Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Am J Transplant ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38552961

ABSTRACT

Recently published studies in both murine models and a meta-analysis of non-human primate renal transplant studies showed that anti-CD154 reagents conferred a significant survival advantage over CD40 blockers in both animal models and across multiple organs. Here we sought to compare the induction of donor-reactive forkhead box P3+-induced regulatory T cells (Foxp3+ iTreg) in mice treated with anti-CD154 versus anti-CD40 monoclonal antibodies (mAbs). Results indicated that while treatment with anti-CD154 mAb resulted in a significant increase in the frequency of donor-reactive CD4+ Foxp3+ iTreg following transplantation, treatment with anti-CD40 or Cd40 deficiency failed to recapitulate this result. Because we recently identified CD11b as an alternate receptor for CD154 during alloimmunity, we interrogated the role of CD154:CD11b interactions in the generation of Foxp3+ iTreg and found that blockade of CD11b in Cd40-/- recipients resulted in increased donor-reactive Foxp3+ iTreg as compared with CD40 deficiency alone. Mechanistically, CD154:CD11b inhibition decreased interleukin (IL)-1ß from CD11b+ and CD11c+ dendritic cells, and blockade of IL-1ß synergized with CD40 deficiency to promote Foxp3+ iTreg induction and prolong allograft survival. Taken together, these data provide a mechanistic basis for the observed inferiority of anti-CD40 blockers as compared with anti-CD154 mAb and illuminate an IL-1ß-dependent mechanism by which CD154:CD11b interactions prevent the generation of donor-reactive Foxp3+ iTreg during transplantation.

2.
Autophagy ; 20(3): 629-644, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37963060

ABSTRACT

PYCARD (PYD and CARD domain containing), a pivotal adaptor protein in inflammasome assembly and activation, contributes to innate immunity, and plays an essential role in the pathogenesis of atherosclerosis and restenosis. However, its roles in microRNA biogenesis remain unknown. Therefore, this study aimed to investigate the roles of PYCARD in miRNA biogenesis and neointima formation using pycard knockout (pycard-/-) mice. Deficiency of Pycard reduced circulating miRNA profile and inhibited Mir17 seed family maturation. The systemic pycard knockout also selectively reduced the expression of AGO2 (argonaute RISC catalytic subunit 2), an important enzyme in regulating miRNA biogenesis, by promoting chaperone-mediated autophagy (CMA)-mediated degradation of AGO2, specifically in adipose tissue. Mechanistically, pycard knockout increased PRMT8 (protein arginine N-methyltransferase 8) expression in adipose tissue, which enhanced AGO2 methylation, and subsequently promoted its binding to HSPA8 (heat shock protein family A (Hsp70) member 8) that targeted AGO2 for lysosome degradation through chaperone-mediated autophagy. Finally, the reduction of AGO2 and Mir17 family expression prevented vascular injury-induced neointima formation in Pycard-deficient conditions. Overexpression of AGO2 or administration of mimic of Mir106b (a major member of the Mir17 family) prevented Pycard deficiency-mediated inhibition of neointima formation in response to vascular injury. These data demonstrate that PYCARD inhibits CMA-mediated degradation of AGO2, which promotes microRNA maturation, thereby playing a critical role in regulating neointima formation in response to vascular injury independently of inflammasome activity and suggest that modulating PYCARD expression and function may represent a powerful therapeutic strategy for neointima formation.Abbreviations: 6-AN: 6-aminonicotinamide; ACTB: actin, beta; aDMA: asymmetric dimethylarginine; AGO2: argonaute RISC catalytic subunit 2; CAL: carotid artery ligation; CALCOCO2: calcium binding and coiled-coil domain 2; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSD: cathepsin D; DGCR8: DGCR8 microprocessor complex subunit; DOCK2: dedicator of cyto-kinesis 2; EpiAdi: epididymal adipose tissue; HSPA8: heat shock protein family A (Hsp70) member 8; IHC: immunohistochemical; ISR: in-stent restenosis; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; miRNA: microRNA; NLRP3: NLR family pyrin domain containing 3; N/L: ammonium chloride combined with leupeptin; PRMT: protein arginine methyltransferase; PVAT: peri-vascular adipose tissues; PYCARD: PYD and CARD domain containing; sDMA: symmetric dimethylarginine; ULK1: unc-51 like kinase 1; VSMCs: vascular smooth muscle cells; WT: wild-type.


Subject(s)
Chaperone-Mediated Autophagy , MicroRNAs , Vascular System Injuries , Animals , Mice , MicroRNAs/genetics , Inflammasomes/metabolism , Autophagy/physiology , Neointima , RNA-Binding Proteins , Heat-Shock Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , GTPase-Activating Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism
3.
Autophagy ; 19(5): 1491-1511, 2023 05.
Article in English | MEDLINE | ID: mdl-36300763

ABSTRACT

Ischemia-induced angiogenesis is critical for blood flow restoration and tissue regeneration, but the underlying molecular mechanism is not fully understood. ATG7 (autophagy related 7) is essential for classical degradative macroautophagy/autophagy and cell cycle regulation. However, whether and how ATG7 influences endothelial cell (EC) function and regulates post-ischemic angiogenesis remain unknown. Here, we showed that in mice subjected to femoral artery ligation, EC-specific deletion of Atg7 significantly impaired angiogenesis, delayed the recovery of blood flow reperfusion, and displayed reduction in HIF1A (hypoxia inducible factor 1 subunit alpha) expression. In addition, in cultured human umbilical vein endothelial cells (HUVECs), overexpression of HIF1A prevented ATG7 deficiency-reduced tube formation. Mechanistically, we identified STAT1 (signal transducer and activator of transcription 1) as a transcription suppressor of HIF1A and demonstrated that ablation of Atg7 upregulated STAT1 in an autophagy independent pathway, increased STAT1 binding to HIF1A promoter, and suppressed HIF1A expression. Moreover, lack of ATG7 in the cytoplasm disrupted the association between ATG7 and the transcription factor ZNF148/ZFP148/ZBP-89 (zinc finger protein 148) that is required for STAT1 constitutive expression, increased the binding between ZNF148/ZFP148/ZBP-89 and KPNB1 (karyopherin subunit beta 1), which promoted ZNF148/ZFP148/ZBP-89 nuclear translocation, and increased STAT1 expression. Finally, inhibition of STAT1 by fludarabine prevented the inhibition of HIF1A expression, angiogenesis, and blood flow recovery in atg7 KO mice. Our work reveals that lack of ATG7 inhibits angiogenesis by suppression of HIF1A expression through upregulation of STAT1 independently of autophagy under ischemic conditions, and suggest new therapeutic strategies for cancer and cardiovascular diseases.Abbreviations: ATG5: autophagy related 5; ATG7: autophagy related 7; atg7 KO: endothelial cell-specific atg7 knockout; BECN1: beclin 1; ChIP: chromatin immunoprecipitation; CQ: chloroquine; ECs: endothelial cells; EP300: E1A binding protein p300; HEK293: human embryonic kidney 293 cells; HIF1A: hypoxia inducible factor 1 subunit alpha; HUVECs: human umbilical vein endothelial cells; IFNG/IFN-γ: Interferon gamma; IRF9: interferon regulatory factor 9; KPNB1: karyopherin subunit beta 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MEFs: mouse embryonic fibroblasts; MLECs: mouse lung endothelial cells; NAC: N-acetyl-l-cysteine; NFKB1/NFκB: nuclear factor kappa B subunit 1; PECAM1/CD31: platelet and endothelial cell adhesion molecule 1; RELA/p65: RELA proto-oncogene, NF-kB subunit; ROS: reactive oxygen species; SP1: Sp1 transcription factor; SQSTM1/p62: sequestosome 1; STAT1: signal transducer and activator of transcription 1; ULK1: unc-51 like autophagy activating kinase 1; ulk1 KO: endothelial cell-specific ulk1 knockout; VSMCs: mouse aortic smooth muscle cells; WT: wild type; ZNF148/ZFP148/ZBP-89: zinc finger protein 148.


Subject(s)
Autophagy , Fibroblasts , Mice , Humans , Animals , Autophagy/genetics , HEK293 Cells , STAT1 Transcription Factor , Human Umbilical Vein Endothelial Cells , Ischemia , Hypoxia-Inducible Factor 1 , Karyopherins , DNA-Binding Proteins , Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit
4.
Biomed Res Int ; 2022: 4267673, 2022.
Article in English | MEDLINE | ID: mdl-35425838

ABSTRACT

Objective: Preventive effect of hippocampal sparing on cognitive dysfunction of patients undergoing whole-brain radiotherapy and imaging assessment of hippocampal volume changes. Methods: Forty patients with brain metastases who attended Liaoning Cancer Hospital from January 2018 to December 2019 were identified as research subjects and were randomly divided into a control group and an experimental group, with 20 cases in each group. The control group was treated with whole-brain radiotherapy (WBRT), and the experimental group was treated with hippocampal sparing-WBRT (HS-WBRT). The Montreal Cognitive Assessment (MoCA) score, Eastern Cooperative Oncology Group (ECOG) score, cancer quality-of-life questionnaire (QLQ-C3O) score, hippocampal volume changes, and prognosis of the two groups were compared. Results: The MoCA scores decreased in both groups at 3, 6, and 12 months after radiotherapy, with significantly higher scores in the experimental group than in the control group (P < 0.05). After radiotherapy, both groups had lower ECOG scores, with those in the experimental group being significantly lower than those in the control group (P < 0.05). After radiotherapy, the QLQ-C30 score was elevated in both groups, and that of the experimental group was significantly higher than that of the control group (P < 0.05). The experimental group outperformed the control group in terms of the prognosis (P < 0.05). The hippocampal volume of the control group was significantly smaller than that of the experimental group (P < 0.05). Conclusion: The application of hippocampal sparing in patients receiving whole-brain radiotherapy is effective in preventing cognitive dysfunction, improving the quality of life and prognosis of patients, and avoiding shrinkage of hippocampal volume.


Subject(s)
Brain Neoplasms , Cognitive Dysfunction , Radiotherapy, Intensity-Modulated , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/radiotherapy , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Cranial Irradiation/adverse effects , Cranial Irradiation/methods , Hippocampus/diagnostic imaging , Humans , Quality of Life , Radiotherapy, Intensity-Modulated/methods
5.
Oncol Rep ; 46(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34296306

ABSTRACT

Accumulating evidence indicates that circular (circ)RNAs exhibit complex functions in diverse malignant tumors, including non­small cell lung cancer (NSCLC). The role of the circRNA transcription adaptor 2A (circTADA2A) in NSCLC remains unclear. The expression, function and mechanism of circTADA2A in NSCLC development were investigated in the present study. The results revealed that circTADA2A was upregulated in NSCLC, and that knockdown of circTADA2A inhibited cell proliferation and migration in the NSCLC cell lines A549 and H1299. Functional assays demonstrated that circTADA2A promoted proliferation and migration via interacting with microRNA (miR)­638. Bioinformatics and reverse transcription­quantitative PCR assay confirmed that miR­638 was expressed at low levels in NSCLC. In addition, it was found that miR­638 served a tumor­suppressive role and suppressed proliferation and migration via PCNA clamp associated factor (KIAA0101) inhibition in A549 and H1299 cells. Lastly, it was verified that circTADA2A promoted cell proliferation and migration, at least partially, via miR­638/KIAA0101 signaling in A549 and H1299 cells. In summary, the present study showed that circTADA2A promoted NSCLC cell proliferation and migration via modulating miR­638/KIAA0101 signaling.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Circular , Transcription Factors/metabolism , A549 Cells , Aged , Aged, 80 and over , Area Under Curve , Cell Movement , Cell Proliferation , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Dactinomycin/metabolism , Eukaryotic Initiation Factor-4A/genetics , Exoribonucleases/metabolism , Female , Humans , Male , MicroRNAs/genetics , Middle Aged , Prognosis , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors/genetics , Up-Regulation
6.
Front Oncol ; 11: 651632, 2021.
Article in English | MEDLINE | ID: mdl-34150619

ABSTRACT

BACKGROUND: Inflammation-related gene polymorphisms are some of the most important determinants for cancer susceptibility, clinical phenotype diversity, and the response to radiotherapy and chemotherapy. However, the relationship between these polymorphisms and head and neck squamous cell carcinoma (HNSCC) remains unclear. The aim of this study was to investigate the role of inflammation-related gene polymorphisms in the developmental risk and radiotherapy sensitivity of HNSCC. METHODS: The Matrix-Assisted Laser Desorption Ionization Time of Flight (MALDI-TOF) genotyping system was used to genotype 612 individuals from a Chinese population for 28 inflammation-related gene polymorphisms. RESULTS: The protein kinase B (AKT1) rs1130233 TT, dominance model (CT+TT vs. CC), recessive model (TT vs. CT+CC), and rs2494732 CC genotypes were associated with reduced risk of HNSCC (P=0.014; P=0.041; P=0.043). The polymeric immunoglobulin receptor (PIGR) rs291097 GA, dominance model (GA+AA vs. GG), and rs291102 dominance model (GA+AA vs. GG) were associated with increased risk of HNSCC (P=0.025; P=0.025; P=0.040). The interleukin-4 receptor-α (IL-4RA) rs1801275 AA genotype was significantly correlated with increased radiotherapy sensitivity of HNSCC patients (P=0.030). In addition, age ≤ 60 years, non-smoker status, and normal levels of squamous cell carcinoma antigen (SCC) were found to be associated with increased radiotherapy sensitivity of HNSCC patients (P=0.033; P=0.033; P=0.030). CONCLUSION: The AKT1 rs1130233, AKT1 rs2494732, PIGR rs291097, and PIGR rs291102 polymorphisms were significantly related to the risk of HNSCC. The IL-4RA rs1801275 polymorphism, age ≤ 60 years, non-smoker status, and normal levels of SCC were significantly associated with increased radiotherapy sensitivity of HNSCC.

7.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L606-L616, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29074487

ABSTRACT

Dysfunction and injury of endothelial cells play critical roles in pulmonary arterial hypertension, including aberrant proliferation, suppressed apoptosis, and excessive angiogenesis. The 12-lipoxygenase and 12-hydroxyeicosatetraenoic acid pathway, which has been considered as a crucial mediator, elevates pulmonary vascular resistance and pulmonary arterial pressure. However, the mechanisms underlying the bioactivity of 12-hydroxyeicosatetraenoic acid in pulmonary vasculature, especially in endothelial cells, are still elusive. Thus we aim to determine the key role of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid in angiogenesis and survival of pulmonary artery endothelial cells and ascertain the signaling pathways participating in the pathological process. Here we establish that hypoxia increases the formation of endogenous 12-hydroxyeicosatetraenoic acid through stimulation of 12-lipoxygenase. Furthermore, we put forward new information that 12-hydroxyeicosatetraenoic acid promotes endothelial cell migration and tube formation, whereas it inhibits the serum deprivation-induced apoptotic responses under hypoxia. Particularly, the regulatory effects of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid on pulmonary artery endothelial cells, at least in part, depend on phosphatidylinositol 3-kinase (PI3K)/Akt signaling activation. Taken together, these results may have significant implications for understanding of pulmonary hypertension and offer a potential therapeutic concept focusing on the 12-lipoxygenase/12-hydroxyeicosatetraenoic acid signaling system.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Arachidonate 12-Lipoxygenase/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/physiopathology , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Arachidonate 12-Lipoxygenase/genetics , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Male , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinase/genetics , Proto-Oncogene Proteins c-akt/genetics , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats , Rats, Wistar , Signal Transduction/drug effects
8.
J Mol Med (Berl) ; 95(9): 977-993, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28593577

ABSTRACT

Abnormal pulmonary arterial smooth muscle cells (PASMCs) proliferation is an important pathological process in hypoxic pulmonary arterial hypertension. Mitochondrial dynamics and quality control have a central role in the maintenance of the cell proliferation-apoptosis balance. However, the molecular mechanism is still unknown. We used hypoxic animal models, cell biology, and molecular biology to determine the effect of mitofusin 1 (Mfn1) on hypoxia-mediated PASMCs mitochondrial homeostasis. We found that Mfn1 expression was increased in hypoxia, which was crucial for hypoxia-induced mitochondrial dysfunction and smooth muscle cell proliferation as well as hypoxia-stimulated cell-cycle transition from the G0/G1 phase to S phase. Subsequently, we studied the role of microRNAs in mitochondrial function associated with PASMC proliferation under hypoxic conditions. The promotive effect of Mfn1 on pulmonary vascular remodeling was alleviated in the presence of miR-125a agomir, and miR-125a antagomir mimicked the hypoxic damage effects to mitochondrial homeostasis. Moreover, in vivo and in vitro treatment with miR-125a agomir protected the pulmonary vessels from mitochondrial dysfunction and abnormal remodeling. In the present study, we determined that mitochondrial homeostasis, particularly Mfn1, played an important role in PASMCs proliferation. MiR-125a, an important underlying factor, which inhibited Mfn1 expression and decreased PASMCs disordered growth during hypoxia. These results provide a theoretical basis for the prevention and treatment of pulmonary vascular remodeling. KEY MESSAGES: Hypoxia leads to upregulation of mitofusin 1 (Mfn1) both in vivo and in vitro. Mfn1 is involved in hypoxia-induced PASMCs proliferation. Mfn1-mediated mitochondrial homeostasis is regulated by miR-125a. MiR-125a plays a role in PASMCs oxidative phosphorylation and glycolysis.


Subject(s)
Hypoxia/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , RNA Interference , Vascular Remodeling/genetics , Animals , Cell Proliferation , Gene Expression , Glycolysis , Homeostasis , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/metabolism , Lung/metabolism , Lung/physiopathology , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Rats
9.
Br J Pharmacol ; 171(14): 3352-63, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24467360

ABSTRACT

BACKGROUND AND PURPOSE: Dysfunction and injury of endothelial cells in the pulmonary artery play critical roles in the hypertension induced by chronic hypoxia. One consequence of hypoxia is increased activity of 15-hydroxyprostaglandin dehydrogenase (PGDH). Here, we have explored, in detail, the effects of hypoxia on the proliferation of pulmonary artery endothelial cells. EXPERIMENTAL APPROACH: We used bromodeoxyuridine incorporation, cell-cycle analysis, immunohistochemistry and Western blot analysis to study the effects of hypoxia, induced 15-PGDH) activity and its product, 15-keto-6Z, 8Z, 11Z, 13E-eicosatetraenoic acid (15-KETE), on endothelial cell proliferation. Scratch-wound and tube formation assays were also used to study migration of endothelial cells. KEY RESULTS: 15-KETE increased DNA synthesis and enhanced the transition from the G0 /G1 phase to the S phase in hypoxia. Inhibition of 15-PGDH or siRNA for 15-PGDH reversed these effects. 15-KETE also activated the ERK1/2 signalling pathway. 15-KETE-induced cell migration and tube formation were reversed by blocking ERK1/2, but not the p38 MAPK pathway. CONCLUSIONS AND IMPLICATIONS: Hypoxia-induced endothelial proliferation and migration, an important underlying mechanism contributing to hypoxic pulmonary vascular remodelling, appears to be mediated by 15-PGDH and 15-KETE, via the ERK1/2 signalling pathway.


Subject(s)
Arachidonic Acids/metabolism , Cell Proliferation , Endothelial Cells/cytology , Hydroxyprostaglandin Dehydrogenases/metabolism , Hypoxia/metabolism , MAP Kinase Signaling System , Pulmonary Artery/cytology , Animals , Cell Movement , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pulmonary Artery/metabolism , Rats , Rats, Wistar , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...