Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
FASEB J ; 34(6): 8702-8720, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32385864

RESUMO

Disruption of colonic homeostasis caused by aberrant M1/M2 macrophage polarization and dysbiosis contributes to inflammatory bowel disease (IBD) pathogenesis. However, the molecular factors mediating colonic homeostasis are not well characterized. Here, we found that Ninjurin1 (Ninj1) limits colon inflammation by regulating macrophage polarization and microbiota composition under homeostatic conditions and during colitis development. Ninj1 deletion in mice induced hypersusceptibility to colitis, with increased prevalence of colitogenic Prevotellaceae strains and decreased immunoregulatory Lachnospiraceae strains. Upon co-housing (CoH) with WT mice, Ninj1-/- mice showed increased Lachnospiraceae and decreased Prevotellaceae abundance, with subsequent improvement of colitis. Under homeostatic conditions, M1 macrophage frequency was higher in the Ninj1-/- mouse colons than wild-type (WT) mouse colons, which may contribute to increased basal colonic inflammation and microbial imbalance. Following colitis induction, Ninj1 expression was increased in macrophages; meanwhile Ninj1-/- mice showed severe colitis development and impaired recovery, associated with decreased M2 macrophages and escalated microbial imbalance. In vitro, Ninj1 knockdown in mouse and human macrophages activated M1 polarization and restricted M2 polarization. Finally, the transfer of WT macrophages ameliorated severe colitis in Ninj1-/- mice. These findings suggest that Ninj1 mediates colonic homeostasis by modulating M1/M2 macrophage balance and preventing extensive dysbiosis, with implications for IBD prevention and therapy.


Assuntos
Moléculas de Adesão Celular Neuronais/deficiência , Colite/metabolismo , Colite/patologia , Microbioma Gastrointestinal/fisiologia , Macrófagos/metabolismo , Macrófagos/patologia , Fatores de Crescimento Neural/deficiência , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Homeostase/fisiologia , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Ativação de Macrófagos/fisiologia , Masculino , Camundongos , Fatores de Crescimento Neural/metabolismo , Células THP-1/metabolismo
2.
Korean J Women Health Nurs ; 26(1): 72-83, 2020 Mar 31.
Artigo em Coreano | MEDLINE | ID: mdl-36311850

RESUMO

Purpose: The purpose of this study was to investigate the effects of uncertainty and spousal support on infertility-related quality of life (QoL) in women undergoing assisted reproductive technologies. Methods: In this correlational survey study, 172 infertile women undergoing assisted reproductive technologies for infertility treatment at M hospital in Seoul participated. Data collection took place at the outpatient department of M hospital using a self-report questionnaire from July to August 2019. Data were analyzed using SPSS for Windows version 28.0. Results: The mean scores for uncertainty, spousal support, and infertility-related quality of life (QoL) were 28.35 (out of 50), 86.67 (out of 115), and 57.98 (out of 100), respectively. Infertility-related quality of life (QoL) was positively correlated with spousal support and negatively correlated with uncertainty. According to the regression analysis, infertility-related quality of life (QoL) was significantly affected by uncertainty, total number of assisted reproductive technology treatments, marriage duration, subjective health status, the financial burden of infertility testing, and the presence of a burdensome person. These variables had an explanatory power of 35.0% for infertility-related quality of life (QoL). Conclusion: Uncertainty was an important factor influencing infertility-related quality of life (QoL) among women undergoing assisted reproductive technologies. It is necessary to develop and implement a nursing intervention program focused on reducing various forms of uncertainty during assisted reproductive procedures and to consider other factors affecting infertility-related quality of life (QoL) in the clinical setting.

3.
Exp Mol Med ; 51(1): 1-16, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30700695

RESUMO

Osteoclasts (OCs) are bone-resorbing cells that originate from hematopoietic stem cells and develop through the fusion of mononuclear myeloid precursors. Dysregulation of OC development causes bone disorders such as osteopetrosis, osteoporosis, and rheumatoid arthritis. Although the molecular mechanisms underlying osteoclastogenesis have been well established, the means by which OCs maintain their survival during OC development remain unknown. We found that Ninjurin1 (Ninj1) expression is dynamically regulated during osteoclastogenesis and that Ninj1-/- mice exhibit increased trabecular bone volume owing to impaired OC development. Ninj1 deficiency did not alter OC differentiation, transmigration, fusion, or actin ring formation but increased Caspase-9-dependent intrinsic apoptosis in prefusion OCs (preOCs). Overexpression of Ninj1 enhanced the survival of mouse macrophage/preOC RAW264.7 cells in osteoclastogenic culture, suggesting that Ninj1 is important for the survival of preOCs. Finally, analysis of publicly available microarray data sets revealed a potent correlation between high NINJ1 expression and destructive bone disorders in humans. Our data indicate that Ninj1 plays an important role in bone homeostasis by enhancing the survival of preOCs.


Assuntos
Osso Esponjoso/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Fatores de Crescimento Neural/genética , Osteoclastos/metabolismo , Osteogênese , Animais , Apoptose , Osso Esponjoso/crescimento & desenvolvimento , Moléculas de Adesão Celular Neuronais/metabolismo , Células Cultivadas , Humanos , Masculino , Camundongos , Fatores de Crescimento Neural/metabolismo , Osteoclastos/citologia , Células RAW 264.7
4.
Oncol Lett ; 16(5): 5907-5915, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30344741

RESUMO

AKAP12 belongs to A-kinase anchoring protein (AKAP) family of scaffold proteins and is known as a tumor suppressor in several human cancer types. Its role as a tumor suppressor in hepatocellular carcinoma (HCC) was proposed due to its downregulation and epigenetic modification in human HCC; however, the effect of its deficiency on liver injuries, such as liver fibrosis and cancer has been poorly studied. By analyzing tumor and non-tumor tissues of 15 patients with HCC, it was confirmed that AKAP12 expression was downregulated in human HCC as compared with adjacent non-tumor tissues. Immunohistochemical staining of mouse liver tissue for AKAP12 revealed that its sinusoidal expression was diminished in capillarized endothelium after 8 weeks of thioacetamide (TAA) administration. AKAP12 deficiency resulted in the promotion of ductular response of biliary epithelial cells, whereas overall fibrosis and myofibroblast activation were comparable between genotypes after short-term TAA treatment. The mRNA expressions of some fibrosis-related genes such as those encoding epithelial cell adhesion molecule, collagen type 1 α1 and elastin were upregulated in liver tissues of AKAP12-knockout mice. Long-term administration of TAA for 26 weeks led to the development of liver tumors; the incidence of tumor development was higher in AKAP12-deficient mice than in wild-type littermates. Together, these results suggest that AKAP12 functions as a tumor suppressor in liver cancer and is associated with the regulation of hepatic non-parenchymal cells.

5.
Exp Mol Med ; 50(4): 1-13, 2018 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-29700280

RESUMO

Liver fibrosis can be reversed by removing its causative injuries; however, the molecular mechanisms mediating the resolution of liver fibrogenesis are poorly understood. We investigate the role of a scaffold protein, A-Kinase Anchoring Protein 12 (AKAP12), during liver fibrosis onset, and resolution. Biliary fibrogenesis and fibrosis resolution was induced in wild-type (WT) or AKAP12-deficient C57BL/6 mice through different feeding regimens with 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing chow. AKAP12 expression in portal fibroblasts (PFs) and liver sinusoidal endothelial cells (LSECs) gradually decreased as fibrosis progressed but was restored after cessation of the fibrotic challenge. Histological analysis of human liver specimens with varying degrees of fibrosis of different etiologies revealed that AKAP12 expression diminishes in hepatic fibrosis from its early stages onward. AKAP12 KO mice displayed reduced fibrosis resolution in a DDC-induced biliary fibrosis model, which was accompanied by impaired normalization of myofibroblasts and capillarized sinusoids. RNA sequencing of the liver transcriptome revealed that genes related to ECM accumulation and vascular remodeling were mostly elevated in AKAP12 KO samples. Gene ontology (GO) and bioinformatic pathway analyses identified that the differentially expressed genes were significantly enriched in GO categories and pathways, such as the adenosine 3',5'-cyclic monophosphate (cAMP) pathway. Knockdown of the AKAP12 gene in cultured primary PFs revealed that AKAP12 inhibited PF activation in association with the adenosine 3',5'-cyclic monophosphate (cAMP) pathway. Moreover, AKAP12 knockdown in LSECs led to enhanced angiogenesis, endothelin-1 expression and alterations in laminin composition. Collectively, this study demonstrates that AKAP12-mediated regulation of PFs and LSECs has a central role in resolving hepatic fibrosis.


Assuntos
Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ciclo Celular/genética , Fibroblastos/patologia , Regulação da Expressão Gênica , Cirrose Hepática/genética , Fígado/patologia , Proteínas de Ancoragem à Quinase A/análise , Animais , Proteínas de Ciclo Celular/análise , Linhagem Celular , Fibroblastos/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcriptoma
6.
Oncotarget ; 8(40): 68517-68529, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28978134

RESUMO

SAM domain and HD domain containing protein 1 (SAMHD1) is a deoxynucleotide triphosphohydrolase (dNTPase) that inhibits retroviruses by depleting intracellular deoxynucleotide triphosphates (dNTPs) in non-cycling myeloid cells. Although SAMHD1 is expressed ubiquitously throughout the human body, the molecular mechanisms regulating its enzymatic activity and function in non-immune cells are relatively unexplored. Here, we demonstrate that the dNTPase activity of SAMHD1 is regulated by acetylation, which promotes cell cycle progression in cancer cells. SAMHD1 is acetylated at residue lysine 405 (K405) in vitro and in vivo by an acetylatransferase, arrest defective protein 1 (ARD1). Acetylated SAMHD1 wildtype proteins have enhanced dNTPase activity in vitro, whereas non-acetylated arginine substituted mutants (K405R) do not. K405R mutant expressing cancer cells have reduced G1/S transition and slower proliferation compared to wildtype. SAMHD1 acetylation levels are strongest during the G1 phase, indicating a role during G1 phase. Collectively, these findings suggest that SAMHD1 acetylation enhances its dNTPase activity and promotes cancer cell proliferation. Therefore, SAMHD1 acetylation may be a potent therapeutic target for cancer treatment.

7.
Oncotarget ; 8(34): 57216-57230, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28915666

RESUMO

Aurora kinase A (AuA) is a prerequisite for centrosome maturation, separation, and mitotic spindle assembly, thus, it is essential for cell cycle regulation. Overexpression of AuA is implicated in poor prognosis of many types of cancer. However, the regulatory mechanisms underlying the functions of AuA are still not fully understood. Here, we report that AuA colocalizes with arrest defective protein 1 (ARD1) acetyltransferase during cell division and cell migration. Additionally, AuA is acetylated by ARD1 at lysine residues at positions 75 and 125. The double mutations at K75/K125 abolished the kinase activity of AuA. Moreover, the double mutant AuA exhibited diminished ability to promote cell proliferation and cell migration. Mechanistic studies revealed that AuA acetylation at K75/K125 promoted cell proliferation via activation of cyclin E/CDK2 and cyclin B1. In addition, AuA acetylation stimulated cell migration by activating the p38/AKT/MMP-2 pathway. Our findings indicate that ARD1-mediated acetylation of AuA enhances cell proliferation and migration, and probably contributes to cancer development.

8.
PLoS One ; 12(8): e0182047, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28771527

RESUMO

Left-right asymmetric organ development is critical to establish a proper body plan of vertebrates. In zebrafish, the Kupffer's vesicle (KV) is a fluid-filled sac which controls asymmetric organ development, and a properly inflated KV lumen by means of fluid influx is a prerequisite for the asymmetric signal transmission. However, little is known about the components that support the paracellular tightness between the KV luminal epithelial cells to sustain hydrostatic pressure during KV lumen expansion. Here, we identified that the claudin5a (cldn5a) is highly expressed at the apical surface of KV epithelial cells and tightly seals the KV lumen. Downregulation of cldn5a in zebrafish showed a failure in organ laterality that resulted from malformed KV. In addition, accelerated fluid influx into KV by combined treatment of forskolin and 3-isobutyl-1-methylxanthine failed to expand the partially-formed KV lumen in cldn5a morphants. However, malformed KV lumen and defective heart laterality in cldn5a morphants were significantly rescued by exogenous cldn5a mRNA, suggesting that the tightness between the luminal epithelial cells is important for KV lumen formation. Taken together, these findings suggest that cldn5a is required for KV lumen inflation and left-right asymmetric organ development.


Assuntos
Padronização Corporal/fisiologia , Cílios/fisiologia , Claudina-5/metabolismo , Embrião não Mamífero/citologia , Lateralidade Funcional/fisiologia , Morfogênese/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/crescimento & desenvolvimento , Animais , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peixe-Zebra/metabolismo
9.
Int J Oncol ; 51(2): 573-578, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627586

RESUMO

Cancer cells are continuously challenged by adverse environmental factors including hypoxia, metabolite restriction, and immune reactions, and must adopt diverse strategies to survive. Heat shock protein (Hsp) 70 plays a central role in protection against stress-induced cell death by maintaining protein homeostasis and interfering with the process of programmed cell death. Recent findings have suggested that Hsp70 acetylation is a key regulatory modification required for its chaperone activity, but its relevance in the process of programmed cell death and the underlying mechanisms involved are not well understood. In this study, we sought to investigate mechanisms mediated by Hsp70 acetylation in relation to apoptotic and autophagic programmed cell death. Upon stress-induced apoptosis, Hsp70 acetylation inhibits apoptotic cell death, mediated by Hsp70 association with apoptotic protease-activating factor (Apaf)-1 and apoptosis-inducing factor (AIF), key modulators of caspase-dependent and -independent apoptotic pathways, respectively. Hsp70 acetylation also attenuated autophagic cell death associated with upregulation of autophagy-related genes and autophagosome induction. Collectively, these results suggest that the acetylation of Hsp70 plays key regulatory roles in cell death pathways as well as in its function as a chaperone, together enabling cellular protection in response to stress.


Assuntos
Fator de Indução de Apoptose/genética , Fator Apoptótico 1 Ativador de Proteases/genética , Proteínas de Choque Térmico HSP70/genética , Neoplasias/genética , Acetilação , Apoptose/genética , Autofagossomos/metabolismo , Autofagia/genética , Caspases , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Dobramento de Proteína , Transdução de Sinais/genética , Estresse Fisiológico/genética
10.
Sci Rep ; 7: 42723, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28205544

RESUMO

Macrophages exhibit phenotypic plasticity, as they have the ability to switch their functional phenotypes during inflammation and recovery. Simultaneously, the mechanical environment actively changes. However, how these dynamic alterations affect the macrophage phenotype is unknown. Here, we observed that the extracellular matrix (ECM) constructed by AKAP12+ colon mesenchymal cells (CMCs) generated M2 macrophages by regulating their shape during recovery. Notably, rounded macrophages were present in the linear and loose ECM of inflamed colons and polarized to the M1 phenotype. In contrast, ramified macrophages emerged in the contracted ECM of recovering colons and mainly expressed M2 macrophage markers. These contracted structures were not observed in the inflamed colons of AKAP12 knockout (KO) mice. Consequently, the proportion of M2 macrophages in inflamed colons was lower in AKAP12 KO mice than in WT mice. In addition, clinical symptoms and histological damage were more severe in AKAP12 KO mice than in WT mice. In experimentally remodeled collagen gels, WT CMCs drove the formation of a more compacted structure than AKAP12 KO CMCs, which promoted the polarization of macrophages toward an M2 phenotype. These results demonstrated that tissue contraction during recovery provides macrophages with the physical cues that drive M2 polarization.


Assuntos
Diferenciação Celular , Matriz Extracelular/metabolismo , Macrófagos/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Colo/citologia , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
11.
Mol Neurobiol ; 54(9): 7353-7368, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-27815839

RESUMO

Over the last few decades, molecular neurobiology has uncovered many genes whose deficiency in mice results in behavioral traits associated with human neuropsychiatric disorders such as autism, obsessive-compulsive disorder (OCD), and schizophrenia. However, the etiology of these common diseases remains enigmatic with the potential involvement of a battery of genes. Here, we report abnormal behavioral phenotypes of mice deficient in a cell adhesion molecule Ninjurin 1 (Ninj1), which are relevant to repetitive and anxiety behaviors of neuropsychiatric disorders. Ninj1 knockout (KO) mice exhibit compulsive grooming-induced hair loss and self-made lesions as well as increased anxiety-like behaviors. Histological analysis reveals that Ninj1 is predominantly expressed in cortico-thalamic circuits, and neuron-specific Ninj1 conditional KO mice manifest aberrant phenotypes similar to the global Ninj1 KO mice. Notably, the brains of Ninj1 KO mice display altered synaptic transmission in thalamic neurons as well as a reduced number of functional synapses. Moreover, the disruption of Ninj1 leads to glutamatergic abnormalities, including increased ionotropic glutamate receptors but reduced glutamate levels. Furthermore, chronic treatment with fluoxetine, a drug reportedly ameliorates compulsive behaviors in mice, prevents progression of hair loss and alleviates the compulsive grooming and anxiety-like behavior of Ninj1 KO mice. Collectively, our results suggest that Ninj1 could be involved in neuropsychiatric disorders associated with impairments of repetitive and anxiety behaviors.


Assuntos
Ansiedade/genética , Ansiedade/metabolismo , Moléculas de Adesão Celular Neuronais/deficiência , Moléculas de Adesão Celular Neuronais/genética , Comportamento Compulsivo/genética , Comportamento Compulsivo/metabolismo , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Animais , Ansiedade/psicologia , Células Cultivadas , Comportamento Compulsivo/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Nat Commun ; 7: 12882, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27708256

RESUMO

Heat shock protein (Hsp)70 is a molecular chaperone that maintains protein homoeostasis during cellular stress through two opposing mechanisms: protein refolding and degradation. However, the mechanisms by which Hsp70 balances these opposing functions under stress conditions remain unknown. Here, we demonstrate that Hsp70 preferentially facilitates protein refolding after stress, gradually switching to protein degradation via a mechanism dependent on ARD1-mediated Hsp70 acetylation. During the early stress response, Hsp70 is immediately acetylated by ARD1 at K77, and the acetylated Hsp70 binds to the co-chaperone Hop to allow protein refolding. Thereafter, Hsp70 is deacetylated and binds to the ubiquitin ligase protein CHIP to complete protein degradation during later stages. This switch is required for the maintenance of protein homoeostasis and ultimately rescues cells from stress-induced cell death in vitro and in vivo. Therefore, ARD1-mediated Hsp70 acetylation is a regulatory mechanism that temporally balances protein refolding/degradation in response to stress.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Acetiltransferase N-Terminal A/metabolismo , Acetiltransferase N-Terminal E/metabolismo , Redobramento de Proteína , Acetilação , Animais , Apoptose , Caspases/metabolismo , Sobrevivência Celular , Proteínas de Fluorescência Verde/química , Células HEK293 , Humanos , Chaperonas Moleculares/química , Mutação , Ligação Proteica , Domínios Proteicos , Processamento de Proteína Pós-Traducional , RNA Interferente Pequeno/metabolismo , Estresse Fisiológico , Peixe-Zebra
13.
Int J Oncol ; 49(4): 1407-14, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27498767

RESUMO

Cirrhosis, the end-stage of hepatic fibrosis, is not only life-threatening by itself, but also a causative factor of liver cancer. Despite efforts to develop treatment for liver fibrosis, there are no approved agents as anti-fibrotic drugs to date. In the present study, we aimed to investigate the anti-fibrotic effect of the AMP-activated protein kinase (AMPK) activator, HL156A. A mouse model of thioacetamide (TAA)-induced liver fibrosis was used to examine the effect of HL156A in vivo. Mice received either TAA alone or a combination of TAA and HL156A intraperitoneally for a total duration of 6 weeks. Including HL156A during exposure to TAA significantly reduced extracellular matrix (ECM) deposition and production of the hepatic transforming growth factor-ß1 (TGF-ß1). Immunohistochemical analysis revealed that the activation of hepatic stellate cells and the capillarization of liver sinusoids were also diminished significantly by HL156A co-treatment. The anti-fibrotic effect of HL156A was further studied in vitro by using a rat hepatic stellate cell line, HSC-T6 cells. The induction of α-smooth muscle actin (α-SMA) by TGF-ß1 treatment was reversed by HL156A, which was likely via the activation of AMPK. Moreover, HL156A showed anti-inflammatory effects on macrophages. Treatment with HL156A diminished LPS-induced activation of both Raw264.7 macrophage cells and primary cultured mouse macrophages. Taken together, these results imply that the AMPK activator HL156A inhibits hepatic fibrosis via multiple mechanisms and could be a potentially effective agent for fibrosis treatment.


Assuntos
Guanidinas/administração & dosagem , Células Estreladas do Fígado/efeitos dos fármacos , Cirrose Hepática/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Pirrolidinas/administração & dosagem , Tioacetamida/efeitos adversos , Actinas/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Guanidinas/farmacologia , Células Estreladas do Fígado/metabolismo , Humanos , Injeções Intraperitoneais , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Macrófagos/metabolismo , Camundongos , Pirrolidinas/farmacologia , Células RAW 264.7 , Ratos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
14.
Int J Oncol ; 48(2): 821-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26677008

RESUMO

Ninjurin1 is a transmembrane protein involved in macrophage migration and adhesion during inflammation. It was recently reported that repression of Ninjurin1 attenuated the lipopolysaccharide (LPS)-induced inflammatory response in macrophages; however, the precise mechanism by which Ninjurin1 modulates LPS-induced inflammation remains poorly understood. In the present study, we found that the interaction between Ninjurin1 and LPS contributed to the LPS-induced inflammatory response. Notably, pull-down assays using lysates from HEK293T cells transfected with human or mouse Ninjurin1 and biotinylated LPS (LPS-biotin) showed that LPS directly bound Ninjurin1. Subsequently, LPS binding assays with various truncated forms of Ninjurin1 protein revealed that amino acids (aa) 81-100 of Ninjurin1 were required for LPS binding. In addition, knockdown experiments using Ninj1 siRNA resulted in decreased nitric oxide (NO) and tumor necrosis factor-α (TNFα) secretion upon LPS treatment in Raw264.7 cells. Collectively, our results suggest that Ninjurin1 regulates the LPS-induced inflammatory response through its direct binding to LPS, thus, identifying Ninjurin1 as a putative target for the treatment of inflammatory diseases, such as sepsis and inflammation-associated carcinogenesis.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Fatores de Crescimento Neural/metabolismo , Aminoácidos/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/fisiologia , Células HEK293 , Humanos , Macrófagos/metabolismo , Camundongos , Óxido Nítrico/metabolismo , Ligação Proteica/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Development ; 142(24): 4363-73, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26586223

RESUMO

Angiogenesis in the developing central nervous system (CNS) is regulated by neuroepithelial cells, although the genes and pathways that couple these cells to blood vessels remain largely uncharacterized. Here, we have used biochemical, cell biological and molecular genetic approaches to demonstrate that ß8 integrin (Itgb8) and neuropilin 1 (Nrp1) cooperatively promote CNS angiogenesis by mediating adhesion and signaling events between neuroepithelial cells and vascular endothelial cells. ß8 integrin in the neuroepithelium promotes the activation of extracellular matrix (ECM)-bound latent transforming growth factor ß (TGFß) ligands and stimulates TGFß receptor signaling in endothelial cells. Nrp1 in endothelial cells suppresses TGFß activation and signaling by forming intercellular protein complexes with ß8 integrin. Cell type-specific ablation of ß8 integrin, Nrp1, or canonical TGFß receptors results in pathological angiogenesis caused by defective neuroepithelial cell-endothelial cell adhesion and imbalances in canonical TGFß signaling. Collectively, these data identify a paracrine signaling pathway that links the neuroepithelium to blood vessels and precisely balances TGFß signaling during cerebral angiogenesis.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Cadeias beta de Integrinas/metabolismo , Neovascularização Fisiológica , Neuropilina-1/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Animais , Encéfalo/patologia , Adesão Celular , Perda do Embrião/patologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Deleção de Genes , Masculino , Camundongos , Modelos Biológicos , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Peixe-Zebra
16.
Mol Med Rep ; 12(2): 1817-23, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25873382

RESUMO

Reactive gliosis is a glial response to a wide range of central nervous system insults, which results in cellular and molecular changes to resting glial cells. Despite its fundamental effect on neuropathologies, the identification and characterization of the molecular mechanisms underlying this process remain to be fully elucidated. The aim of the present study was to analyze the expression profile and functions of the astrocytic neurotrophic factor, meteorin, in the progression of reactive gliosis. A mouse model of photothrombotic ischemia, and a primary astrocyte culture were used in the present study. Reverse transcription quantitative polymerase chain reaction, western blotting and immunofluorescence staining were performed to examine the expression levels of meteorin and reactive gliosis markers. Increased expression levels of meteorin were observed in reactive astrocytes in a photothrombotic ischemia mouse model, as well as in cultured astrocytes, which were stimulated by transforming growth factor-ß1. Exogenous treatment of the astrocytes with meteorin did not induce janus kinase-signal transducer and activator of transcription 3 signaling, however, silencing the expression of meteorin in the astrocytes resulted in an upregulation of reactive astrocyte markers, including glial fibrillary acidic protein and S100ß, indicating that endogenous meteorin is required for the maintenance of astrocytic homeostasis. These results suggested a novel role for meteorin as a negative feedback effector in reactive gliosis.


Assuntos
Astrócitos/metabolismo , Gliose , Proteínas do Tecido Nervoso/metabolismo , Animais , Astrócitos/citologia , Células Cultivadas , Modelos Animais de Doenças , Imuno-Histoquímica , Isquemia/induzido quimicamente , Isquemia/metabolismo , Isquemia/patologia , Luz , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Fármacos Fotossensibilizantes/toxicidade , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/efeitos dos fármacos
17.
Mol Cell Biol ; 35(8): 1401-13, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25666508

RESUMO

Directional cell motility is essential for normal development and physiology, although how motile cells spatiotemporally activate signaling events remains largely unknown. Here, we have characterized an adhesion and signaling unit comprised of protein tyrosine phosphatase (PTP)-PEST and the extracellular matrix (ECM) adhesion receptor ß8 integrin that plays essential roles in directional cell motility. ß8 integrin and PTP-PEST form protein complexes at the leading edge of migrating cells and balance patterns of Rac1 and Cdc42 signaling by controlling the subcellular localization and phosphorylation status of Rho GDP dissociation inhibitor 1 (RhoGDI1). Translocation of Src-phosphorylated RhoGDI1 to the cell's leading edge promotes local activation of Rac1 and Cdc42, whereas dephosphorylation of RhoGDI1 by integrin-bound PTP-PEST promotes RhoGDI1 release from the membrane and sequestration of inactive Rac1/Cdc42 in the cytoplasm. Collectively, these data reveal a finely tuned regulatory mechanism for controlling signaling events at the leading edge of directionally migrating cells.


Assuntos
Movimento Celular , Cadeias beta de Integrinas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 12/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Proteínas F-Box/metabolismo , Proteína 7 com Repetições F-Box-WD , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Masculino , Camundongos , Fosforilação , Ligação Proteica , Mapas de Interação de Proteínas , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
J Biol Chem ; 289(32): 21926-36, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24917672

RESUMO

Ninjurin1 is involved in the pathogenesis of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, by mediating leukocyte extravasation, a process that depends on homotypic binding. However, the precise regulatory mechanisms of Ninjurin1 during inflammation are largely undefined. We therefore examined the pro-migratory function of Ninjurin1 and its regulatory mechanisms in macrophages. Interestingly, Ninjurin1-deficient bone marrow-derived macrophages exhibited reduced membrane protrusion formation and dynamics, resulting in the impairment of cell motility. Furthermore, exogenous Ninjurin1 was distributed at the membrane of filopodial structures in Raw264.7 macrophage cells. In Raw264.7 cells, RNA interference of Ninjurin1 reduced the number of filopodial projections, whereas overexpression of Ninjurin1 facilitated their formation and thus promoted cell motility. Ninjurin1-induced filopodial protrusion formation required the activation of Rac1. In Raw264.7 cells penetrating an MBEC4 endothelial cell monolayer, Ninjurin1 was localized to the membrane of protrusions and promoted their formation, suggesting that Ninjurin1-induced protrusive activity contributed to transendothelial migration. Taking these data together, we conclude that Ninjurin1 enhances macrophage motility and consequent extravasation of immune cells through the regulation of protrusive membrane dynamics. We expect these findings to provide insight into the understanding of immune responses mediated by Ninjurin1.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Movimento Celular/fisiologia , Macrófagos/fisiologia , Fatores de Crescimento Neural/fisiologia , Animais , Adesão Celular/fisiologia , Moléculas de Adesão Celular Neuronais/deficiência , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Membrana Celular/fisiologia , Células Cultivadas , Células Endoteliais/fisiologia , Técnicas de Silenciamento de Genes , Inflamação/etiologia , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Neuropeptídeos/metabolismo , Pseudópodes/fisiologia , Interferência de RNA , Proteínas rac1 de Ligação ao GTP/metabolismo
19.
Methods Mol Biol ; 1135: 261-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24510871

RESUMO

Most organs and tissues of the vertebrate body harbor elaborate network of blood vessels with diverse functions that are determined, in part, by cues within the local environment (Warren and Iruela-Arispe, Curr Opin Hematol 17:213-218, 2010). How vascular endothelial cells decipher these cues to promote normal blood vessel development and physiology remains largely uncharacterized. In this review, we will focus on genetic strategies to analyze glial regulation of blood vessel growth and sprouting within the microenvironment of the retina, a component of the central nervous system (CNS) that contains a complex web of blood vessels with many unique features, including a blood-retinal barrier (Abbott et al., Nat Rev Neurosci 7:41-53, 2006). Blood vessels promote retinal development and homeostasis and alterations in vascular functions can lead to various developmental and adult-onset retinal pathologies (Fruttiger, Angiogenesis 10:77-88, 2007). How glial cells control retinal endothelial cell growth and sprouting remains largely uncharacterized. We will detail methodologies involving inducible Cre-lox technologies to acutely ablate genes of interest in CNS glial cells. These methods allow for precise spatial and temporal regulation of gene expression to study how glial cells in the retinal microenvironment control angiogenesis and blood-retinal barrier development.


Assuntos
Sistema Nervoso Central/irrigação sanguínea , Neovascularização Fisiológica , Neuroglia/fisiologia , Animais , Deleção de Genes , Camundongos , Camundongos Transgênicos , Vasos Retinianos/citologia , Vasos Retinianos/fisiologia , Fixação de Tecidos
20.
J Biol Chem ; 289(6): 3328-38, 2014 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-24347169

RESUMO

Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26-37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


Assuntos
Células da Medula Óssea/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular , Encefalomielite Autoimune Experimental/metabolismo , Macrófagos/metabolismo , Fatores de Crescimento Neural/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Células da Medula Óssea/patologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Suscetibilidade a Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/terapia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Esclerose Múltipla/genética , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Esclerose Múltipla/terapia , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA