Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Infect Dis ; 227(12): 1417-1427, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36281765

RESUMO

Liver injury is a common complication during infection of Toxoplasma gondii. However, the Toxoplasma effector proteins involved remain unknown. Herein, we identified that T. gondii macrophage migration inhibitory factor (TgMIF) is a critical pathogenic factor of liver injury in acute toxoplasmosis mouse model induced by a less virulent strain, which is widely prevalent in humans. We show that TgMIF is a novel activator of nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3) inflammasome in hepatocytes, resulting in subsequent pyroptosis. Furthermore, T. gondii promotes the TgMIF-dependent infiltration of Ly6Chi proinflammatory macrophages to release cytokines, leading to hepatocyte apoptosis. Although the intense inflammation induced by TgMIF inhibits the proliferation of intracellular parasites, it results in fatal liver damage. In contrast, parasites with TgMIF gene deletion significantly alleviate liver injury and prolong mice survival. The discovery of novel Toxoplasma virulence factor may expedite the development of human toxoplasmosis control strategies.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Toxoplasma , Toxoplasmose , Camundongos , Humanos , Animais , Fatores Inibidores da Migração de Macrófagos/metabolismo , Piroptose , Toxoplasmose/genética , Toxoplasma/genética , Inflamassomos/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(23): E5344-E5352, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784816

RESUMO

The neurotropic parasite Toxoplasma gondii is a globally distributed parasitic protozoan among mammalian hosts, including humans. During the course of infection, the CNS is the most commonly damaged organ among invaded tissues. The polymorphic rhoptry protein 18 (ROP18) is a key serine (Ser)/threonine (Thr) kinase that phosphorylates host proteins to modulate acute virulence. However, the basis of neurotropism and the specific substrates through which ROP18 exerts neuropathogenesis remain unknown. Using mass spectrometry, we performed proteomic analysis of proteins that selectively bind to active ROP18 and identified RTN1-C, an endoplasmic reticulum (ER) protein that is preferentially expressed in the CNS. We demonstrated that ROP18 is associated with the N-terminal portion of RTN1-C and specifically phosphorylates RTN1-C at Ser7/134 and Thr4/8/118. ROP18 phosphorylation of RTN1-C triggers ER stress-mediated apoptosis in neural cells. Remarkably, ROP18 phosphorylation of RTN1-C enhances glucose-regulated protein 78 (GRP78) acetylation by attenuating the activity of histone deacetylase (HDAC), and this event is associated with an increase of neural apoptosis. These results clearly demonstrate that both RTN1-C and HDACs are involved in T. gondii ROP18-mediated pathogenesis of encephalitis during Toxoplasma infection.


Assuntos
Síndrome da Imunodeficiência Adquirida/microbiologia , Encefalite Infecciosa/microbiologia , Proteínas Serina-Treonina Quinases/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/microbiologia , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/metabolismo , Síndrome da Imunodeficiência Adquirida/patologia , Animais , Apoptose/fisiologia , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Feminino , HIV-1/isolamento & purificação , Interações Hospedeiro-Parasita , Encefalite Infecciosa/metabolismo , Encefalite Infecciosa/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Proteínas de Protozoários , Toxoplasma/patogenicidade , Toxoplasmose/genética , Toxoplasmose/metabolismo , Toxoplasmose/patologia
3.
BMC Complement Altern Med ; 19(1): 272, 2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31638956

RESUMO

BACKGROUND: This study aimed to investigate the effect of the Phellinus linteus (Mesima) decoction on podocyte injury in a rat model of focal and segmental glomerulosclerosis (FSGS) and evaluate the potential mechanisms. METHODS: FSGS resembling primary FSGS in humans was established in rats by uninephrectomy and the repeated injection of doxorubicin. The FSGS rats were randomly divided into the model group, low-dose group of P. linteus decoction (PLD-LD), medium-dose group of P. linteus decoction (PLD-MD), and high-dose group of P. linteus decoction (PLD-HD). Blood and urine analysis were performed after 12 weeks and the molecular indicators of renal function and the renal pathological changes were examined. RESULTS: FSGS developed within 12 weeks in the test group and showed progressive proteinuria and segmental glomerular scarring. Urinary protein, serum creatinine, urea nitrogen, triglycerides and cholesterol were significantly reduced following the 12-week intervention with P.linteus decoction, especially in the PLD-LD group. Renal nephrin and podocin were markedly increased. Moreover, the pathological damage in the renal tissue was alleviated by the PLD-LD intervention. CONCLUSION: The P. linteus decoction alleviated the podocyte injury in the FSGS rat model, thus minimizing the progression of glomerular sclerosis and improving renal function.


Assuntos
Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Podócitos/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/fisiopatologia , Masculino , Proteínas de Membrana/metabolismo , Phellinus , Podócitos/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Exp Biol Med (Maywood) ; 249: 10051, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38881848

RESUMO

Podocyte injury or dysfunction can lead to proteinuria and glomerulosclerosis. Zonula occludens 1 (ZO-1) is a tight junction protein which connects slit diaphragm (SD) proteins to the actin cytoskeleton. Previous studies have shown that the expression of ZO-1 is decreased in chronic kidney disease (CKD). Thus, elucidation of the regulation mechanism of ZO-1 has considerable clinical importance. Triptolide (TP) has been reported to exert a strong antiproteinuric effect by inhibiting podocyte epithelial mesenchymal transition (EMT) and inflammatory response. However, the underlying mechanisms are still unclear. We found that TP upregulates ZO-1 expression and increases the fluorescence intensity of ZO-1 in a puromycin aminonucleoside (PAN)-induced podocyte injury model. Permeablity assay showed TP decreases podocyte permeability in PAN-treated podocyte. TP also upregulates the DNA demethylase TET2. Our results showed that treatment with the DNA methyltransferase inhibitors 5-azacytidine (5-AzaC) and RG108 significantly increased ZO-1 expression in PAN-treated podocytes. Methylated DNA immunoprecipitation (MeDIP) and hydroxymethylated DNA immunoprecipitation (hMeDIP) results showed that TP regulates the methylation status of the ZO-1 promoter. Knockdown of TET2 decreased ZO-1 expression and increased methylation of its promoter, resulting in the increase of podocyte permeability. Altogether, these results indicate that TP upregulates the expression of ZO-1 and decreases podocyte permeability through TET2-mediated 5 mC demethylation. These findings suggest that TP may alleviate podocyte permeability through TET2-mediated hydroxymethylation of ZO-1.


Assuntos
Dioxigenases , Diterpenos , Compostos de Epóxi , Fenantrenos , Podócitos , Proteína da Zônula de Oclusão-1 , Podócitos/metabolismo , Podócitos/efeitos dos fármacos , Podócitos/patologia , Proteína da Zônula de Oclusão-1/metabolismo , Fenantrenos/farmacologia , Diterpenos/farmacologia , Compostos de Epóxi/farmacologia , Dioxigenases/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Permeabilidade/efeitos dos fármacos , Humanos , Metilação de DNA/efeitos dos fármacos
5.
Int Immunopharmacol ; 122: 110584, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37454630

RESUMO

Proteinuria is an independent risk factor for the progression of diabetic nephropathy (DN) and an imbalance in podocyte function aggravates proteinuria. Celastrol is the primary active ingredient of T. wilfordii, effective in treating DN renal injury; however, the mechanisms underlying its effect are unclear. We explored how celastrol prevents DN podocyte damage using in vivo and in vitro experiments. We randomly divided 24 male C57BLKS/J mice into three groups: db/m (n = 8), db/db (n = 8), and celastrol groups (db/db + celastrol, 1 mg/kg/d, gavage administration, n = 8). In vivo experiments lasted 12 weeks and intervention lasted ten weeks. Serum samples and kidney tissues were collected for biochemical tests, pathological staining, transmission electron microscopy, fluorescencequantitation polymerase chain reaction, and western blotting analysis. In vitro experiments to elaborate the mechanism of celastrol protection were performed on high glucose (HG)-induced podocyte injury. Celastrol reduced blood glucose levels and renal function index in db/db mice, attenuated renal histomorphological injury and glomerular podocyte foot injuries, and induced significant anti-inflammatory effects. Celastrol upregulated silent information regulator 2 related enzyme 1(SIRT1) expression and downregulated enhancer of zeste homolog (EZH2), inhibiting the wnt/ß-catenin pathway-related molecules, such as wnt1, wnt7a, and ß-catenin. SIRT1 repressed the promoter activity of EZH2, and was co-immunoprecipitated with EZH2 in mouse podocyte cells (MPC5). SIRT1 knockdown aggravated the protective effects of celastrol on MPC5 cells. Celastrol protected podocyte injury via SIRT1/EZH2, which participates in the wnt/ß-catenin pathway. Overall, celastrol-mediated SIRT1 upregulation inhibited the EZH2-related wnt/ß-catenin signaling pathway to attenuate DN and podocyte injury, providing a theoretical basis for celastrol clinical application.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Podócitos , Camundongos , Masculino , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteinúria , Diabetes Mellitus/patologia
6.
Exp Biol Med (Maywood) ; 247(21): 1947-1955, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36046983

RESUMO

Renal injury is an important factor in the development of chronic kidney diseases that pathologically manifested as renal fibrosis and podocyte damage. In the disease state, renal fibroblasts lead to high expression levels of α-smooth muscle actin (α-SMA), while podocytes undergo epithelial-mesenchymal transition, leading to proteinuria. Celastrol, a bioactive compound in the medicinal plant Tripterygium wilfordii, was found to delay the progression of early diabetic nephropathy and attenuate renal fibrosis in mice with unilateral ureteral obstruction. However, its effect on the renal system in 5/6 nephrectomized (Nx) rats remains unknown. The aim of this study was to explore the protective effects of celastrol and its underlying mechanisms in 5/6 Nx rats. We found that 24 h proteinuria and levels of blood urea nitrogen, serum creatinine, triglycerides, serum P, renal index and cholesterol significantly increased (P < 0.05), while that of serum albumin decreased significantly in 5/6 Nx rats. After intervention with celastrol, 24 h proteinuria and levels of blood urea nitrogen, serum creatinine, triglycerides, serum P, renal index, and cholesterol significantly decreased, while that of serum albumin significantly increased. Renal tissue pathological staining and transmission electron microscopy showed that celastrol ameliorated kidney injury and glomerular podocyte foot injury and induced significant anti-inflammatory effects. Quantitative polymerase chain reaction (PCR) and western blotting results revealed that nephrin and NEPH1 expression levels were upregulated, whereas α-SMA and Col4a1 expression levels were downregulated in the celastrol group. Celastrol inhibited the expression of transforming growth factor (TGF)-ß1/Smad3 signaling pathway-related molecules such as TGF-ß1 and P-Smad3. In summary, celastrol contributes to renal protection by inhibiting the epithelial-mesenchymal transdifferentiation and TGF-ß1/Smad3 pathways.


Assuntos
Transição Epitelial-Mesenquimal , Rim , Triterpenos Pentacíclicos , Proteína Smad3 , Fator de Crescimento Transformador beta1 , Animais , Camundongos , Ratos , Colesterol , Creatinina , Fibrose , Rim/efeitos dos fármacos , Rim/patologia , Albumina Sérica , Triglicerídeos , Triterpenos Pentacíclicos/farmacologia , Nefrectomia
7.
Eur J Med Res ; 27(1): 118, 2022 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-35820962

RESUMO

AIM: The aim of this study was to investigate the renoprotective effects of exosomes derived from rat bone marrow mesenchymal stem cells (rBMSCs) in a rat model of 5/6 nephrectomy (Nx)-induced chronic kidney disease (CKD). METHODS: A rat model of 5/6 Nx-induced CKD was established using conventional method. rBMSC-derived exosomes were isolated using ultracentrifugation and characterized. The exosomes were injected into 5/6 Nx rats through the caudal vein. After 12 weeks, 24 h proteinuria, serum creatinine (SCr), and blood urea nitrogen (BUN) levels were evaluated, and renal pathology was analyzed by H&E and Masson staining, and transmission electron microscopy. The expression of klotho was analyzed and the activity of the klotho promoter was evaluated using a luciferase reporter assay. RESULTS: The isolated exosomes showed typical morphological features. Exosomes transplantation reduced 24 h urinary protein excretion, and SCr and BUN levels in 5/6 Nx-induced CKD rats. Furthermore, renal pathology was improved in the exosome-treated 5/6 Nx rats. Mechanistically, the exosomes significantly upregulated the activity of klotho promoter and its expression. CONCLUSIONS: Transplantation of rBMSC-derived exosomes may protect against kidney injury, probably by regulating klotho activity and expression. Our results provide a theoretical basis for the application of rBMSC-derived exosomes in CKD therapy.


Assuntos
Exossomos , Insuficiência Renal Crônica , Animais , Modelos Animais de Doenças , Exossomos/metabolismo , Rim/patologia , Nefrectomia , Ratos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
8.
Am J Transl Res ; 13(3): 1233-1244, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841652

RESUMO

The epithelial-mesenchymal transition (EMT) is usually considered the central mechanism of podocyte injury that eventually leads to proteinuria. We used an in vitro TGF-ß1 induced podocyte EMT model and an in vivo rat focal segmental glomerulosclerosis (FSGS) model to uncover the mechanism underlying the protective effect of triptolide (TP) on podocytes. We found that TP could reverse the podocyte EMT process and upregulate the expression of TET2 in the TGF-ß1-induced podocyte injury model. Bisulfite amplicon sequencing (BSAS) showed TP could alter the methylation status at some specific sites of the medium CpG density region in the promoters of NEPH1 and nephrin, two main markers of the podocyte slit diaphragm. Knockdown of TET2 with shRNA lentivirus (Lv) leads to high methylation of the promoters of NEPH1 and nephrin such that their expression can not return to normal levels, even after treatment with TP. In vivo, we found that TP could protect against podocyte injury in the FSGS rat and increase TET2 expression. These results suggested TET2-mediated DNA demethylation may be partly involved in podocyte injury. We believe these findings can help uncover a novel molecular mechanism of TP in alleviating podocyte-associated glomerular diseases.

9.
Adv Sci (Weinh) ; 7(1): 1901785, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31921559

RESUMO

Generation of hematopoietic stem/progenitor cells (HSPCs) via cell expansion or cell reprogramming has been widely achieved by overexpression of transcription factors. Herein, it is reported that without introducing exogenous genes, mouse fibroblasts can be reprogrammed into hemogenic cells based on lineage tracing analysis, which further develop into hematopoietic cells, by treatment of cocktails of chemical compounds. The chemical cocktails also reprogram differentiated hematopoietic cells back into HSPC-like cells. Most importantly, the chemical cocktails enabling hematopoietic reprogramming robustly promote HSPC proliferation ex vivo. The expanded HSPCs acquire enhanced capacity of hematopoietic reconstruction in vivo. Single-cell sequencing analysis verifies the expansion of HSPCs and the cell reprogramming toward potential generation of HSPCs at the same time by the chemical cocktail treatment. Thus, the proof-of-concept findings not only demonstrate that hematopoietic reprogramming can be achieved by chemical compounds but also provide a promising strategy for acquisition of HSPCs by chemical cocktail-enabled double effects.

10.
Cell Rep ; 24(5): 1355-1362.e3, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30067988

RESUMO

Neural stem cells (NSCs) are valuable for both basic research and clinical application. We previously reported a chemical cocktail that could reprogram somatic cells into neural progenitor cells. However, the reprogramming process is complex, and the underlying mechanism remains largely elusive. Here, we identified a culture condition that greatly promotes the efficiency of NSC generation directly from mouse fibroblasts based on our reported chemical cocktail. Transcriptome and epigenome analyses demonstrated that growth factors including IL-6, FGF5, and LIF were dynamically activated and contributed to the cell fate changes. Treatment of these growth factors together can enable fibroblast to neural progenitor-like cell conversion. Moreover, the reprogramming capacity of both chemical cocktail and growth factors requires nucleoporin Nup210 to activate SoxB1 transcription factors to initiate NSC fate. Altogether, our findings reveal important roles of both extracellular signals and internal factors in direct cellular reprogramming.


Assuntos
Diferenciação Celular , Fator 5 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/citologia , Células-Tronco Neurais/citologia , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Animais , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Fator 5 de Crescimento de Fibroblastos/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Pirazóis/farmacologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Transcriptoma , Ácido Valproico/farmacologia
11.
Protein Cell ; 8(4): 273-283, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28190217

RESUMO

Tissue damage induces cells into reprogramming-like cellular state, which contributes to tissue regeneration. However, whether factors promoting the cell reprogramming favor tissue regeneration remains elusive. Here we identified combination of small chemical compounds including drug cocktails robustly promoting in vitro cell reprogramming. We then administrated the drug cocktails to mice with acute liver injuries induced by partial hepatectomy or toxic treatment. Our results demonstrated that the drug cocktails which promoted cell reprogramming in vitro improved liver regeneration and hepatic function in vivo after acute injuries. The underlying mechanism could be that expression of pluripotent genes activated after injury is further upregulated by drug cocktails. Thus our study offers proof-of-concept evidence that cocktail of clinical compounds improving cell reprogramming favors tissue recovery after acute damages, which is an attractive strategy for regenerative purpose.


Assuntos
Técnicas de Reprogramação Celular/métodos , Reprogramação Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
12.
Cell Death Dis ; 8(10): e3108, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29022921

RESUMO

Neural stem cells (NSCs) have a unique role in neural regeneration. Cell therapy based on NSC transplantation is a promising tool for the treatment of nervous system diseases. However, there are still many issues and controversies associated with the derivation and therapeutic application of these cells. In this review, we summarize the different sources of NSCs and their derivation methods, including direct isolation from primary tissues, differentiation from pluripotent stem cells and transdifferentiation from somatic cells. We also review the current progress in NSC implantation for the treatment of various neural defects and injuries in animal models and clinical trials. Finally, we discuss potential optimization strategies for NSC derivation and propose urgent challenges to the clinical translation of NSC-based therapies in the near future.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Doenças do Sistema Nervoso/terapia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante , Traumatismos da Medula Espinal/terapia , Animais , Transdiferenciação Celular , Humanos , Regeneração Nervosa/fisiologia , Neurônios/citologia
13.
Cell Death Dis ; 8(10): e3080, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28981095

RESUMO

The reticulon family has been found to induce apoptosis, inhibit axon regeneration and regulate protein trafficking. However, little is known about the mechanisms of how reticulon proteins are involved in neuronal death-promoting processes during ischemia. Here, we report that the expression of Reticulon Protein 1-C (RTN1-C) was associated with the progression of cerebral ischemia/reperfusion (I/R) injury. Using a combination of rat middle cerebral artery occlusion (MCAO) stroke and oxygen-glucose deprivation followed by reoxygenation (OGD/R) models, we determined that the expression of RTN1-C was significantly increased during cerebral ischemic/reperfusion. RTN1-C overexpression induced apoptosis and increased the cell vulnerability to ischemic injury, whereas RTN1-C knockdown reversed ischemia-induced apoptosis and attenuated the vulnerability of OGD/R-treated neural cells. Mechanistically, we demonstrated that RTN1-C mediated OGD/R-induced apoptosis through ER stress and mitochondria-associated pathways. RTN1-C interacted with Bcl-xL and increased its localization in the ER, thus reducing the anti-apoptotic activity of Bcl-xL. Most importantly, knockdown of Rtn1-c expression in vivo attenuated apoptosis in MCAO rats and reduced the extent of I/R-induced brain injury, as assessed by infarct volume and neurological score. Collectively, these data support for the first time that RTN1-C may represent a novel candidate for therapies against cerebral ischemia/reperfusion injury.


Assuntos
Isquemia Encefálica/genética , Proteínas do Tecido Nervoso/genética , Traumatismo por Reperfusão/genética , Acidente Vascular Cerebral/genética , Animais , Apoptose/genética , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Proteínas do Tecido Nervoso/metabolismo , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia
14.
Acta Trop ; 174: 106-113, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28669563

RESUMO

T. gondii is an obligate intracellular parasite, belonging to the Phylum Apicomplexa, infecting all warm-blooded animals including humans. During host cell invasion, specialized cytoskeletal and secretory organelles play a pivotal role. ROP18, as a member of the ROP2 family, has been identified as a key virulence factor mediating pathogenesis in T. gondii. Here, we identify an ER-resident protein, Derlin2, a factor implicated in the removal of misfolded proteins from the ER for cytosolic degradation, as a component of the machinery required for ER-associated protein degradation (ERAD). We identified Derlin2 interacting with ROP18 by yeast two-hybrid screening system. The interaction between ROP18 and Derlin2 was further confirmed through in vitro GST pull-down and in vivo immunoprecipitation assays. By immunofluorescence assay, we found that ROP18 co-localized with Derlin2 in the endoplasmic reticulum. Using overexpression and knockdown approaches, we demonstrated that Derlin2 was required for T. gondii ROP18 degradation. Consistently, cycloheximide chase experiments showed that the degradation of ROP18 relied on the Derlin2, but not Derlin1. These results indicate that interaction between Derlin2 and ROP18 is functionally relevant and leads ultimately to degradation of ROP18. The finding provides the basis for future studies on Derlin2-dependent ERAD of T. gondii ROP18 and subsequent antigen generation.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Proteólise , Toxoplasma/metabolismo , Toxoplasma/patogenicidade , Fatores de Virulência/metabolismo , Animais , Humanos , Toxoplasma/genética
15.
Parasit Vectors ; 8: 554, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26489755

RESUMO

BACKGROUND: The neurotropic parasite T. gondii is widespread among mammalian hosts including humans. During the course of T. gondii infection, the central nervous system is the most commonly damaged of all invasive organs. The polymorphic rhoptry protein ROP18 has been identified as a key factor in the pathogenesis of T. gondii; however, the molecular mechanism by which this protein exerts neuropathogenesis remains elusive. METHODS: Immunofluorescence staining was performed to detect neuropathogenesis of the mouse brain tissues. The apoptosis of neural cells and the expressions of related proteins in the endoplasmic reticulum stress (ER Stress)-mediated apoptosis pathway were detected by flow cytometry and Western blotting. RESULTS: Immunofluorescence staining reveals induction of the propidium iodide (PI) - positive neural cells in mouse cerebral cortex and hippocampus infected with ROP18 over-expressing transgenic tachyzoites. Western blotting analyses reveal that ROP18 increases the expressions of cleaved caspase-12, CHOP and cleaved caspase-3 when compared to the control groups. After the pretreatment of Z-ATAD-FMK (a specific caspase-12 inhibitor), the apoptotic level of neural cells had an apparent decline, and correspondingly, the expressions of those related proteins were notably decreased. CONCLUSIONS: Our findings here highlight that the virulence factor ROP18 in T. gondii may contribute to neuronal apoptosis through the ER stress-mediated apoptosis pathway, which may be a potential molecular mechanism responsible for neurological disorders of toxoplasmosis.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Interações Hospedeiro-Patógeno , Neurônios/fisiologia , Neurônios/parasitologia , Proteínas Serina-Treonina Quinases/metabolismo , Toxoplasma/fisiologia , Animais , Western Blotting , Encéfalo/patologia , Citometria de Fluxo , Camundongos , Proteínas de Protozoários , Toxoplasmose Animal/parasitologia , Toxoplasmose Animal/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA