Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Pediatr Nephrol ; 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39097532

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a life-threatening thrombotic microangiopathy. Genetic defects in the alternative complement (AP) pathway have been identified in 60-70% of individuals. Eculizumab is recommended as a first-line therapy. METHODS: We collected the clinical data of a pediatric patient with aHUS accompanied by protein-losing enteropathy (PLE). Genetic testing was performed. Related literature on aHUS combined with PLE was reviewed. RESULTS: A 15-year-old Chinese girl was diagnosed with aHUS at 3.7 years of age and experienced five episodes; her symptoms completely resolved with plasma treatment. Severe gastrointestinal symptoms and hypoalbuminemia presented after the first episode, and PLE was diagnosed. A novel homozygous CD46 variant was identified, and FACS revealed significantly decreased CD46 expression. She presented at a recent relapse with persistent GI symptoms and headache and progressed to chronic kidney failure; peritoneal dialysis was initiated. Eculizumab was given 8 months after the last recurrence. Surprisingly, PLE was cured. Afterward, dialysis was discontinued, and eGFR recovered to 44.8 ml/min/1.73 m2. A review of the literature indicated that PLE with thrombosis was caused by CD55 variants via hyperactivation of the AP system. We report an aHUS patient with PLE caused by CD46 variants. Symptoms of both PLE and aHUS were significantly alleviated in our patient and patients with CD55 variants treated with eculizumab, indicating that PLE was a new symptom of aHUS in our patient with a CD46 variant. CONCLUSIONS: Our case expands the phenotype of aHUS caused by a CD46 mutation and provides evidence of the efficacy of eculizumab after a long phase of chronic kidney failure.

2.
Exp Cell Res ; 407(2): 112753, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34499887

RESUMO

Peroxisome proliferator-activated receptor α (PPARα) activation has been reported to exert protective effects on podocytes, whereas angiopoietin-like 3 (ANGPTL3) has been shown to exert significant pathogenic effects on these cells. This study aimed to investigate the link between the protective effects of PPARα activation and the pathogenic effects of ANGPTL3 in podocytes. Both PPARα and ANGPTL3 were expressed in cultured podocytes. PPARα mRNA and protein levels decreased whereas ANGPTL3 mRNA and protein levels increased in a time-dependent manner in podocytes treated with puromycin aminonucleoside (PAN). Gemfibrozil, a pharmacological agonist of PPARα, increased PPARα levels and activity in podocytes. The drug also decreased ANGPTL3 levels by potentially weakening ANGPTL3 promoter activity in both normal and PAN-treated podocytes. Furthermore, gemfibrozil significantly decreased PAN-induced apoptosis and F-actin rearrangement. Primary podocytes from Angptl3-knockout mice were cultured. There was no significant difference between Angptl3-/- podocytes treated with or without gemfibrozil in the lamellipodia numbers after PAN treatment. The results suggested that the protective effects of gemfibrozil on podocytes were not exerted following knockout of the Angptl3 gene. This study identified a novel mechanism of the PPARα agonist gemfibrozil that exerts its protective effects by inhibiting PAN-induced apoptosis and cytoskeleton rearrangements through inhibition of ANGPTL3 expression.


Assuntos
Citoesqueleto de Actina/efeitos dos fármacos , Proteínas Semelhantes a Angiopoietina/fisiologia , Genfibrozila/farmacologia , PPAR alfa/agonistas , Podócitos/efeitos dos fármacos , Pseudópodes/efeitos dos fármacos , Puromicina Aminonucleosídeo/farmacologia , Proteína 3 Semelhante a Angiopoietina , Animais , Apoptose , Hipolipemiantes/farmacologia , Camundongos , Camundongos Knockout , Podócitos/metabolismo , Podócitos/patologia , Fatores de Proteção , Pseudópodes/metabolismo
3.
Biochem Biophys Res Commun ; 516(3): 812-818, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31256934

RESUMO

Proteinuria is an important marker and is closely related to the progressive decline of renal function. Our previous research showed that angiopoietin-like-3 (ANGPTL3) plays a crucial role in proteinuria. In this study, we prepared an antibody against ANGPTL3 coil-coiled domain (ANGPTL3-CCD) and investigated the protective effect of anti-ANGPTL3-CCD antibody in mice with adriamycin-induced nephropathy. Nephropathy was established by adriamycin injection at a dose of 25 mg per kg in 8-12 week-old male mice in the ADR group. Blockade of ANGPTL3 by anti-ANGPTL3-CCD antibody (20 mg per kg) was performed every three days nine times after adriamycin injection in the ADR plus anti-angptl3-antibody group. The anti-ANGPTL3-CCD antibody can specifically recognize ANGPTL3. After anti-ANGPTL3-CCD antibody intervention, the urinary protein level in the ADR plus anti-angptl3-antibody group was significantly lower than that in the ADR group. Serum albumin was higher and triglyceride and total cholesterol were lower in the ADR plus anti-angptl3-antibody group than in the ADR group. The levels of serum creatinine did not significantly differ among the groups. Focal sclerotic glomeruli and podocyte foot processes extensive fusion were found in the renal tissue of the ADR group, whereas no sclerotic glomeruli and only partial fusion were found in the ADR plus anti-angptl3-antibody group. This study demonstrated that the anti-ANGPTL3-CCD antibody ameliorated proteinuria and podocyte dysfunction in adriamycin-induced nephropathy in mice.


Assuntos
Proteínas Semelhantes a Angiopoietina/genética , Anti-Inflamatórios/farmacologia , Anticorpos/farmacologia , Nefrite/tratamento farmacológico , Proteinúria/tratamento farmacológico , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/antagonistas & inibidores , Proteínas Semelhantes a Angiopoietina/metabolismo , Animais , Anti-Inflamatórios/isolamento & purificação , Anticorpos/isolamento & purificação , Especificidade de Anticorpos , Colesterol/sangue , Creatinina/sangue , Doxorrubicina/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Nefrite/induzido quimicamente , Nefrite/genética , Nefrite/patologia , Domínios Proteicos , Proteinúria/induzido quimicamente , Proteinúria/genética , Proteinúria/patologia , Coelhos , Albumina Sérica/antagonistas & inibidores , Albumina Sérica/metabolismo , Resultado do Tratamento , Triglicerídeos/antagonistas & inibidores , Triglicerídeos/sangue
4.
BMC Nephrol ; 20(1): 185, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-31126248

RESUMO

BACKGROUND: Angiopoietin-like-3 (Angptl3) knockout is known for its protective effects on podocyte injury and proteinuria in the early stage of adriamycin (ADR) nephropathy. The current study re-evaluated the renoprotective effect of Angptl3 knockout in chronic ADR nephropathy and attempted to explore the mechanism underlying the effect associated with Angptl3 knockout in glomerulosclerosis. METHODS: B6; 129S5 mice were injected with ADR to induce nephropathy. Kidney structure and serum and urine parameters were observed during long-term follow-up. Cultured primary mouse podocytes were exposed to ADR and analyzed for the expression of some relative proteins. Podocyte loss was analyzed in both in vivo and in vitro experiments. RESULTS: Angptl3 knockout attenuated proteinuria and hypoproteinemia, protected renal structure and function, and improved the survival of mice over the whole process of ADR nephropathy. Furthermore, Angptl3 knockout reduced the numbers of the detached and apoptotic cells in the renal tissue and alleviated podocyte loss in mice with ADR chronic nephropathy, thereby, delaying the glomerulosclerosis formation. Additional results in vitro showed that Angptl3 knockout attenuated ADR-induced primary podocyte loss, including podocyte detachment and apoptosis. CONCLUSION: In addition to serving a renoprotective role in the early stage of ADR nephropathy, Angptl3 knockout contributed to disease amelioration throughout the ADR nephropathy process. Angptl3 knockout effectively delayed glomerulosclerosis formation by attenuating podocyte loss through rescuing podocytes from detachment and apoptosis. Angptl3 antagonists or inhibitors might have therapeutic potential in the occurrence and progression of nephropathy.


Assuntos
Proteínas Semelhantes a Angiopoietina/deficiência , Antibióticos Antineoplásicos/toxicidade , Doxorrubicina/toxicidade , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/metabolismo , Podócitos/metabolismo , Proteína 3 Semelhante a Angiopoietina , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Glomerulosclerose Segmentar e Focal/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Podócitos/patologia , Podócitos/ultraestrutura
5.
Sensors (Basel) ; 16(9)2016 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-27598161

RESUMO

For high-speed aircraft, a conformal window is used to optimize the aerodynamic performance. However, the local shape of the conformal window leads to large amounts of dynamic aberrations varying with look angle. In this paper, deformable mirror (DM) and model-based wavefront sensorless adaptive optics (WSLAO) are used for dynamic aberration correction of an infrared remote sensor equipped with a conformal window and scanning mirror. In model-based WSLAO, aberration is captured using Lukosz mode, and we use the low spatial frequency content of the image spectral density as the metric function. Simulations show that aberrations induced by the conformal window are dominated by some low-order Lukosz modes. To optimize the dynamic correction, we can only correct dominant Lukosz modes and the image size can be minimized to reduce the time required to compute the metric function. In our experiment, a 37-channel DM is used to mimic the dynamic aberration of conformal window with scanning rate of 10 degrees per second. A 52-channel DM is used for correction. For a 128 × 128 image, the mean value of image sharpness during dynamic correction is 1.436 × 10(-5) in optimized correction and is 1.427 × 10(-5) in un-optimized correction. We also demonstrated that model-based WSLAO can achieve convergence two times faster than traditional stochastic parallel gradient descent (SPGD) method.

6.
Exp Ther Med ; 28(2): 302, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38873048

RESUMO

[This retracts the article DOI: 10.3892/etm.2020.8708.].

7.
Sci Rep ; 14(1): 19557, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174626

RESUMO

The lockbolt structure is essential in railway wagons, and a scientific lockbolt layout can ensure uniform load distribution, thereby preventing failure. However, current engineering lacks layout optimization methods that address multidimensional failure modes. This paper presents a new lockbolt structure layout optimization method based on submodel, parametric models, and a multi-strategy integrated NSGA-III (MSNSGA-III), adhering to the DVS EFB 3435-2 standard. This method simultaneously optimizes the number and spacing of lockbolts to prevent tensile, bearing, shear, and other static failure modes under specified load conditions. The proposed method was applied during the design phase of a container flatcar. Optimization results indicate that, compared to NSGA-III, this method achieves the best IGD and HV values across multiple complex test functions, demonstrating superior performance in solving complex Pareto front optimization problems. Additionally, the optimized lockbolt structure's safety margins increased by a maximum of 59.81%, passing the full vehicle strength test and significantly enhancing resistance to multidimensional failure modes. These results highlight the method's significant practical application value in addressing the optimization of railway wagon lockbolt structures under complex multidimensional failure modes.

8.
Gene ; 907: 148286, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38367852

RESUMO

BACKGROUND: Osteosarcoma (OS), with a peak incidence during the adolescent growth spurt, is correlated with poor prognosis for its high malignancy. The tumor microenvironment (TME) is highly complicated, with frequent interactions between tumor and stromal cells. The cancer-associated fibroblasts (CAFs) in the TME have been considered to actively involve in the progression, metastasis, and drug resistance of OS. This study aimed to characterize cellular heterogeneity and molecular characterization in CAFs subtypes and explore the potential targeting therapeutic strategies to improve the prognosis of OS patients. METHODS: The single-cell atlas of human OS tumor lesions were constructed from the GEO database. Then significant marker genes and potential biological functions for each CAFs subtype were identified and explored using the Seurat R package. Next, by performing the survival analyses and constructing the risk scores for CAFs subtypes, we aimed to identify and characterize the prognostic values of specific marker genes and different CAFs subtypes. Furthermore, we explored the therapeutic targets and innovative drugs targeting different CAFs subtypes based on the GDSC database. Finally, prognoses related CAFs subtypes were further validated through immunohistochemistry (IHC) on clinical OS specimens. RESULTS: Overall, nine main cell clusters and five subtypes of CAFs were identified. The differentially expressed marker genes for each CAFs clusters were then identified. Moreover, through Gene Ontology (GO) enrichment analysis, we defined the CAFs_2 (upregulated CXCL14 and C3), which was closely related to leukocyte migration and chemotaxis, as inflammatory CAFs (iCAFs). Likewise, we defined the CAFs_4 (upregulated CD74, HLA-DRA and HLA-DRB1), which was closely related to antigen process and presentation, as antigen-presenting CAFs (apCAFs). Furthermore, Kaplan-Meier analyses showed that CAFs_2 and CAFs_4 were correlated with poor clinical prognosis of OS patients. Meanwhile, therapeutic drugs targeting CAFs_2 and CAFs_4, such as 17-AAG/Docetaxel/Bleomycin and PHA-793887/NG-25/KIN001-102, were also explored, respectively. Finally, IHC assay confirmed the abundant CAFs_2 and CAFs_4 subtypes infiltration in the OS microenvironment compared with adjacent tissues. CONCLUSION: Our study revealed the diversity, complexity, and heterogeneity of CAFs in OS, and complemented the single-cell atlas in OS TME.


Assuntos
Neoplasias Ósseas , Fibroblastos Associados a Câncer , Osteossarcoma , Adolescente , Humanos , Osteossarcoma/genética , Perfilação da Expressão Gênica , Expressão Gênica , Neoplasias Ósseas/genética , Microambiente Tumoral/genética
9.
Int Immunopharmacol ; 115: 109715, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37724955

RESUMO

Diabetic nephropathy (DN) is a common leading cause of end-stage renal disease (ESRD). Podocyte injury is a major pathogenesis of DN. Pharmacological inhibition of the renin-angiotensin-aldosterone system (RAAS) is insufficient to fully prevent the development of ESRD. The present investigation aims to evaluate the protective function of valsartan, an angiotensin receptor blocker, alone and in combination with angiopoietin-like protein 3 (Angptl3) knockout against renal damage and podocyte injury in streptozotocin (STZ)-induced diabetic mice. The mice were divided into four groups: normal control group, STZ-induced DN group, valsartan + DN group (val, 100 mg/kg, po), and Angptl3-/- + valsartan + DN group. Tests on kidney function, renal pathology, podocyte ultrastructure, podocyte apoptosis, reactive oxygen species (ROS) production, and autophagy were performed. The combined Angptl3 knockout/valsartan treatment significantly attenuated diabetes-induced renal pathological damage and improved podocyte ultrastructure compared with valsartan alone. The combined administration ameliorated glomerular injury by increasing nephrin, podocin, and CD2-associated protein (CD2AP) expression levels and inhibiting podocyte loss by apoptosis. Compared with valsartan alone, Angptl3-/- and valsartan combination therapy significantly improved the renal function, as demonstrated by decreasing levels of serum urea nitrogen, creatinine, and urinary albumin. Additionally, the combination treatment significantly activated autophagy and reduced the ROS production than valsartan alone. These findings highlight the role of valsartan to Angptl3 knockout could have much better outcome that opens the future for drugs that could inhibit Angptl3.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Falência Renal Crônica , Podócitos , Animais , Camundongos , Nefropatias Diabéticas/tratamento farmacológico , Valsartana/uso terapêutico , Estreptozocina , Proteína 3 Semelhante a Angiopoietina , Diabetes Mellitus Experimental/tratamento farmacológico , Espécies Reativas de Oxigênio
10.
Mater Today Bio ; 20: 100675, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37304579

RESUMO

In recent years, immune checkpoint blockades (ICBs) have made great progress in the treatment of cancer. However, most ICBs have not yet been observed to be satisfactory in the treatment of osteosarcoma. Herein, we designed composite nanoparticles (NP-Pt-IDOi) from a reactive oxygen species (ROS) sensitive amphiphilic polymer (PHPM) with thiol-ketal bonds in the main chain to encapsulate a Pt(IV) prodrug (Pt(IV)-C12) and an indoleamine-(2/3)-dioxygenase (IDO) inhibitor (IDOi, NLG919). Once NP-Pt-IDOi enter the cancer cells, the polymeric nanoparticles could dissociate due to the intracellular ROS, and release Pt(IV)-C12 and NLG919. Pt(IV)-C12 induces DNA damage and activates the cGAS-STING pathway, increasing infiltration of CD8+ T cells in the tumor microenvironment. In addition, NLG919 inhibits tryptophan metabolism and enhances CD8+ T cell activity, ultimately activating anti-tumor immunity and enhancing the anti-tumor effects of platinum-based drugs. NP-Pt-IDOi were shown to have superior anti-cancer activity in vitro and in vivo in mouse models of osteosarcoma, providing a new clinical paradigm for combining chemotherapy with immunotherapy for osteosarcoma.

11.
Front Oncol ; 13: 1158857, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37361567

RESUMO

Introduction: Tumor progression is driven by intrinsic malignant behaviors caused by gene mutation or epigenetic modulation, as well as crosstalk with the components in the tumor microenvironment (TME). Considering the current understanding of the tumor microenvironment, targeting the immunomodulatory stromal cells such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) could provide a potential therapeutic strategy. Here, we investigated the effect of sulfatinib, a multi-targeted tyrosine kinase inhibitor (TKI) of FGFR1, CSF1R, and VEGFR1-3, on the treatment of osteosarcoma (OS). Methods: In vitro, the antitumor effect was tested by clony formation assay and apoptosis assay.The inhibition of tumor migration and invasion was detected by Transwell assay, and the de-polarization of macrophage was detected by flow cytometry.In vivo, subcutaneous and orthotopic tumor models were established to verify antitumor effect, and the underlying mechanism was verified by immunohistochemistry(IHC), immunofluorescence(IF) and flow cytometry. Results: Sulfatinib suppressed OS cell migration and invasion by inhibiting epithelial-mesenchymal transition (EMT) by blocking the secretion of basic fibroblast growth factor (bFGF) in an autocrine manner. In addition, it regulated immune TME via inhibition of the migration of skeletal stem cells (SSCs) to the TME and the differentiation from SSCs to CAFs. Moreover, sulfatinib can suppress OS by modulation of the TME by inhibiting M2 polarization of macrophages. Systemic treatment of sulfatinib can reduce immunosuppression cells M2-TAMs, Tregs, and myeloid-derived suppressor cells (MDSCs) and increase cytotoxic T-cell infiltration in tumors, the lungs, and the spleens. Discussion: Our preclinical experiments have shown that sulfatinib can inhibit the proliferation, migration, and invasion of OS by playing a dual role on tumor cells and the tumor microenvironment simultaneously and systematically reverse immunosuppression to immune activation status, which could be translated into clinical trials.

12.
Front Pediatr ; 11: 1113484, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37266537

RESUMO

Background: Angiopoietin-like 3 (ANGPTL3) is a secretory glycoprotein. It has been demonstrated that ANGPTL3 level was upregulated in minimal change nephrotic syndrome (MCNS) kidney tissues. Subsequently, our group found that ANGPTL3 level was closely correlated with nephropathy in vivo and in vitro. Hence, whether ANGPTL3 level could be correlated with the proteinuria level, and assessment of disease severity of nephrotic syndrome (NS) remained to be investigated. This study aimed to analyzed the correlation between the levels of ANGPTL3 in serum and urine of patients with nephrotic syndrome and proteinuria, and assessed the severity of the patients' disease. In future clinical translation, the level of ANGPTL3 in serum, urine will be used as a biomarker to better predict the development of nephrotic syndrome. Methods: A total of 200 NS patients and 80 healthy controls (age, 1-18 years) were admitted to our institution between 2021 and 2022. The etiology of NS included primary nephrotic syndrome (PNS, n = 144) and NS with other causes (n = 56). A total of 280 serum samples and 244 urinary samples were collected to determine ANGPTL3 level using enzyme-linked immunosorbent assay (ELISA). Results: Serum ANGPTL3 and urinary ANGPTL3/Cre were remarkably elevated in NS patients compared with those in healthy controls. Furthermore, serum ANGPTL3 and urinary ANGPTL3/Cre were significantly correlated with proteinuria level. Additionally, multivariate linear regression analysis demonstrated that serum ALB was independently correlated with serum ANGPTL3 and PRO/CR was independently correlated with urinary ANGPTL3/Cre in NS patients. Conclusion: Serum ANGPTL3 and urinary ANGPTL3/Cre showed a promising performance in the diagnosis of NS, and served as novel potential noninvasive biomarkers to assess disease severity of NS. Further exploration of the role of ANGPTL3 level may shed a new light on the treatment of NS.

13.
Int Immunopharmacol ; 105: 108549, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35086056

RESUMO

Acute kidney injury (AKI) caused by sepsis has a high incidence and poor prognosis. Thus, novel strategies that minimize AKI are urgently needed. In our previous research, we found that angiopoietin-like protein 3 (Angptl3) knockout exerts protective effects on kidney injury in adriamycin nephropathy. However, the role of Angptl3 in the pathogenesis of AKI is largely unclear. This study aimed to explore the renal protective effects and molecular mechanisms of Angptl3 knockout in lipopolysaccharide (LPS)-induced AKI in mice. B6;129S5 mice were injected intraperitoneally with 10 mg/kg of LPS to induce AKI. Then, the changes in renal function, podocyte apoptosis, inflammatory factors (tumor necrosis factor-α, TNF-α; interleukin-6, IL-6; and interleukin-1ß, IL-1ß), reactive oxygen species (ROS), and endoplasmic reticulum (ER) stress were measured. The mechanism of Angptl3 in the apoptosis of podocytes was also investigated. Results showed that Angptl3 knockout significantly alleviated the renal dysfunction and apoptosis of podocytes induced by LPS. Angptl3 knockout was associated with the (1) downregulation of Bax and upregulation of Bcl-2; (2) amelioration of the abnormal expression of nephrin, podocin, and CD2AP; (3) reduced ER stress; (4) reduced secretions of TNF-α, IL-6, and IL-1ß; and (5) regulation of Bax expression via the ROS-related ER stress pathway. Our findings revealed that Angptl3 knockout alleviated the apoptosis of podocytes by regulating the ROS/GRP78 signaling pathway.


Assuntos
Injúria Renal Aguda , Podócitos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/genética , Proteínas Semelhantes a Angiopoietina/metabolismo , Animais , Apoptose , Chaperona BiP do Retículo Endoplasmático , Lipopolissacarídeos/farmacologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo
14.
Cell Death Dis ; 13(10): 867, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229446

RESUMO

Proteinuria, an indication of kidney disease, is caused by the malfunction of podocytes, which play a key role in maintaining glomerular filtration. Angiopoietin-like 3 (ANGPTL3) has been documented to have a cell-autonomous involvement in podocytes, and deletion of Angptl3 in podocytes reduced proteinuria in adriamycin-induced nephropathy. Here, we developed a monoclonal antibody (mAb) against ANGPTL3 to investigate its effects on podocyte injury in an ADR nephropathy mouse model and puromycin (PAN) induced podocyte damage in vitro. The mAb against the human ANGPTL3-FLD sequence (5E5F6) inhibited the binding of ANGPTL3-FLD to integrin ß3. Treatment with the 5E5F6 mAb in ADR nephropathy mice mitigated proteinuria and led to a significant decline in podocyte apoptosis, reactive oxygen species (ROS) generation and mitochondrial fragmentation. In PAN-induced podocyte damage in vitro, the 5E5F6 mAb blocked the ANPGPLT3-mediated activation of integrin αvß3 and Rac1, which regulated the mitochondrial homeostasis. Altogether, anti-ANGPLT3-FLD mAb attenuates proteinuria and podocyte lesions in ADR mice models, as well as PAN-induced podocyte damage, in part through regulating mitochondrial functions. Our study provides a therapeutic approach for targeting ANGPTL3 in proteinuric kidney disease.


Assuntos
Nefropatias , Podócitos , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/metabolismo , Angiopoietinas/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Doxorrubicina/farmacologia , Humanos , Integrina alfaVbeta3/metabolismo , Integrina beta3/metabolismo , Nefropatias/patologia , Camundongos , Podócitos/metabolismo , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Puromicina/farmacologia , Espécies Reativas de Oxigênio/metabolismo
15.
Ann Palliat Med ; 10(5): 5218-5230, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33977746

RESUMO

BACKGROUND: Rheumatoid arthritis is a long-term systemic disease that primarily affects multiple synovial joints throughout the body. Some patients with severe joint effusion even require repeated arthrocentesis or arthroscopic debridement to relieve symptoms, which causes them much suffering mentally and physically. This text-mining study was designed to find potential drugs that target key genes in this disease. METHODS: Firstly, we performed text mining by two keywords ("rheumatoid synovitis" and "joint effusion") to get a common set of genes. Secondly, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis performed on these genes, and protein-protein interaction (PPI) network was constructed. Subsequently, the significant genes clustered in the PPI network were chose to execute gene-drug interaction analysis for potential drug discovery. RESULTS: Through text mining, 68 overlapping genes were identified as an initial set of key genes. Construction of the initial gene set's PPI network showed that 25 genes clustered in a significant gene module. Ultimately, 8 out of 25 genes could be targetable by a total of 19 drugs. CONCLUSIONS: The final 8 genes (PTGS2, TNF, VEGFA, IL1B, CCL2, VWF, IL6, and ESR1) and 19 drugs may provide significant therapeutic value for rheumatoid arthritis patients with joint effusion.


Assuntos
Artrite Reumatoide , Descoberta de Drogas , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/genética , Biologia Computacional , Mineração de Dados , Perfilação da Expressão Gênica , Humanos
16.
Exp Ther Med ; 20(1): 359-366, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32537002

RESUMO

The aim of the present study was to investigate the potential therapeutic effects of molecular hydrogen on type 2 diabetes mellitus (T2DM) in rats. Following maintenance on a high-fat diet for 4 weeks, a T2DM model was established using an injection of 30 mg/kg streptozotocin via the caudal vein into Sprague-Dawley rats. On day 0 and Day 80, the blood samples were obtained from each rat for the measurement of biochemical indicators including blood lipids, fasting blood glucose, hepatic glycogen, fasting serum insulin, insulin sensitivity index, insulin resistance index, serum superoxide dismutase (SOD) and serum malondialdehyde (MDA) using an automatic biochemical analyzer. The kidneys and pancreas tissues were harvested for HE staining and Western blot assay of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), phosphorylated (p)-p65, p65, p-IκB and IκB. The results showed that in rats with T2DM, molecular hydrogen treatment decreased fasting blood glucose levels, increased hepatic glycogen synthesis and improved insulin sensitivity. Treatment with molecular hydrogen also increased the production of SOD whilst decreasing the production of MDA. In addition, molecular hydrogen alleviated the pathological changes exhibited by pancreatic islets and kidney during T2DM. Mechanistically, molecular hydrogen decreased TLR4 and MyD88 expression levels whilst also decreasing p65 and NF-κB inhibitor phosphorylation. In conclusion, molecular hydrogen exerted therapeutic effects against T2DM by improving hyperglycemia and inhibiting oxidative stress through mechanisms that are associated with the TLR4/MyD88/NF-κB signaling pathway.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA