MeCP2 modulates the canonical Wnt pathway activation by targeting SFRP4 in rheumatoid arthritis fibroblast-like synoviocytes in rats

2013 ◽  
Vol 25 (3) ◽  
pp. 598-608 ◽  
Author(s):  
Cheng-gui Miao ◽  
Cheng Huang ◽  
Yan Huang ◽  
Ying-ying Yang ◽  
Xu He ◽  
...  
Biochimie ◽  
2014 ◽  
Vol 106 ◽  
pp. 149-156 ◽  
Author(s):  
Cheng-gui Miao ◽  
Ying-ying Yang ◽  
Xu He ◽  
Cheng Huang ◽  
Yan Huang ◽  
...  

2014 ◽  
Vol 26 (5) ◽  
pp. 1068-1074 ◽  
Author(s):  
Jinxiao Chen ◽  
Hongwei Yan ◽  
Dan-ni Ren ◽  
Yan Yin ◽  
Zhi Li ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 741-741
Author(s):  
Marina Bolzoni ◽  
Simona Colla ◽  
Paola Storti ◽  
Gaetano Donofrio ◽  
Manuela Abeltino ◽  
...  

Abstract Abstract 741 Osteogenic differentiation of human mesenchymal stem cells (hMSC) is typically impaired in multiple myeloma (MM) patients leading to osteoblast deficiency. Canonical Wnt signal pathway is critical in the regulation of bone formation process and its activation in osteoblastic cells improves bone mass reducing the development of osteolytic lesions in MM mouse model. Together to canonical Wnt signaling, a non-canonical Wnt pathway, independently to β-catenin activation, has been identified. Non-canonical Wnt signaling is transduced through FZD receptor and Ror2 co-receptor to several cascades either disheveled pathways involving Rho family small GTPase and JNK or Ca++ dependent pathways involving the nuclear factor of activated T cells (NFAT). Interestingly, recent evidences suggest that non-canonical Wnt pathway activation by Wnt5a or Wnt4, rather than canonical one by Wnt3a, stimulates the osteogenic properties of hMSC through Ror2 activation. The effect of MM cells on non-canonical Wnt pathway as well as the role of the activation of this pathway in hMSC on the osteogenic differentiation impairment induced by MM cells are not known and have been investigated in the present study. First we checked the expression of non-canonical Wnt related molecules by bone marrow (BM) hMSC and osteoprogenitor cells (PreOB) at the first passage by oligonucleotide arrays. We found that both cells expressed the activator of non-canonical Wnt pathways Wnt5a but lack of express the main activators of canonical Wnt signaling as Wnt1, Wnt3a and Wnt8. The presence of the Wnt5a receptor FZD2 and FZD5 was also detected in both cells as well as of Ror2. Interestingly we found that osteogenic differentiation of hMSC towards preOB significantly increased Ror2 but not Wnt5a expression. Secondly, we performed a series of co-culture between PreOB and MM cells using either the human myeloma cell lines (JJN3, XG-1, XG-6, KMS12, KMS27) or purified CD138+ cells obtained from MM patients finding that MM cells inhibit Ror2 protein expression by PreOB and consistently the activity of NFATc1 at nuclear level. Following activation of non-canonical Wnt signaling pathway either by Wnt5a treatment or by the induction of both Wnt5a and Ror2 overexpression by lentivirus vectors have been performed in primary hMSC obtained from MM patients. The efficiency of Wnt5a and Ror2 trasduction was checked by GFP expression using flow cytometry whereas the efficacy was evaluated by the level of Wnt5a and Ror2 mRNA and protein expression as well as by the intracytoplasmatic increase of Ca++ influx, phospho-PKC expression and NFATc1 activity. We found that Wnt5a treatment as well as Wnt5a or Ror2 overexpression significantly increased osteogenic differentiation and the expression of alkaline phosphatase in hMSC. Consistently, in the co-culture system with MM cells, Wnt5a and Ror2 overexpression by hMSC blunted the inhibitory effect of MM cells on alkaline phosphatase expression and osteogenic differentiation. Finally, these observations were further confirmed showing that Wnt5a or Ror2 silencing in PreOB by siRNA or shRNA trasfection, respectively inhibited the expression of osteogenic markers alkaline phosphatase, osteocalcin and collagen I. In conclusion our data indicate that activation of non-canonical Wnt5a/Ror2/Ca++ signal pathway in hMSC increases osteogenic differentiation and counterbalance the inhibitory effect of MM cell suggesting that this pathway could represent a potential target in MM microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1460-1460
Author(s):  
Teppei Sakoda ◽  
Yoshikane Kikushige ◽  
Toshihiro Miyamoto ◽  
Koichi Akashi

Abstract We originally identified T-cell immunoglobulin mucin-3 (TIM-3) as a leukemic stem cells (LSCs)-specific surface molecule and a useful marker for discriminating LSCs from hematopoietic stem cells (HSCs). Furthermore, we recently identified an unique autocrine loop composed of TIM-3 and its ligand galectin-9 (Gal-9). This TIM-3/Gal-9 autocrine loop enhances self-renewal capacity of AML-LSCs and contributes to leukemia progression(Kikushige et al., Cell Stem Cell 2015). To clarify the molecular mechanism how TIM-3 signaling enhances stem cell properties of AML-LSCs, we performed shRNA-mediated knock-down(KD) of TIM-3 in KASUMI-3 (TIM-3+ AML cell line). As a result, TIM-3-KD significantly reduce the proliferation of KASUMI-3. To investigate how TIM-3-KD attenuated leukemia propagation, we compared gene expression profile of scramble-infected control and TIM-3-KD KASUMI-3 cells. Gene Set Enrichment Analysis (GSEA) revealed TIM-3-KD resulted in the significaltly attenuated expression of HSCs and LSCs-related genes (Eppert et al., Nature Med 2011) and canonical Wnt pathway-related genes (BioCarta). Since canonical Wnt pathway is known to regulate stem cell properties via inducing the nucleus accumulation ofβ-catenin in many types of stem cells including HSCs, we next examined the β-catenin level in TIM-3-KD-KASUMI-3 cells. Array scan analysis revealed that KD of TIM-3 significantly impaired the the nucleus accumulation of β-catenin in KASUMI-3 cells. In various types of cancers, the constitutive activation of β-catenin is one of the most common oncogenic signatures, and it is mainly driven by somatic mutations in the genes related to the canonical Wnt pathway. However, such mutations are rarely detected in AML.We, therefore, hypothesized that TIM-3/Gal-9 autocrine loop signaling might be a novel mutation-independent machinery for the constitutive activation of canonical Wnt pathway in primary AML. To test this hypothethis, we first evaluated the phosphorylation status of LDL receptor-related protein 6 (LRP6), because LRP6 phosphorylation is a crucial step for canonical Wnt pathway activation. Surprisingly, Gal-9 stimulation induced phosphorylation of LRP6 and subsequent β-catenin accumulation in primary TIM-3+ AML cells even in the absence of Wnt ligand. On the other hand, LRP6 phosphorylation induced by Gal-9 stimulation was canceled by the blocking antibody to TIM-3 (F38-2E2). These results indicate that Gal-9 stimulation should induce canonical Wnt pathway activation via TIM-3 dependent manner in primary AML cells. Since Gal-9 ligation to TIM-3 is known to induce phosphorylation of tyrosine residues of TIM-3 cytoplasmic tail, leading to the recruitment and activation of several Src family kinases (SFKs) for downstream signal transduction. We found that TIM-3/Gal-9 interaction recruited hematopoietic cell kinase (HCK), a member of SFKs, to TIM-3 and activated HCK by phosphorylation of the SH1 domain (Y410 in HCK) in TIM-3+ AML cells. Furthermore, we confirmed a specific HCK inhibitor A-419259 efficiently blocked the LRP6 phosphorylation and subsequent β-catenin accumulation induced by TIM-3/Gal-9 interaction in primary AML cells. These results suggest that the canonical Wnt pathway activation induced by Gal-9 ligation to TIM-3 is mainly mediated by HCK in primary AML cells. We next focused on p120-catenin as a bridging molecule between HCK activation and LRP6 phosphorylation because, as previously reported, this molecule can be directly activated by SFKs and the activation of this molecule is an initial step for canonical Wnt pathway activation preceding LRP6 phosphorylation. We found that Gal-9 ligation to TIM-3 immediately induced the direct association of HCK and p120-catenin, leading to the activation of p120-catenin in TIM-3+ AML cells. We also confirmed that specific inhibition of HCK completely abrogated the activation of p120-catenin and subsequent canonical Wnt pathway activation in primary TIM-3+ AML cells, indicating that the activation of p120-catenin mediated by HCK is a bridging moleculer machinery for connecting TIM-3 signaling and previously identified canonical Wnt pathway. Since TIM-3 as well as HCK is specifically expressed in LSCs but not in HSCs, we conclude that TIM-3/Gal-9 autocrine loop is a novel Wnt-ligands independent machinery for the constitutive activation of canonical Wnt pathway specific for AML-LSCs. (summarized in figure) Figure. Figure. Disclosures Akashi: Celgene: Research Funding, Speakers Bureau; Pfizer: Research Funding; Bristol-Myers Squibb: Research Funding, Speakers Bureau; Kyowa Hakko Kirin: Research Funding, Speakers Bureau; Taiho Pharmaceutical: Research Funding; MSD: Research Funding; Asahi-kasei: Research Funding; Chugai Pharma: Research Funding; Ono Pharmaceutical: Research Funding; Otsuka Pharmaceutical: Research Funding; Eli Lilly Japan: Research Funding; Novartis pharma: Research Funding; Eisai: Research Funding; sanofi: Research Funding; Astellas Pharma: Research Funding.


2018 ◽  
Vol 50 (07) ◽  
pp. 575-581 ◽  
Author(s):  
Carlos Jucá ◽  
Leandro Colli ◽  
Clarissa Martins ◽  
Marina Campanini ◽  
Beatriz Paixão ◽  
...  

Abstract CTNNB1 mutations and abnormal β-catenin distribution are associated with the pathogenesis of adamantinomatous craniopharyngioma (aCP). We evaluated the expression of the canonical Wnt pathway components in aCPs and its association with CTNNB1 mutations and tumor progression. Tumor samples from 14 aCP patients and normal anterior pituitary samples from eight individuals without pituitary disease were studied. Gene expression of Wnt pathway activator (WNT4), inhibitors (SFRP1, DKK3, AXIN1, and APC), transcriptional activator (TCF7), target genes (MYC, WISP2, and, CDH1), and Wnt modulator (TP53) was evaluated by qPCR. β-Catenin, MYC, and WISP2 expression was determined by immunohistochemistry (IHC). The transcription levels of all genes studied, except APC, were higher in aCPs as compared to controls and TCF7 mRNA levels correlated with CTNNB1 mutation. CDH1 mRNA was overexpressed in tumor samples of patients with disease progression in comparison to those with stable disease. β-Catenin was positive and aberrantly distributed in 11 out of 14 tumor samples. Stronger β-catenin immunostaining associated positively with tumor progression. MYC positive staining was found in 10 out of 14 cases, whereas all aCPs were negative for WISP2. Wnt pathway genes were overexpressed in aCPs harboring CTNNB1 mutations and in patients with progressive disease. Recurrence was associated with stronger staining for β-catenin. These data suggest that Wnt pathway activation contributes to the pathogenesis and prognosis of aCPs.


2021 ◽  
Vol 12 ◽  
Author(s):  
Xiao Wang ◽  
Jun Chang ◽  
Guoliang Zhou ◽  
Chenglong Cheng ◽  
Youyi Xiong ◽  
...  

The pathogenesis of rheumatoid arthritis (RA) is still not fully clarified, and the development of therapeutic drugs for RA is particularly urgent. Our group studies a possibility that circ_ 0015756/miR-942-5p may participate in the pathogenesis of RA through disordered Cullin 4B (CUL4B) and the traditional Chinese medicine compound Huangqin Qingre Chubi Capsule (HQC) may inhibit the pathogenesis of RA through the CUL4B/Wnt pathway. Data showed that the expression of circ_0015756 increased not only in fibroblast-like synoviocytes (FLS) of RA, but also in synovium and FLS of CIA mice, and the expression of miR-942-5p decreased. Abnormal circ_0015756 up-regulated the CUL4B expression and activated the canonical Wnt signaling pathway by inhibiting the expression of miR-942-5p. Circ_0015756 participated in the pathogenesis of RA and promoted the abnormal proliferation of FLS. Further, circ_0015756 activated the secretion of IL-1 and IL-8 and promoted the production of RA pathological gene MMP3 and fibronectin. Further analysis showed that HQC inhibited the pathogenesis of RA through the CUL4B/Wnt pathway, and the specific target was CUL4B. HQC interfered with the effects of circ_0015756 on the pathogenesis of RA by inhibiting the CUL4B, showing a good therapeutic effect on RA.


2005 ◽  
Vol 65 (14) ◽  
pp. 6199-6206 ◽  
Author(s):  
Aykut Üren ◽  
Shannon Fallen ◽  
Hang Yuan ◽  
Alp Usubütün ◽  
Türkan Küçükali ◽  
...  

2009 ◽  
Vol 28 (2) ◽  
pp. 121-122
Author(s):  
D. Takashi ◽  
B. John ◽  
P. Prem ◽  
T. Jennifer

Sign in / Sign up

Export Citation Format

Share Document