2. Slit2 and TGF-ß1 played a mutual inhibitory role in RTFs. Slit2 supplementation and Robo1 downregulation inhibited excessive extracellular matrix (ECM) deposition induced by TGF-ß1 in RTFs via the TGF-ß1/Smad3 pathway. Ultimately, exogenous Slit2 and Robo1 knockdown-mediated attenuation of airway fibrosis were validated in a trauma-induced rat airway obstruction model. These findings demonstrate that recombinant Slit2 alleviated pathologic tracheobronchial healing by attenuating excessive ECM deposition. Slit2-Robo1 is an attractive target for further exploring the mechanisms and treatment of benign central airway obstruction.


Assuntos
Obstrução das Vias Respiratórias , Fibrose Pulmonar , Animais , Humanos , Ratos , Obstrução das Vias Respiratórias/metabolismo , Fibroblastos/metabolismo , Fibrose , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fibrose Pulmonar/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/farmacologia
4.
J Biochem Mol Toxicol ; 38(4): e23638, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38613466

RESUMO

The pancreas is a heterocrine gland that has both exocrine and endocrine parts. Most pancreatic cancer begins in the cells that line the ducts of the pancreas and is called pancreatic ductal adenocarcinoma (PDAC). PDAC is the most encountered pancreatic cancer type. One of the most important characteristic features of PDAC is neuropathy which is primarily due to perineural invasion (PNI). PNI develops tumor microenvironment which includes overexpression of fibroblasts cells, macrophages, as well as angiogenesis which can be responsible for neuropathy pain. In tumor microenvironment inactive fibroblasts are converted into an active form that is cancer-associated fibroblasts (CAFs). Neurotrophins they also increase the level of Substance P, calcitonin gene-related peptide which is also involved in pain. Matrix metalloproteases are the zinc-associated proteases enzymes which activates proinflammatory interleukin-1ß into its activated form and are responsible for release and activation of Substance P which is responsible for neuropathic pain by transmitting pain signal via dorsal root ganglion. All the molecules and their role in being responsible for neuropathic pain are described below.


Assuntos
Neuralgia , Neoplasias Pancreáticas , Humanos , Substância P , Neuralgia/etiologia , Pâncreas , Neoplasias Pancreáticas/complicações , Fibroblastos , Microambiente Tumoral
5.
J Infect Dis ; 2023 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-37158474

RESUMO

During infection, Mycobacterium tuberculosis (Mtb) rewires distinct host signaling pathways that results in pathogen-favorable outcomes. Oxidative stress build-up is a key cellular manifestation that occurs due to the cumulative effect of elevated reactive oxygen species generation (ROS) and the inept ability of the cell to mitigate ROS levels. Here, we report the Mtb-induced expression of the neuronal ligand, SLIT2, to be instrumental in ROS accumulation during infection. Loss of function analysis revealed the heightened expression of SLIT2 to be dependent on the Mtb-mediated phosphorylation of the P38/JNK pathways. Activation of these kinases resulted in the loss of the repressive H3K27me3 signature on the Slit2 promoter. Furthermore, SLIT2 promoted the expression of Vanin1 (VNN1), that contributed to copious levels of ROS within the host. Thus, we dissect the pathway leading to the robust expression of SLIT2 during Mtb infection while outlining the potential consequences of SLIT2 upregulation in infected macrophages.

6.
J Neuroinflammation ; 20(1): 249, 2023 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-37899442

RESUMO

BACKGROUND: Germinal matrix hemorrhage (GMH) is a devastating neonatal stroke, in which neuroinflammation is a critical pathological contributor. Slit2, a secreted extracellular matrix protein, plays a repulsive role in axon guidance and leukocyte chemotaxis via the roundabout1 (Robo1) receptor. This study aimed to explore effects of recombinant Slit2 on neuroinflammation and the underlying mechanism in a rat model of GMH. METHODS: GMH was induced by stereotactically infusing 0.3 U of bacterial collagenase into the germinal matrix of 7-day-old Sprague Dawley rats. Recombinant Slit2 or its vehicle was administered intranasally at 1 h after GMH and daily for 3 consecutive days. A decoy receptor recombinant Robo1 was co-administered with recombinant Slit2 after GMH. Slit2 siRNA, srGAP1 siRNA or the scrambled sequences were administered intracerebroventricularly 24 h before GMH. Neurobehavior, brain water content, Western blotting, immunofluorescence staining and Cdc42 activity assays were performed. RESULTS: The endogenous brain Slit2 and Robo1 expressions were increased after GMH. Robo1 was expressed on neuron, astrocytes and infiltrated peripheral immune cells in the brain. Endogenous Slit2 knockdown by Slit2 siRNA exacerbated brain edema and neurological deficits following GMH. Recombinant Slit2 (rSlit2) reduced neurological deficits, proinflammatory cytokines, intercellular adhesion molecules, peripheral immune cell markers, neuronal apoptosis and Cdc42 activity in the brain tissue after GMH. The anti-neuroinflammation effects were reversed by recombinant Robo1 co-administration or srGAP1 siRNA. CONCLUSIONS: Recombinant Slit2 reduced neuroinflammation and neuron apoptosis after GMH. Its anti-neuroinflammation effects by suppressing onCdc42-mediated brain peripheral immune cells infiltration was at least in part via Robo1-srGAP1 pathway. These results imply that recombinant Slit2 may have potentials as a therapeutic option for neonatal brain injuries.


Assuntos
Proteínas do Tecido Nervoso , Transdução de Sinais , Ratos , Animais , Ratos Sprague-Dawley , Proteínas do Tecido Nervoso/metabolismo , Doenças Neuroinflamatórias , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Encéfalo/metabolismo , Hemorragia Cerebral , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , RNA Interferente Pequeno/farmacologia , Proteínas Ativadoras de GTPase/metabolismo
7.
FASEB J ; 36(11): e22618, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36250924

RESUMO

Triple-negative breast cancer (TNBC) is a group of fatal malignancies characterized by high metastatic capacity, the underlying mechanisms of which remain largely elusive. We have found here that insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is highly expressed in TNBC and correlates clinically with distant metastasis-free survival of TNBC patients. IGF2BP3 promotes the migration and invasion capabilities of TNBC cells dependent upon cellular RNA N6-methyladenosine (m6A) modification. Mechanistically, IGF2BP3 binds to and destabilizes m6A-methylated mRNA of the extracellular matrix glycoprotein, SLIT2, impairs its downstream signaling via the cognate receptor ROBO1, and consequently triggers the activation of canonical PI3K/AKT and MEK/ERK pathways. The IGF2BP3/SLIT2 axis is critically involved in the regulation of TNBC metastasis in vivo. These findings shed light into the regulatory network of distant metastasis of breast cancer and provide rationale for targeting the m6A machinery in the treatment of TNBC.


Assuntos
Proteínas de Ligação a RNA , Somatomedinas , Neoplasias de Mama Triplo Negativas , Humanos , Glicoproteínas , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas do Tecido Nervoso/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Receptores Imunológicos/genética , RNA , RNA Mensageiro/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas de Ligação a RNA/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética
8.
Transfus Apher Sci ; 62(1): 103500, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35853810

RESUMO

BACKGROUND: Transfusion-related acute lung injury (TRALI) is the infusion of blood or blood system. OBJECTIVE: To explore the mechanism of TLR4-mediated T cell immune effect in TRALI. METHODS: In this animal study, a mouse model of LPS-induced TRALI was established. Sixty adult C57/BL6 mice (wild-type, WT) were randomly divided into 5 groups: 1) normal WT type, 2) LPS control group of WT type lipopolysaccharide, 3) WT type TRALI group (LPS + MHC-I mAb), 4) (TLR4 antibody) lipopolysaccharide LPS control group, 5) (TLR4 antibody) TRALI group (LPS + MHC-I mAb). Mice were injected with LPS (0.1 mg/kg) and MHC-I mAb (2 mg/kg) into the tail vein. H&E staining was performed to detect pathological features. The myeloperoxidase (MPO) activity and the level of inflammatory cytokines in lung tissue homogenate supernatant were measured. Blood, spleen single-cell suspension, and bronchoalveolar lavage fluid were collected to detect the ratio of Treg and Th17 cells by flow cytometry. RT-PCR and WB were used to detect mRNA or protein expression. RESULTS: TLR4 mAb treatment alleviated the pathogenesis of LPS-induced TRALI in vivo, the MPO activity, and the level of proinflammatory factors in lung tissues. TLR4 exerted its function by changing of Treg/Th17 ratio via the SLIT2/ROBO4 signaling pathway and downregulating CDH5 and SETSIP. CONCLUSION: TLR4 mediates immune response in the LPS-induced TRALI model through the SLIT2/ROBO4 signaling pathway.


Assuntos
Lesão Pulmonar Aguda , Lesão Pulmonar Aguda Relacionada à Transfusão , Camundongos , Animais , Lipopolissacarídeos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Transdução de Sinais , Receptores de Superfície Celular/metabolismo
9.
Int J Mol Sci ; 24(21)2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-37958523

RESUMO

METTL3, a methyltransferase responsible for N6-methyladenosine (m6A) modification, plays key regulatory roles in mammal central neural system (CNS) development. However, the specific epigenetic mechanisms governing human CNS development remain poorly elucidated. Here, we generated small-molecule-assisted shut-off (SMASh)-tagged hESC lines to reduce METTL3 protein levels, and found that METTL3 is not required for human neural progenitor cell (hNPC) formation and neuron differentiation. However, METTL3 deficiency inhibited hNPC proliferation by reducing SLIT2 expression. Mechanistic studies revealed that METTL3 degradation in hNPCs significantly decreased the enrichment of m6A in SLIT2 mRNA, consequently reducing its expression. Our findings reveal a novel functional target (SLIT2) for METTL3 in hNPCs and contribute to a better understanding of m6A-dependent mechanisms in hNPC proliferation.


Assuntos
Metiltransferases , Células-Tronco Neurais , Humanos , Proliferação de Células/genética , Metiltransferases/genética , Metiltransferases/metabolismo , Células-Tronco Neurais/citologia
10.
J Periodontal Res ; 57(3): 578-586, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35426130

RESUMO

BACKGROUND AND OBJECTIVES: Periodontitis is a chronic multifactorial inflammatory disease associated with dental plaque biofilms. Slit guidance ligand 2 (SLIT2) has been shown to guide neuronal migration, regulate the inflammatory response and cancer progression. However, the role of SLIT2 in periodontitis is poorly understood. In this study, we investigated the expression of SLIT2 in the gingiva of periodontitis and its role in periodontitis progression. METHODS: Gingiva and gingival crevicular fluid (GCF) were collected from healthy people and periodontitis patients. Immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) were used to analyze SLIT2 secretion level. Healthy human gingival fibroblasts (hGFs) were isolated and the expression of SLIT2 in lipopolysaccharide (LPS)-treated hGFs was detected. The effect of SLIT2 on inflammation was analyzed using western blot and immunofluorescence. SLIT2 knockdown (KD) and overexpression assays in hGFs were performed to investigate the role of SLIT2 in the LPS-induced inflammatory response. RESULTS: Gingival tissues and GCF of periodontitis patients displayed higher expression of SLIT2. Similarly, SLIT2 was upregulated in hGFs in an inflammatory environment (LPS treatment). In addition, SLIT2 treatment increased the expression of the inflammatory mediators interleukin-6 (IL-6) and IL-8 in hGFs. Mechanistically, SLIT2 stimulated the activation of nuclear factor-κB (NF-κB) signaling, as well as LPS. Lastly, SLIT2 KD impaired LPS-induced IL-6 production in hGFs, while SLIT2 overexpression amplified the inflammatory response. CONCLUSION: SLIT2 may be involved in the aggravation of periodontitis by activating NF-κB signaling in hGFs.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , NF-kappa B , Proteínas do Tecido Nervoso/metabolismo , Periodontite , Células Cultivadas , Fibroblastos , Gengiva/metabolismo , Humanos , Interleucina-6/metabolismo , Ligantes , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Periodontite/metabolismo , Transdução de Sinais
11.
J Am Soc Nephrol ; 32(9): 2255-2272, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34341180

RESUMO

BACKGROUND: Kidney function requires continuous blood filtration by glomerular capillaries. Disruption of glomerular vascular development or maintenance contributes to the pathogenesis of kidney diseases, but the signaling events regulating renal endothelium development remain incompletely understood. Here, we discovered a novel role of Slit2-Robo signaling in glomerular vascularization. Slit2 is a secreted polypeptide that binds to transmembrane Robo receptors and regulates axon guidance as well as ureteric bud branching and angiogenesis. METHODS: We performed Slit2-alkaline phosphatase binding to kidney cryosections from mice with or without tamoxifen-inducible Slit2 or Robo1 and -2 deletions, and we characterized the phenotypes using immunohistochemistry, electron microscopy, and functional intravenous dye perfusion analysis. RESULTS: Only the glomerular endothelium, but no other renal endothelial compartment, responded to Slit2 in the developing kidney vasculature. Induced Slit2 gene deletion or Slit2 ligand trap at birth affected nephrogenesis and inhibited vascularization of developing glomeruli by reducing endothelial proliferation and migration, leading to defective cortical glomerular perfusion and abnormal podocyte differentiation. Global and endothelial-specific Robo deletion showed that both endothelial and epithelial Robo receptors contributed to glomerular vascularization. CONCLUSIONS: Our study provides new insights into the signaling pathways involved in glomerular vascular development and identifies Slit2 as a potential tool to enhance glomerular angiogenesis.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/genética , Glomérulos Renais/irrigação sanguínea , Néfrons/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Animais , Animais Recém-Nascidos , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Camundongos , Néfrons/patologia , Transdução de Sinais , Proteínas Roundabout
12.
Br J Haematol ; 193(2): 375-379, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33481259

RESUMO

SLIT2 constitutes a known tumour suppressor gene, which has not yet been implicated in the pathogenesis of primary central nervous system lymphoma (PCNSL). Performing exome sequencing on paired blood and tumour DNA samples from six treatment-naïve PCNSL patients, we identified novel SLIT2 variants (p.N63S, p.T590M, p.T732S) that were associated with shorter progression-free survival in our cohort and shorter overall survival in a large validation cohort of lymphoid malignancies from the cBio Cancer Genomics Portal. WNT- and NF-κB-reporter luciferase assays suggest detected alterations are loss-of-function variants. Given the possible prognostic implications, the role of SLIT2 in PCNSL pathogenesis and progression warrants further investigation.


Assuntos
Neoplasias do Sistema Nervoso Central/genética , Sequenciamento do Exoma/métodos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfoma não Hodgkin/genética , Proteínas do Tecido Nervoso/genética , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Central/virologia , Estudos de Coortes , Feminino , Variação Estrutural do Genoma/genética , Genômica/métodos , Herpesvirus Humano 4/genética , Humanos , Linfoma não Hodgkin/diagnóstico , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/mortalidade , Masculino , NF-kappa B/genética , Prognóstico , Intervalo Livre de Progressão , Estudos Retrospectivos
13.
Development ; 145(19)2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30177526

RESUMO

Trio, a member of the Dbl family of guanine nucleotide exchange factors, activates Rac1 downstream of netrin 1/DCC signalling in axon outgrowth and guidance. Although it has been proposed that Trio also activates RhoA, the putative upstream factors remain unknown. Here, we show that Slit2 induces Trio-dependent RhoA activation, revealing a crosstalk between Slit and Trio/RhoA signalling. Consistently, we found that RhoA activity is hindered in vivo in Trio mutant mouse embryos. We next studied the development of the ventral telencephalon and thalamocortical axons, which have been previously shown to be controlled by Slit2. Remarkably, this analysis revealed that Trio knockout (KO) mice show phenotypes that bear strong similarities to the ones that have been reported in Slit2 KO mice in both guidepost corridor cells and thalamocortical axon pathfinding in the ventral telencephalon. Taken together, our results show that Trio induces RhoA activation downstream of Slit2, and support a functional role in ensuring the proper positioning of both guidepost cells and a major axonal tract. Our study indicates a novel role for Trio in Slit2 signalling and forebrain wiring, highlighting its role in multiple guidance pathways as well as in biological functions of importance for a factor involved in human brain disorders.


Assuntos
Padronização Corporal , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Telencéfalo/embriologia , Telencéfalo/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Orientação de Axônios , Axônios/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cones de Crescimento/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos Knockout , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tálamo/embriologia , Tálamo/metabolismo
14.
Biochem Biophys Res Commun ; 522(3): 757-762, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31791578

RESUMO

Loss of E-cadherin elicits epithelial-mesenchymal transition (EMT). While both the Src family of membrane-associated non-receptor tyrosine kinases (SFKs) and Slit2 binding to Roundabout 1 (Robo1) have been shown to induce E-cadherin repression and EMT, whether these two signaling pathways are mechanistically coupled remains unknown in epithelial cells. Here we found that Slit2 and Robo1 overexpression activated Src kinases for tyrosine phosphorylation, degradation of E-cadherin and induction of EMT. Specific blockade of Slit2 binding to Robo1 inactivated Src, prevented E-cadherin phosphorylation and EMT induction. Biochemically, the cytoplasmic CC3 motif of Robo1 (CC3) bound directly to the SH2 and 3 domains of c-Src and the cytoplasmic domains of E-cadherin. Slit2 induced Robo1 association with endogenous c-Src and E-cadherin, whereas ectopic expression of CC3 dissociated this protein complex in colorectal epithelial cells. These results indicate that Slit2 not only induces Robo1 binding to Src, but also recruits Src to E-cadherin for tyrosine phosphorylation of E-cadherin, leading to E-cadherin degradation and EMT induction in colorectal epithelial cells.


Assuntos
Caderinas/metabolismo , Transição Epitelial-Mesenquimal , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Quinases da Família src/metabolismo , Proteína Tirosina Quinase CSK/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Humanos , Fosforilação , Mapas de Interação de Proteínas , Proteínas Roundabout
15.
Biochem Biophys Res Commun ; 523(4): 931-938, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-31964527

RESUMO

BACKGROUND: Brain damage in premature infants often occurs in very low birth weight infants (VLBW) as a result of hypoxia-ischemia and can lead to cognitive impairment and movement disorders. Many miRNAs have been demonstrated to participate in hypoxia-ischemic brain damage (HIBD). This study was designed to investigate the roles of miR-200b-3p in brain damage of neonatal rats induced by hypoxia-ischemia. METHODS AND RESULTS: Three-day-old SD rats were used to establish the model of hypoxia-ischemic brain injury mimicking premature infants. RT-qPCR showed that miR-200b-3p was up-regulated in rat brains at the early stage following hypoxia-ischemic treatment. Bioinformatics analysis identified that Slit2 is a target gene of miR-200b-3p and luciferase reporter gene assay confirmed that miR-200b-3p can interact with and target Slit2 mRNA. Inhibition of miR-200b-3p by antagomir increased Slit2 expression at both the mRNA and protein levels in rat brains. TUNEL assay and transmission electron microscopy (TEM) analysis showed decreased numbers of apoptotic neurons in the hypoxia-ischemia-treated animals as a result of administration of miR-200b-3p antagomir. Administration of miR-200b-3p antagomir attenuated spatial and learning memory loss in the animals induced by hypoxia-ischemia as compared to controls. CONCLUSION: Our study has demonstrated that Slit2 is a target gene of miR-200b-3p and that the hypoxia-ischemic brain damage in neonatal rats was alleviated by inhibiting miR-200b-3p via Slit2. miR-200b-3p may be a potential therapeutic target of HIBD for further investigation.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , MicroRNAs/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Antagomirs/farmacologia , Sequência de Bases , Encéfalo/fisiopatologia , Encéfalo/ultraestrutura , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Memória/efeitos dos fármacos , MicroRNAs/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Ratos Sprague-Dawley , Aprendizagem Espacial/efeitos dos fármacos , Regulação para Cima/genética
16.
J Transl Med ; 18(1): 313, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32795291

RESUMO

BACKGROUND: Lung cancer is one of the most common carcinomas in the world, and lung adenocarcinoma (LUAD) is the most lethal and most common subtype of lung cancer. Cigarette smoking is the most leading risk factor of lung cancer, but it is still unclear how normal lung cells become cancerous in cigarette smokers. This study aims to identify potential smoking-related biomarkers associated with the progression and prognosis of LUAD, as well as their regulation mechanism using an in vitro carcinogenesis model and bioinformatics analysis. RESULTS: Based on the integration analysis of four Gene Expression Omnibus (GEO) datasets and our mRNA sequencing analysis, 2 up-regulated and 11 down-regulated genes were identified in both S30 cells and LUAD. By analyzing the LUAD dataset in The Cancer Gene Analysis (TCGA) database, 3 of the 13 genes, viz., glycophorin C (GYPC), NME/NM23 nucleoside diphosphate kinase 1 (NME1) and slit guidance ligand 2 (SLIT2), were found to be significantly correlated with LUAD patients' smoking history. The expression levels of GYPC, NME1 and SLIT2 in S30 cells and lung cancer cell lines were validated by quantitative PCR, immunofluorescence, and western blot assays. Besides, these three genes are associated with tumor invasion depth, and elevated expression of NME1 was correlated with lymph node metastasis. The enrichment analysis suggested that these genes were highly correlated to tumorigenesis and metastasis-related biological processes and pathways. Moreover, the increased expression levels of GYPC and SLIT2, as well as decreased expression of NME1 were associated with a favorable prognosis in LUAD patients. Furthermore, based on the multi-omics data in the TCGA database, these genes were found to be regulated by DNA methylation. CONCLUSION: In conclusion, our observations indicated that the differential expression of GYPC, NME1 and SLIT2 may be regulated by DNA methylation, and they are associated with cigarette smoke-induced LUAD, as well as serve as prognostic factors in LUAD patients.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/genética , Carcinogênese/genética , Biologia Computacional , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Fumar/efeitos adversos
17.
J Surg Res ; 256: 345-354, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32738556

RESUMO

BACKGROUND: The dysfunction of vascular endothelial cells is associated with sepsis development. Long noncoding RNAs take part in the regulation of vascular endothelial cell function. This study aimed to explore the role and mechanism of long noncoding RNA taurine-upregulated gene 1 (TUG1) in lipopolysaccharide (LPS)-induced endothelial cell injury. METHODS: LPS-treated human umbilical vein endothelial cells (HUVECs) were used as a model of sepsis in vitro. Quantitative real-time polymerase chain reaction was performed to detect the expression of TUG1, microRNA-27a-3p (miR-27a-3p) and slit guidance ligand 2 (SLIT2) messenger RNA. Western blot was conducted to measure the protein levels of SLIT2 as well as those involved in apoptosis, autophagy, and inflammatory response. Flow cytometry was used to detect cell apoptotic rate. The targets of TUG1 and miR-27a-3p were predicted via starBase (http://starbase.sysu.edu.cn/index.php). Dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays were carried out to validate the target correlation between miR-27a-3p and TUG1/SLIT2. RESULTS: TUG1 expression was decreased after the treatment of LPS in HUVECs. Overexpression of TUG1 decreased LPS-induced apoptosis, autophagy, and inflammatory response. TUG1 was a sponge of miR-27a-3p. Upregulation of miR-27a-3p reversed the suppressive effect of TUG1 overexpression on LPS-induced apoptosis, autophagy, and inflammatory response. SLIT2 was a target of miR-27a-3p. Knockdown of miR-27a-3p could inhibit LPS-induced injury by increasing SLIT2 in HUVECs. TUG1 could enhance SLIT2 expression by competitively sponging miR-27a-3p. CONCLUSIONS: TUG1 could repress cell apoptosis, autophagy, and inflammatory response in LPS-treated HUVECs by sponging miR-27a-3p to target SLIT2, providing a potential target for the treatment of sepsis.


Assuntos
Endotélio Vascular/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/genética , RNA Longo não Codificante/metabolismo , Sepse/imunologia , Apoptose/genética , Apoptose/imunologia , Autofagia/genética , Autofagia/imunologia , Endotélio Vascular/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipopolissacarídeos/imunologia , Sepse/patologia , Regulação para Cima/imunologia
18.
Exp Cell Res ; 385(1): 111626, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31545977

RESUMO

Liver Cholestasis is a widespread disease of broad etiologies and ultimately results in fibrosis, which is still lacking effective therapeutic strategies. Activation of hepatic stellate cells (HSCs) is the key event of liver fibrosis. Here, we aimed to investigate the effect and mechanism of the Slit2 signaling in cholestasis-induced liver fibrosis. Our findings revealed that the serum levels and hepatic expression of Slit2 were significantly increased in patients with primary biliary cirrhosis (PBC). Additionally, Slit2-Tg mice were much more vulnerable to BDL-induced liver injury and fibrosis compared to WT control. Slit2 up-regulation by Slit2 recombinant protein induced proliferation, and inhibited apoptosis of human HSCs cell line LX-2 via p38 and ERK signaling pathway, resulting in the activation of HSCs. In contrast, Slit2 down-regulation by siRNA silencing inhibit the activation of HSCs. In conclusion, Slit2 is involved in the activation of HSCs and liver fibrogenesis, highlighting Slit2 as a potential therapeutic target for liver fibrosis.


Assuntos
Colestase/metabolismo , Células Estreladas do Fígado/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Células Cultivadas , Regulação para Baixo/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
19.
J Biol Chem ; 293(9): 3039-3055, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29317497

RESUMO

Slit guidance ligand 2 (SLIT2) is a large, secreted protein that binds roundabout (ROBO) receptors on multiple cell types, including neurons and kidney podocytes. SLIT2-ROBO-mediated signaling regulates neuronal migration and ureteric bud (UB) outgrowth during kidney development as well as glomerular filtration in adult kidneys. Additionally, SLIT2 binds Gremlin, an antagonist of bone morphogenetic proteins (BMPs), and BMP-Gremlin signaling also regulates UB formation. However, direct cross-talk between the ROBO2-SLIT2 and BMP-Gremlin signaling pathways has not been established. Here, we report the discovery of negative feedback between the SLIT2 and BMP-Gremlin signaling pathways. We found that the SLIT2-Gremlin interaction inhibited both SLIT2-ROBO2 signaling in neurons and Gremlin antagonism of BMP activity in myoblasts and fibroblasts. Furthermore, BMP2 down-regulated SLIT2 expression and promoter activity through canonical BMP signaling. Gremlin treatment, BMP receptor inhibition, and SMAD family member 4 (SMAD4) knockdown rescued BMP-mediated repression of SLIT2. BMP2 treatment of nephron progenitor cells derived from human embryonic stem cells decreased SLIT2 expression, further suggesting an interaction between the BMP2-Gremlin and SLIT2 pathways in human kidney cells. In conclusion, our study has revealed direct negative cross-talk between two pathways, previously thought to be unassociated, that may regulate both kidney development and adult tissue maintenance.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Proteína Morfogenética Óssea 2/farmacologia , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Domínios Proteicos , Transdução de Sinais/efeitos dos fármacos
20.
Biochem Biophys Res Commun ; 514(3): 868-874, 2019 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-31084928

RESUMO

Axon guidance molecules, originally found to mediate the positioning of axons during nerve development, have been receiving great attention due to their critical roles in regulating bone metabolism. Recently, SLITs, another group of neuronal guidance proteins, were found to be significantly expressed in bone cells. Furthermore, we had provided experimental evidence that SLIT3 is an osteoclast-secreted coupling factor playing an osteoprotective role. Therefore, we hypothesized that SLIT2, a member of the SLIT family, may also affect bone homeostasis. SLIT2 suppressed osteoclast differentiation in a dose-dependent manner and in vitro bone resorption by more than 80%. Consistently, the expression of osteoclast differentiation markers, such as tartrate-resistant acid phosphatase (Trap) and calcitonin receptor (Ctr), was decreased by SLIT2. The migration and fusion of preosteoclasts were markedly reduced in the presence of SLIT2, suggesting that SLIT2 mainly functions in the early stage of osteoclastogenesis. SLIT2 suppressed Cdc42 activity among small GTPases, whereas Cdc42 overexpression almost completely reversed the SLIT2-mediated suppression of osteoclast differentiation. Among ROBO1-4, the SLIT receptors, ROBO1 and ROBO3 were known to be predominantly expressed in osteoclast lineages. A binding ELISA experiment in mouse bone marrow-derived macrophages showed that ROBO1, rather than ROBO3, was directly associated with SLIT2, and gene silencing with Robo1 siRNA blocked the SLIT2-mediated suppression of osteoclastogenesis. Taken together, our results indicated that SLIT2 inhibits osteoclastogenesis and the resultant bone resorption by decreasing Cdc42 activity, suggesting that this was a potential therapeutic target in metabolic bone diseases related to high bone turnover states.


Assuntos
Reabsorção Óssea/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas do Tecido Nervoso/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Proteína cdc42 de Ligação ao GTP/genética , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Diferenciação Celular , Proliferação de Células , Fêmur/citologia , Fêmur/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/citologia , Osteoclastos/patologia , Cultura Primária de Células , Células RAW 264.7 , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores da Calcitonina/genética , Receptores da Calcitonina/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais , Fosfatase Ácida Resistente a Tartarato/genética , Fosfatase Ácida Resistente a Tartarato/metabolismo , Tíbia/citologia , Tíbia/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Roundabout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA