112 results on '"Tgif1"'
Search Results
2. Phosphorylation of TG‐interacting factor 1 at carboxyl‐terminal sites in response to insulin regulates adipocyte differentiation.
- Author
-
Chang, Yu‐Hao, Tseng, Yu‐Hua, Wang, Ju‐Ming, Tsai, Yau‐Sheng, Liu, Xin‐Lei, and Huang, Huei‐Sheng
- Subjects
- *
INSULIN , *PHOSPHORYLATION , *ADIPOGENESIS , *CELL proliferation , *STEM cells , *INSULIN receptors , *FAT cells , *ADIPOSE tissues , *DEPHOSPHORYLATION - Abstract
TG‐interacting factor 1 (TGIF1) contributes to the differentiation of murine white preadipocyte and human adipose tissue‐derived stem cells; however, its regulation is not well elucidated. Insulin is a component of the adipogenic cocktail that induces ERK signaling. TGIF1 phosphorylation and sustained stability in response to insulin were reduced through the use of specific MEK inhibitor U0126. Mutagenesis at T235 or T239 residue of TGIF1 in preadipocytes led to dephosphorylation of TGIF1. The reduced TGIF1 stability resulted in an increase in p27kip1 expression, a decrease in phosphorylated Rb expression and cellular proliferation, and a reduced accumulation of lipids compared to the TGIF1‐overexpressed cells. These findings highlight that insulin/ERK‐driven phosphorylation of the T235 or T239 residue at TGIF1 is crucial for adipocyte differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Tgif1-deficiency impairs cytoskeletal architecture in osteoblasts by activating PAK3 signaling
- Author
-
Simona Bolamperti, Hiroaki Saito, Sarah Heerdmann, Eric Hesse, and Hanna Taipaleenmäki
- Subjects
osteoblast spreading ,bone regeneration ,Tgif1 ,PAK3 ,PTH ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Osteoblast adherence to bone surfaces is important for remodeling bone tissue. This study demonstrates that deficiency of TG-interacting factor 1 (Tgif1) in osteoblasts results in altered cell morphology, reduced adherence to collagen type I-coated surfaces, and impaired migration capacity. Tgif1 is essential for osteoblasts to adapt a regular cell morphology and to efficiently adhere and migrate on collagen type I-rich matrices in vitro. Furthermore, Tgif1 acts as a transcriptional repressor of p21-activated kinase 3 (Pak3), an important regulator of focal adhesion formation and osteoblast spreading. Absence of Tgif1 leads to increased Pak3 expression, which impairs osteoblast spreading. Additionally, Tgif1 is implicated in osteoblast recruitment and activation of bone surfaces in the context of bone regeneration and in response to parathyroid hormone 1–34 (PTH 1–34) treatment in vivo in mice. These findings provide important novel insights in the regulation of the cytoskeletal architecture of osteoblasts.
- Published
- 2024
- Full Text
- View/download PDF
4. miR-139-5p mediates TGIF1 to regulate the TGFβ pathway and inhibit growth of esophageal squamous cell carcinoma cells.
- Author
-
Fan, Xiaowu
- Abstract
Summary: Background: Esophageal squamous cell carcinoma (ESCC) is the most prevalent histological subtype of esophageal cancer. miR-139-5p has been shown to have abnormal expression in ESCC. We intend to probe into the roles of miR-139-5p in ESCC, thus providing references for investigating underlying therapeutic targets. Methods: miR-139-5p expression in esophageal carcinoma was predicted by the Starbase online website. miR-139-5p expression in human ESCC cell lines (KYSE-150, KYSE-410, KSE-30) and human esophageal epithelial cell line Het1A was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR). KYSE-150 cells were manipulated with miR-139-5p mimic to upregulate miR-139-5p. Cell colony formation, viability, apoptosis, migration, and invasion were assessed by colony formation assay, CCK‑8, flow cytometry, wound healing test, and Transwell. The targeted binding between miR-139-5p and TGIF1 was predicted on Starbase, Targetscan, and miRDB online websites and verified by Dual-Luciferase assay. Cells were transfected with miR-139-5p mimic and pcDNA3.1 TGIF1. The phosphorylation levels of TGFβ pathway-related proteins Smad2 and Smad3 were assessed by RT-qPCR and western blot. Results: miR-139-5p was underexpressed in ESCC cells. miR-139-5p overexpression reduced ESCC cell viability, migration, and invasion, and promoted apoptosis. TGIF1 overexpression averted miR-139-5p mimic-inhibited ESCC cell growth. miR-139-5p overexpression decreased the phosphorylation levels of TGFβ pathway-related proteins Smad2 and Smad3, while the phosphorylation levels were increased by additional TGIF1 vector transfection. Conclusion: miR-139-5p mediates TGIF1 to regulate TGFβ pathway and inhibit ESCC cell growth. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Circular RNA CDR1as Mediated by Human Antigen R (HuR) Promotes Gastric Cancer Growth via miR-299-3p/TGIF1 Axis.
- Author
-
Li, Rong, Xu, Xuejing, Gao, Shuo, Wang, Junyi, Hou, Jie, Xie, Zhenfan, Luo, Lan, Shen, Han, Xu, Wenrong, and Jiang, Jiajia
- Subjects
- *
STOMACH tumors , *CIRCULAR RNA , *FLOW cytometry , *IN vitro studies , *XENOGRAFTS , *IN vivo studies , *RNA-binding proteins , *WESTERN immunoblotting , *ANIMAL experimentation , *MICRORNA , *SMALL interfering RNA , *PRECIPITIN tests , *APOPTOSIS , *SIGNAL peptides , *BIOINFORMATICS , *CELL survival , *GENES , *CELL proliferation , *RESEARCH funding , *GENETIC techniques , *COLORIMETRY , *BIOLOGICAL assay , *POLYMERASE chain reaction , *ANTIGENS , *CASPASES , *MICE , *EVALUATION - Abstract
Simple Summary: Gastric cancer (GC) is one of the most common malignancies with limited therapeutic targets available. CDR1as, an endogenous circular RNA with a closed loop structure, has been reported to be a crucial regulator and promising biomarker in various tumors. However, whether and how CDR1as participates in GC progression remains not well characterized. In this study, we explored the biological roles, the downstream molecular mechanism and the upstream regulator of CDR1as in GC. We found that CDR1as mediated by human antigen R (HuR) promotes GC growth through the miR-299-3p/TGIF1 axis, which provides new insights into GC pathogenesis and brings a new potential target for clinical GC therapy. Background: Gastric cancer (GC) remains a common malignancy worldwide with a limited understanding of the disease mechanisms. A novel circular RNA CDR1as has been recently reported to be a crucial regulator of human cancer. However, its biological role and mechanism in the GC growth are still far from clear. Methods: Small interfering RNAs (siRNAs), lentivirus or plasmid vectors were applied for gene manipulation. The CDR1as effects on the GC growth were evaluated in CCK8 and colony formation assays, a flow cytometry analysis and mouse xenograft tumor models. A bioinformatics analysis combined with RNA immunoprecipitation (RIP), RNA pull-down assays, dual-luciferase reporter gene assays, Western blot, reverse transcription–quantitative polymerase chain reaction (RT-qPCR) and functional rescue experiments were used to identify the CDR1as target miRNA, the downstream target gene and its interaction with human antigen R (HuR). Results: The CDR1as overexpression promoted the GC growth in vitro and in vivo and reduced the apoptotic rate of GC cells. Its knockdown inhibited the GC cell proliferation and viability and increased the cell apoptotic rate. Proliferation-related proteins PCNA and Cyclin D1 and apoptosis-related proteins Bax, Bcl-2, Caspase-3 and Caspase-9 were regulated. Mechanically, the cytoplasmic CDR1as acted as a miR-299-3p sponge to relieve its suppressive effects on the GC cell growth. Oncogenic TGIF1 was a miR-299-3p downstream target gene that mediated the promotive effects of CDR1as and regulated the PCNA and Bax levels. HuR interacted with CDR1as via the RRM2 domain and positively regulated the CDR1as level and its oncogenic role as well as downstream target TGIF1. Conclusions: CDR1as promotes the GC growth through the HuR/CDR1as/miR-299-3p/TGIF1 axis and could be used as a new therapeutic target for GC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. TGIF1 overexpression promotes glioma progression and worsens patient prognosis
- Author
-
Baoya Wang, Qiong Ma, Xuelin Wang, Kunshan Guo, Zhendong Liu, and Gang Li
- Subjects
biomarker ,glioma ,prognosis ,TGIF1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Transforming growth factor β‐induced factor homeobox 1 (TGIF1) reportedly promotes the pathological processes of various malignant tumors. However, few studies have investigated the role of TGIF1 in gliomas. We aimed to explore the relationship between TGIF1 expression and the clinical characteristics of patients with glioma, including their overall survival. A total of thousands transcriptome datapoints were downloaded from public databases to determine the correlations between TGIF1 and various clinicopathological features using the Wilcoxon or Kruskal–Wallis tests. The Kaplan–Meier and Cox statistical methods were used to explore the prognostic significance of TGIF1. Gene set enrichment analysis (GSEA) was used to indirectly identify the pathological mechanisms modulated by TGIF1, and compounds that inhibit its expression were determined using a connectivity map (CMap). TGIF1 was significantly overexpressed in gliomas and was correlated with unfavorable prognostic factors and shorter overall survival. Cox analysis confirmed that TGIF1 expression was a significant predictor of poor prognosis in patients with glioma. GSEA revealed that the signaling pathways associated with TGIF1 expression in glioma included extracellular matrix receptor‐ and cell cycle‐modulating proteins. CMap analysis showed that the small molecules scriptaid, torasemide, dexpropranolol, ipratropium bromide, and harmine were potential negative regulators of TGIF1. Finally, in vitro experiments demonstrated that knockdown of TGIF1 significantly inhibited the proliferation and invasion of glioma cell. Taken together, our study, which is the first to comprehensively analyze TGIF1 in gliomas, revealed it to be a novel oncogene in terms of its association with this disease. As such, TGIF1 may be a potential therapeutic target for individualized treatment of patients with glioma.
- Published
- 2022
- Full Text
- View/download PDF
7. Antagonizing microRNA‐19a/b augments PTH anabolic action and restores bone mass in osteoporosis in mice
- Author
-
Hanna Taipaleenmäki, Hiroaki Saito, Saskia Schröder, Miki Maeda, Ramona Mettler, Matthias Ring, Ewa Rollmann, Andreas Gasser, Carl Haasper, Thorsten Gehrke, Alexander Weiss, Steffen K Grimm, and Eric Hesse
- Subjects
anti‐miRNA ,osteoporosis ,parathyroid hormone ,Tgif1 ,treatment ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Postmenopausal bone loss often leads to osteoporosis and fragility fractures. Bone mass can be increased by the first 34 amino acids of human parathyroid hormone (PTH), parathyroid hormone‐related protein (PTHrP), or by a monoclonal antibody against sclerostin (Scl‐Ab). Here, we show that PTH and Scl‐Ab reduce the expression of microRNA‐19a and microRNA‐19b (miR‐19a/b) in bone. In bones from patients with lower bone mass and from osteoporotic mice, miR‐19a/b expression is elevated, suggesting an inhibitory function in bone remodeling. Indeed, antagonizing miR‐19a/b in vivo increased bone mass without overt cytotoxic effects. We identified TG‐interacting factor 1 (Tgif1) as the target of miR‐19a/b in osteoblasts and essential for the increase in bone mass following miR‐19a/b inhibition. Furthermore, antagonizing miR‐19a/b augments the gain in bone mass by PTH and restores bone loss in mouse models of osteoporosis in a dual mode of action by supporting bone formation and decreasing receptor activator of NF‐κB ligand (RANKL)‐dependent bone resorption. Thus, this study identifies novel mechanisms regulating bone remodeling, which opens opportunities for new therapeutic concepts to treat bone fragility.
- Published
- 2022
- Full Text
- View/download PDF
8. NH4Cl promotes apoptosis and inflammation in bovine mammary epithelial cells via the circ02771/miR-194b/TGIF1 axis
- Author
-
Zhi CHEN, Yu-sheng LIANG, Wei-cheng ZONG, Jia-he GUO, Jing-peng ZHOU, Yong-jiang MAO, De-jun JI, Pei-xin JIAO, LOOR Juan J, and Zhang-ping YANG
- Subjects
NH4Cl ,circ02771 ,miR-194b ,TGIF1 ,bovine mammary epithelial cells ,Agriculture (General) ,S1-972 - Abstract
Excess ammonia (NH3) in the circulation of dairy animals can reduce animal health and the quality of products for human consumption. To develop effective prevention and treatment methods, it is essential to examine the molecular mechanisms through which excess NH3 may affect the mammary gland. The present study used bovine mammary epithelial cells (BMECs) to evaluate the effects of exogenous NH4Cl on the abundance of circular RNAs (circRNAs) using high-throughput sequencing. Among the identified circRNAs, circ02771 was the most significantly upregulated by exogenous NH4Cl (P
- Published
- 2022
- Full Text
- View/download PDF
9. Ghrelin alleviates 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y cells
- Author
-
Xin He, Wei Yuan, Chun-Qing Yang, Lu Zhu, Fei Liu, Juan Feng, and Yi-Xue Xue
- Subjects
6-hydroxydopamine ,apoptosis ,ghrelin ,lincrna-p21 ,neuropeptide ,neurotoxicity ,parkinson’s disease ,stau1-mediated mrna decay ,tgif1 ,α-synuclein ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Ghrelin is a neuropeptide that has various physiological functions and has been demonstrated to be neuroprotective in a number of neurological disease models. However, the underlying mechanisms of ghrelin in Parkinson’s disease remain largely unexplored. The current study aimed to study the effects of ghrelin in a 6-hydroxydopamine (6-OHDA)-induced Parkinson’s disease model and evaluate the potential underlying mechanisms. In the present study, we treated an SH-SY5Y cell model with 6-OHDA, and observed that pretreatment with different concentrations of ghrelin (1, 10, and 100 nM) for 30 minutes relieved the neurotoxic effects of 6-OHDA, as revealed by Cell Counting Kit-8 and Annexin V/propidium iodide (PI) apoptosis assays. Reverse transcription quantitative polymerase chain reaction and western blot assay results demonstrated that 6-OHDA treatment upregulated α-synuclein and lincRNA-p21 and downregulated TG-interacting factor 1 (TGIF1), which was predicted as a potential transcription regulator of the gene encoding α-synuclein (SNCA). Ghrelin pretreatment was able to reverse the trends caused by 6-OHDA. The Annexin V/PI apoptosis assay results revealed that inhibiting either α-synuclein or lincRNA-p21 expression with small interfering RNA (siRNA) relieved 6-OHDA-induced cell apoptosis. Furthermore, inhibiting lincRNA-p21 also partially upregulated TGIF1. By retrieving information from a bioinformatics database and performing both double luciferase and RNA immunoprecipitation assays, we found that lincRNA-p21 and TGIF1 were able to form a double-stranded RNA-binding protein Staufen homolog 1 (STAU1) binding site and further activate the STAU1-mediated mRNA decay pathway. In addition, TGIF1 was able to transcriptionally regulate α-synuclein expression by binding to the promoter of SNCA. The Annexin V/PI apoptosis assay results showed that either knockdown of TGIF1 or overexpression of lincRNA-p21 notably abolished the neuroprotective effects of ghrelin against 6-OHDA-induced neurotoxicity. Collectively, these findings suggest that ghrelin exerts neuroprotective effects against 6-OHDA-induced neurotoxicity via the lincRNA-p21/TGIF1/α-synuclein pathway.
- Published
- 2022
- Full Text
- View/download PDF
10. Antagonizing microRNA‐19a/b augments PTH anabolic action and restores bone mass in osteoporosis in mice.
- Author
-
Taipaleenmäki, Hanna, Saito, Hiroaki, Schröder, Saskia, Maeda, Miki, Mettler, Ramona, Ring, Matthias, Rollmann, Ewa, Gasser, Andreas, Haasper, Carl, Gehrke, Thorsten, Weiss, Alexander, Grimm, Steffen K, and Hesse, Eric
- Abstract
Postmenopausal bone loss often leads to osteoporosis and fragility fractures. Bone mass can be increased by the first 34 amino acids of human parathyroid hormone (PTH), parathyroid hormone‐related protein (PTHrP), or by a monoclonal antibody against sclerostin (Scl‐Ab). Here, we show that PTH and Scl‐Ab reduce the expression of microRNA‐19a and microRNA‐19b (miR‐19a/b) in bone. In bones from patients with lower bone mass and from osteoporotic mice, miR‐19a/b expression is elevated, suggesting an inhibitory function in bone remodeling. Indeed, antagonizing miR‐19a/b in vivo increased bone mass without overt cytotoxic effects. We identified TG‐interacting factor 1 (Tgif1) as the target of miR‐19a/b in osteoblasts and essential for the increase in bone mass following miR‐19a/b inhibition. Furthermore, antagonizing miR‐19a/b augments the gain in bone mass by PTH and restores bone loss in mouse models of osteoporosis in a dual mode of action by supporting bone formation and decreasing receptor activator of NF‐κB ligand (RANKL)‐dependent bone resorption. Thus, this study identifies novel mechanisms regulating bone remodeling, which opens opportunities for new therapeutic concepts to treat bone fragility. Synopsis: Due to low bone mass, osteoporosis leads to fractures. Current drugs increase bone mass but have limitations. Thus, additional medicines are needed. Here we established anti‐sense oligonucleotide‐based microRNA‐19a/b inhibition to increase bone mass in osteoporosis by a novel dual mode of action. Post‐menopausal osteoporosis is caused by an increased osteoclast‐dependent bone resorption and a reduced osteoblast‐mediated bone formation often leading to fragility fractures that are associated with an increased morbidity and mortality.Patients with osteoporosis are frequently treated with anti‐resorptive drugs or anabolic agents such as PTH 1‐34 or an anti‐sclerostin antibody to increase bone mass.We uncovered that PTH 1‐34 and an anti‐sclerostin antibody reduced the expression of microRNA‐19a and microRNA‐19b (miR‐19a/b) in bone.Human bones with lower bone mass and bones from osteoporotic mice displayed an increased miR‐19a/b expression.Anti‐sense oligonucleotide‐mediated miR‐19a/b inhibition augmented the PTH 1‐34‐induced gain in bone mass and reconstituted low bone mass in osteoporosis in mice by supporting bone formation and decreasing bone resorption. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. PSPC1 is a new contextual determinant of aberrant subcellular translocation of oncogenes in tumor progression
- Author
-
Yaw-Dong Lang and Yuh-Shan Jou
- Subjects
PSPC1 ,Nucleocytoplasmic shuttling ,Oncogenic switch ,Selective inhibitor of nucleocytoplasmic shuttling ,TGIF1 ,NPM ,Medicine - Abstract
Abstract Dysregulation of nucleocytoplasmic shuttling is commonly observed in cancers and emerging as a cancer hallmark for the development of anticancer therapeutic strategies. Despite its severe adverse effects, selinexor, a selective first-in-class inhibitor of the common nuclear export receptor XPO1, was developed to target nucleocytoplasmic protein shuttling and received accelerated FDA approval in 2019 in combination with dexamethasone as a fifth-line therapeutic option for adults with relapsed refractory multiple myeloma (RRMM). To explore innovative targets in nucleocytoplasmic shuttling, we propose that the aberrant contextual determinants of nucleocytoplasmic shuttling, such as PSPC1 (Paraspeckle component 1), TGIF1 (TGF-β Induced Factor Homeobox 1), NPM1 (Nucleophosmin), Mortalin and EBP50, that modulate shuttling (or cargo) proteins with opposite tumorigenic functions in different subcellular locations could be theranostic targets for developing anticancer strategies. For instance, PSPC1 was recently shown to be the contextual determinant of the TGF-β prometastatic switch and PTK6/β-catenin reciprocal oncogenic nucleocytoplasmic shuttling during hepatocellular carcinoma (HCC) progression. The innovative nucleocytoplasmic shuttling inhibitor PSPC1 C-terminal 131 polypeptide (PSPC1-CT131), which was developed to target both the shuttling determinant PSPC1 and the shuttling protein PTK6, maintained their tumor-suppressive characteristics and exhibited synergistic effects on tumor suppression in HCC cells and mouse models. In summary, targeting the contextual determinants of nucleocytoplasmic shuttling with cargo proteins having opposite tumorigenic functions in different subcellular locations could be an innovative strategy for developing new therapeutic biomarkers and agents to improve cancer therapy.
- Published
- 2021
- Full Text
- View/download PDF
12. TGIF1 Knockdown Inhibits the Proliferation and Invasion of Gastric Cancer via AKT Signaling Pathway
- Author
-
Zhang J, Zhang F, Fan J, and Feng B
- Subjects
tgif1 ,gastric cancer ,akt pathway ,apoptosis ,proliferation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Jing Zhang,1 Feiyan Zhang,2 Jiye Fan,3,4 Bin Feng5 1Pharmacy Department, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei 050026, People’s Republic of China; 2Department of Outpatient Operating Room, Heze Municipal Hospital, Heze City, Shandong Province, 274000, People’s Republic of China; 3Department of Pharmaceutical Engineering, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei 050026, People’s Republic of China; 4College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, People’s Republic of China; 5Department of Gastrointestinal Surgery, Heze Municipal Hospital, Heze City, 274000 Shandong Province, People’s Republic of ChinaCorrespondence: Bin FengDepartment of Gastrointestinal Surgery, Heze Municipal Hospital, No. 2888, Caozhou Road, Mudan District, Heze City 274000, Shandong Province, People’s Republic of ChinaEmail 13020511011@163.comIntroduction: Gastric cancer is a kind of cancer with high mortality. TGIF1, as a transcription inhibitor, can inhibit the transcription of specific genes. The purpose of this study was to investigate the role of TGIF1 in gastric cancer by knocking down TGIF1.Methods: The expression of TGIF1 was detected by qPCR and Western blotting; CCK8 assay, colony formation assay, transwell, and wound-healing assay were used to evaluate the proliferation, migration, and invasion of gastric cancer cells; cell apoptosis was analyzed by flow cytometry and Hoechst-PI double staining; cell cycle was detected by flow cytometry. Gelatinase experiment was performed to detect the expression level of MMP-2; apoptosis related proteins and AKT singling pathway were assessed by Western blotting.Results: Knockdown of TGIF1 inhibited the proliferation, migration, and invasion of gastric cancer cells and promoted apoptosis. TGIF1 knockdown down-regulated the expression levels of MMP-2, Bcl2, CyclinD1, and p-Akt, and up-regulated the expression levels of Bax and Caspase3. These data suggested that knockdown of TGIF1 inhibited the development of gastric cancer via AKT signaling pathway.Conclusion: TGIF1 knockdown inhibited the proliferation, migration, and invasion and promoted apoptosis of gastric cancer cells via the AKT signaling pathway, suggesting that TGIF1 is considered a potential inhibitor in gastric cancer.Keywords: TGIF1, gastric cancer, AKT pathway, apoptosis, proliferation
- Published
- 2021
13. The Chinese Medicine, Shezhi Huangling Decoction, Inhibits the Growth and Metastasis of Glioma Cells via the Regulation of miR-1298-5p/TGIF1 Axis
- Author
-
Liu X, Ju J, Liu Q, Zhu Z, and Liu C
- Subjects
glioma ,chinese medicine ,shezhi huangling decoction ,mir-1298-5p ,tgif1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Xiaoqian Liu,1 Jianfeng Ju,1 Qun Liu,1 Zongmin Zhu,1 Chunxia Liu2 1Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People’s Republic of China; 2Department of Pharmacy, Caoxian People’s Hospital of Heze City, Heze 274400, Shandong, People’s Republic of ChinaCorrespondence: Jianfeng JuDepartment of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jingshi Road, Jinan 250011, Shandong, People’s Republic of ChinaEmail yxbgongyou@163.comPurpose: In recent years, traditional Chinese medicine has achieved good results in treating gliomas. This research aimed to reveal the effect of Shezhi Huangling decoction (SD) on glioma cell process.Methods: U87 and U251 cells were treated with different concentrations (10, 30 and 50 μg/mL) of SD or transfected with miR-1298-5p mimic, inhibitor and siRNA targeting TGIF1. Cell proliferation, migration, invasion and apoptosis were detected. The expression of miR-1298-5p was measured by qRT-PCR, while TGIF1 expression was examined by immunohistochemical analysis and Western blot.Results: SD treatment inhibited the proliferation, migration and invasion of glioma cells and induced the apoptosis. In addition, SD treatment induced the expression of miR-1298-5p in glioma cells. The low expression of miR-1298-5p was examined in glioma tissues and was significantly related to the high histological grade of glioma patients and predicted a poor prognosis. MiR-1298-5p directly targeted the 3ʹ-UTR of transforming growth factor β induced factor 1 (TGIF1) and reduced TGIF1 protein expression. MiR-1298-5p restricted the proliferation, migration and invasion of glioma cells and induced cell apoptosis by targeting TGIF1.Conclusion: Our data reveal that SD acts as a cancer-inhibiting agent in glioma via miR-1298-5p/TGIF1 axis, suggesting a potential therapeutic application of SD in glioma.Keywords: glioma, Chinese medicine, Shezhi Huangling decoction, miR-1298-5p, TGIF1
- Published
- 2020
14. Breast cancer bone metastases are attenuated in a Tgif1-deficient bone microenvironment
- Author
-
Marie-Therese Haider, Hiroaki Saito, Jennifer Zarrer, Kevin Uzhunnumpuram, Sankari Nagarajan, Vijayalakshmi Kari, Michael Horn-Glander, Stefan Werner, Eric Hesse, and Hanna Taipaleenmäki
- Subjects
Breast cancer ,Bone metastases ,Osteoblasts ,Tgif1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Osteoclast activation is a hallmark of breast cancer-induced bone disease while little is known about the role of osteoblasts in this process. Recently, we identified the homeodomain protein TG-interacting factor-1 (Tgif1) as a crucial regulator of osteoblast function. In this study, we demonstrate that lack of Tgif1 also restricts the progression of breast cancer bone metastases. Methods Transwell migration assays were used to investigate the osteoblast-breast cancer cell interaction in vitro. Molecular analyses included RNA sequencing, immunoblotting, and qRT-PCR. To determine the role of Tgif1 in metastatic bone disease, 4T1 breast cancer cells were injected intracardially into mice with a germ line deletion of Tgif1 (Tgif1 −/− ) or control littermates (Tgif1 +/+ ). Progression of bone metastases and alterations in the bone microenvironment were assessed using bioluminescence imaging, immunofluorescence staining, confocal microscopy, and histomorphometry. Results Medium conditioned by osteoblasts stimulated breast cancer cell migration, indicating a potential role of osteoblasts during bone metastasis progression. Tgif1 expression was strongly increased in osteoblasts upon stimulation by breast cancer cells, demonstrating the implication of Tgif1 in the osteoblast-breast cancer cell interaction. Indeed, conditioned medium from osteoblasts of Tgif1 −/− mice failed to induce breast cancer cell migration compared to control, suggesting that Tgif1 in osteoblasts augments cancer cell motility. Semaphorin 3E (Sema3E), which is abundantly secreted by Tgif1 −/− osteoblasts, dose-dependently reduced breast cancer cell migration while silencing of Sema3E expression in Tgif1 −/− osteoblasts partially restored the impaired migration. In vivo, we observed a decreased number of breast cancer bone metastases in Tgif1 −/− mice compared to control littermates. Consistently, the presence of single breast cancer cells or micro-metastases in the tibiae was reduced in Tgif1 −/− mice. Breast cancer cells localized in close proximity to Endomucin-positive vascular cells as well as to osteoblasts. Although Tgif1 deficiency did not affect the bone marrow vasculature, the number and activity of osteoblasts were reduced compared to control. This suggests that the protective effect on bone metastases might be mediated by osteoblasts rather than by the bone marrow vasculature. Conclusion We propose that the lack of Tgif1 in osteoblasts increases Sema3E expression and attenuates breast cancer cell migration as well as metastases formation.
- Published
- 2020
- Full Text
- View/download PDF
15. Inhibition of ERN1 affects the expression of TGIF1 and other homeobox gene expressions in U87MG glioblastoma cells.
- Author
-
Minchenko, Oleksandr H., Khita, Olena O., Krasnytska, Daria A., Viletska, Yuliia M., Rudnytska, Olha V., Hnatiuk, Oksana S., and Minchenko, Dmytro O.
- Subjects
- *
GENE expression , *HOMEOBOX genes , *GENETIC regulation , *GLIOBLASTOMA multiforme , *REGULATOR genes , *PROTEIN expression - Abstract
The ERN1 (endoplasmic reticulum to nucleus signaling 1) pathway plays an important role in the regulation of gene expression in glioblastoma, but molecular mechanism has not yet been fully elucidated. The aim of this study was to evaluate the relative relevance of ERN1 activity as a kinase in comparison to its endoribonuclease activity in the regulation of homeobox gene expression. Two sublines of U87MG glioblastoma cells with different ways of ERN1 inhibition were used: dnERN1 (overexpressed transgene without protein kinase and endoribonuclease) and dnrERN1 (overexpressed transgene with mutation in endoribonuclease). ERN1 suppression was also done using siRNA for ERN1. Silencing of XBP1 mRNA by specific siRNA was used for suppression of ERN1 endoribonuclease function mediated by XBP1s. The expression levels of homeobox genes and microRNAs were evaluated by qPCR. The expression of TGIF1 and ZEB2 genes was downregulated in both types of glioblastoma cells with inhibition of ERN1 showing the ERN1 endoribonuclease-dependent mechanism of their regulation. However, the expression of PBX3 and PRPRX1 genes did not change significantly in dnrERN1 glioblastoma cells but was upregulated in dnERN1 cells indicating the dependence of these gene expressions on the ERN1 protein kinase. At the same time, the changes in PAX6 and PBXIP1 gene expressions introduced in glioblastoma cells by dnrERN1 and dnERN1 were different in direction and magnitude indicating the interaction of ERN1 protein kinase and endoribonuclease activities in regulation of these gene expressions. The impact of ERN1 and XBP1 silencing on the expression of studied homeobox genes is similar to that observed in dnERN1 and dnrERN1 glioblastoma cells, correspondingly. The expression of TGIF1 and other homeobox genes is dependent on the ern1 signaling pathways by diverse mechanisms because inhibition of ERN1 endoribonuclease and both ERN1 enzymatic activities had dissimilar impacts on the expression of most studied genes showing that ERN1 protein kinase plays an important role in controlling homeobox gene expression associated with glioblastoma cell invasion. [Display omitted] • Inhibition of ERN1 suppresses TGIF1 and ZEB2 expressions via ERN1 endoribonuclease. • ERN1 knockdown affects PBX3 and PRRX1 gene expressions through ERN1 protein kinase. • The changes in PAX6 gene expression are mediated by ERN1 endoribonuclease and kinase. • ERN1 protein kinase pathway is an important regulator of homeobox genes expressions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. MircoRNA-25-3p in skin precursor cell-induced Schwann cell-derived extracellular vesicles promotes axon regeneration by targeting Tgif1.
- Author
-
Cong, Meng, Li, Jiyu, Wang, Lijuan, Liu, Chang, Zheng, Mengru, Zhou, Qiang, Du, Mingzhi, Ye, Xinli, Feng, Min, Ye, Yujiao, Zhang, Shuyu, Xu, Wenqing, Lu, Yi, Wang, Cheng, Xia, Yingjie, Xie, Huimin, Zhang, Yide, He, Qianru, Gong, Leilei, and Gu, Yun
- Subjects
- *
EXTRACELLULAR vesicles , *NERVOUS system regeneration , *AXONS , *DORSAL root ganglia , *GENE expression - Abstract
Nerve injury often leads to severe dysfunction because of the lack of axon regeneration in adult mammal. Intriguingly a series of extracellular vesicles (EVs) have the obvious ability to accelerate the nerve repair. However, the detailed molecular mechanisms to describe that EVs switch neuron from a transmitter to a regenerative state have not been elucidated. This study elucidated the microRNA (miRNA) expression profiles of two types of EVs that promote nerve regeneration. The functions of these miRNAs were screened in vitro. Among the 12 overlapping miRNAs, miR-25-3p was selected for further analysis as it markedly promoted axon regeneration both in vivo and in vitro. Furthermore, knockdown experiments confirmed that PTEN and Klf4 , which are the major inhibitors of axon regeneration, were the direct targets of miR-25-3p in dorsal root ganglion (DRG) neurons. The utilization of luciferase reporter assays and functional tests provided evidence that miR-25-3p enhances axon regeneration by targeting Tgif1. Additionally, miR-25-3p upregulated the phosphorylation of Erk. Furthermore, Rapamycin modulated the expression of miR-25-3p in DRG neurons. Finally, the pro-axon regeneration effects of EVs were confirmed by overexpressing miR-25-3p and Tgif1 knockdown in the optic nerve crush model. Thus, the enrichment of miR-25-3p in EVs suggests that it regulates axon regeneration, proving a potential cell-free treatment strategy for nerve injury. • miR-25-3p promotes axon regeneration by targeting Tgif1. • miR-25-3p up-regulate p-Erk expression level in DRG neuron. • SKP-SC-EVs containing miR-25-3p was a potential treatment strategy for nerve injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. TG-interacting factor 1 regulates mitotic clonal expansion during adipocyte differentiation.
- Author
-
Chang, Yu-Hao, Tseng, Yu-Hua, Wang, Ju-Ming, Tsai, Yau-Sheng, and Huang, Huei-Sheng
- Subjects
- *
ADIPOGENESIS , *GENE expression , *STAINS & staining (Microscopy) , *FAT cells , *PEROXISOME proliferator-activated receptors , *CELL cycle - Abstract
Obesity is one of the significant health challenges in the world and is highly associated with abnormal adipogenesis. TG-interacting factor 1 (TGIF1) is essential for differentiating murine adipocytes and human adipose tissue-derived stem cells. However, the mode of action needs to be better elucidated. To investigate the roles of TGIF1 in differentiation in-depth, CRISPR/Cas9 knockout technology was performed to generate TGIF1-silenced preadipocytes. The absence of TGIF1 in 3 T3-F442A preadipocytes abolished lipid accumulation throughout the differentiation using Oil Red O staining. Conversely, we established 3 T3-F442A preadipocytes stably expressing TGIF1 and doxycycline-inducible TGIF1 in TGIF1-silenced 3 T3-F442A preadipocytes. Remarkably, the induction of TGIF1 by doxycycline during the initial differentiation phase successfully promoted lipid accumulation in TGIF1-silenced 3 T3-F442A cells. We further explored the mechanisms of TGIF1 in early differentiation. We demonstrated that TGIF1 promoted the mitotic clonal expansion via upregulation of CCAAT/enhancer-binding proteins β expression, interruption with peroxisome proliferators activated receptor γ downstream regulation, and inhibition of p27kip1 expression. In conclusion, we strengthen the pivotal roles of TGIF1 in early differentiation, which might contribute to resolving obesity-associated metabolic syndromes. • We demonstrated that TGIF1 was essential for early differentiation by using several advanced techniques in preadipocytes, including enforced overexpression, CRISPR/Cas9 knockout, and Tet-inducible gene expression. • TGIF1 contributes to the early differentiation through novel mechanisms, including upregulation of C/EBP β expression, interference in PPAR γ downstream regulation, and inhibition of p27kip1 expression, thereby promoting mitotic clonal expansion in the early differentiation. • We conclude that p27kip1, a PPAR γ -regulated gene, is suppressed by TGIF1 in early differentiation until the endogenous PPAR γ ligands production which is tightly linked to mitotic clonal expansion to strengthen the binding ability of PPAR γ to the promoter of p27kip1 and stop the cell cycle to proceed to differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. TGIF1-Twist1 axis in pancreatic ductal adenocarcinoma
- Author
-
Mohammed S. Razzaque and Azeddine Atfi
- Subjects
TGIF1 ,TGF-β ,Twist1 ,Tumor suppressor ,Pancreatic ductal adenocarcinoma ,Biotechnology ,TP248.13-248.65 - Abstract
TG-interacting factor 1 (TGIF1) exerts inhibitory effects on transforming growth factor-beta (TGF-β) signaling by suppressing Smad signaling pathway at multiple levels. TGIF1 activity is important for normal embryogenesis and organogenesis, yet its dysregulation can culminate in tumorigenesis. For instance, increased expression of TGIF1 correlates with poor prognosis in triple-negative breast cancer patients, and enforced expression of TGIF1 facilitates Wnt-driven mammary tumorigenesis, suggesting that TGIF1 might function as an oncoprotein. Quite surprisingly, TGIF1 has recently been shown to function as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC), possibly owing to its ability to antagonize the pro-malignant transcription factor Twist1. In this article, we will briefly elaborate on the biological and clinical significance of the unique tumor-suppressive function of TGIF1 and its functional interaction with Twist1 in the context of PDAC pathogenesis and progression.
- Published
- 2020
- Full Text
- View/download PDF
19. Loss of the transcriptional repressor TGIF1 results in enhanced Kras-driven development of pancreatic cancer
- Author
-
Ching-Chieh Weng, Mei-Jen Hsieh, Chia-Chen Wu, Yu-Chun Lin, Yan-Shen Shan, Wen-Chun Hung, Li-Tzong Chen, and Kuang-Hung Cheng
- Subjects
Pancreatic cancer ,TGIF1 ,Has2 ,PD-L1 ,M2 macrophage ,Metastatic PDAC model ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The TG-interacting factor 1 (TGIF1) gene, which encodes a nuclear transcriptional corepressor of the TGFβ1/Smad signaling pathway, has been implicated in the pathogenesis of various types of human cancer; however, its role in pancreatic ductal adenocarcinoma (PDAC) has yet to be elucidated. Methods The expression of TGIF1 in human and murine PDAC specimens were detected by IHC analysis. The functions of TGIF1 in in vivo PDAC growth, dissemination, and metastasis were assessed using conditional inactivation of TGIF1 in well-established autochthonous mouse models of PDAC. Primary cells from TGIF1 null or wild type PDAC mice were examined by assays for cell proliferation, migration, invasion, soft agar and xenograft tumorigenesis. Gene expression profiling, pathway analyses, epigenetic changes associated with TGIF1 loss, and in vitro and in vivo effects of 4-MU were assessed. Results Conditional deletion of TGIF1 in the mouse pancreas had no discernible effect on pancreatic development or physiology. Notably, TGIF1 loss induced KrasG12D-driven PDAC models exhibited shorter latency and greater propensity for distant metastases. Deciphering the molecular mechanisms highlighted the TGIF1 loss-induced activation of the hyaluronan synthase 2 (HAS2)-CD44 signaling pathway and upregulation of the immune checkpoint regulator PD-L1 to facilitate the epithelial–mesenchymal transition (EMT) and tumor immune suppression. We also founded that TGIF1 might function as an epigenetic regulator and response for aberrant EMT gene expression during PDAC progression. Conclusions Our results imply that targeting the HAS2 pathway in TGIF1 loss of PDAC could be a promising therapeutic strategy for improving the clinical efficacy against PDAC metastasis.
- Published
- 2019
- Full Text
- View/download PDF
20. The homeobox gene TGIF1 is required for chicken ovarian cortical development and generation of the juxtacortical medulla.
- Author
-
Estermann, Martin Andres, Hirst, Claire Elizabeth, Major, Andrew Thomas, and Smith, Craig Allen
- Subjects
- *
HOMEOBOX genes , *SERTOLI cells , *SEX differentiation (Embryology) , *INTERSTITIAL cells , *CELL populations , *GONADS , *CHICKEN embryos - Abstract
During early embryogenesis in amniotic vertebrates, the gonads differentiate into either ovaries or testes. The first cell lineage to differentiate gives rise to the supporting cells: Sertoli cells in males and pre-granulosa cells in females. These key cell types direct the differentiation of the other cell types in the gonad, including steroidogenic cells. The gonadal surface epithelium and the interstitial cell populations are less well studied, and little is known about their sexual differentiation programs. Here, we show the requirement of the homeobox transcription factor gene TGIF1 for ovarian development in the chicken embryo. TGIF1 is expressed in the two principal ovarian somatic cell populations: the cortex and the pre-granulosa cells of the medulla. TGIF1 expression is associated with an ovarian phenotype in estrogen-mediated sex reversal experiments. Targeted misexpression and gene knockdown indicate that TGIF1 is required, but not sufficient, for proper ovarian cortex formation. In addition, TGIF1 is identified as the first known regulator of juxtacortical medulla development. These findings provide new insights into chicken ovarian differentiation and development, specifically cortical and juxtacortical medulla formation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
21. PSPC1 is a new contextual determinant of aberrant subcellular translocation of oncogenes in tumor progression.
- Author
-
Lang, Yaw-Dong and Jou, Yuh-Shan
- Subjects
NUCLEOCYTOPLASMIC interactions ,CANCER invasiveness ,ONCOGENES ,LABORATORY mice ,MULTIPLE myeloma - Abstract
Dysregulation of nucleocytoplasmic shuttling is commonly observed in cancers and emerging as a cancer hallmark for the development of anticancer therapeutic strategies. Despite its severe adverse effects, selinexor, a selective first-in-class inhibitor of the common nuclear export receptor XPO1, was developed to target nucleocytoplasmic protein shuttling and received accelerated FDA approval in 2019 in combination with dexamethasone as a fifth-line therapeutic option for adults with relapsed refractory multiple myeloma (RRMM). To explore innovative targets in nucleocytoplasmic shuttling, we propose that the aberrant contextual determinants of nucleocytoplasmic shuttling, such as PSPC1 (Paraspeckle component 1), TGIF1 (TGF-β Induced Factor Homeobox 1), NPM1 (Nucleophosmin), Mortalin and EBP50, that modulate shuttling (or cargo) proteins with opposite tumorigenic functions in different subcellular locations could be theranostic targets for developing anticancer strategies. For instance, PSPC1 was recently shown to be the contextual determinant of the TGF-β prometastatic switch and PTK6/β-catenin reciprocal oncogenic nucleocytoplasmic shuttling during hepatocellular carcinoma (HCC) progression. The innovative nucleocytoplasmic shuttling inhibitor PSPC1 C-terminal 131 polypeptide (PSPC1-CT131), which was developed to target both the shuttling determinant PSPC1 and the shuttling protein PTK6, maintained their tumor-suppressive characteristics and exhibited synergistic effects on tumor suppression in HCC cells and mouse models. In summary, targeting the contextual determinants of nucleocytoplasmic shuttling with cargo proteins having opposite tumorigenic functions in different subcellular locations could be an innovative strategy for developing new therapeutic biomarkers and agents to improve cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
22. TGIF1 and SF1 polymorphisms are associated with litter size in Small Tail Han sheep.
- Author
-
Zhang, Zhuangbiao, He, Xiaoyun, Liu, Qiuyue, Tang, Jishun, Di, Ran, and Chu, Mingxing
- Subjects
- *
SHEEP , *SHEEP breeds , *ANIMAL litters , *GENETIC markers , *TAILS , *EWES , *SIZE - Abstract
TGF‐β induced factor homeobox 1 (TGIF1) and splicing factor 1 (SF1) are important for mammalian reproduction; however, the effects of these genes on litter size in sheep remain unexplored. In this study, we genotyped 768 ewes from seven sheep breeds at two loci: g.37871539C>T, a synonymous mutation of TGIF1; and g.42314637T>C, a 3′UTR variant of SF1. Our analysis of polymorphism revealed only two genotypes at locus g.37871539C>T in TGIF1, with most sheep populations being moderately polymorphic (0.25 < PIC < 0.5) at this site. In contrast, most breeds exhibited low polymorphism (PIC ≤0.25) at the SF1 locus g.42314637T>C. The association analysis revealed that a synonymous mutation at g.37871539C>T in TGIF1 was highly associated with litter size in Small Tail Han sheep, in which it causes a significant decrease in litter size. Conversely, while the SF1 3′UTR variant g.42314637T>C was also highly associated with litter size in sheep, it causes a significant increase in the number of litter size. Combined, these data provide valuable information regarding candidate genetic markers for sheep breeding programs. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
23. Effect of TGIF1 expression on epithelial cadherin and Twist1 protein expression in breast cancer cells.
- Author
-
Ying Liu, Ru-Li Qiao, Li Shang, Feng-Jun Zhang, and Qing-Li Zhao
- Subjects
TRANSFORMING growth factors-beta ,BREAST cancer ,CANCER cells ,CADHERINS ,METASTASIS - Abstract
Objective: To observe the effect of the expression of transforming growth factor ß-homologous inducible factor 1 (TGIF1) on the expression of epithelial cadherin (E-cadherin) and human Twist-related protein 1 (Twist1) in breast cancer cells. Methods: Total of twenty-four 6-weekold female SPF Balb/c mice were randomly divided into a control group, a TGIF1-silencing group, a TGIF1-normal group, and a TGIF1-overexpression group. In the TGIF1-silencing group, 4T1 breast cancer cells were interfered by lentivirus shRNA (H) lentiviral particles (sc- 36659-v) to construct a breast cancer model. TGIF1-normal group used breast cancer cells (4T1) to construct a mouse model of breast cancer. And the TGIF1-overexpression group used 4T1 breast cancer cells with TGIF1 overexpression to establish a mouse model of breast cancer. Determination of TGIF1, E-cadherin and Twist1 protein levels in breast tumor tissue of mice in each group. Results: The tumor volume of mice in the TGIF1-overexpression group was significantly larger than that in the TGIF1-normal group and the TGIF1-silencing group, and the differences between the groups were statistically significant (P <0.05).The expression levels of TGIF1 and Twist1 protein in TGIF1-normal group, TGIF1-silencing group, and TGIF1- overexpression group were significantly higher than those in control group, and E-cadherin was significantly lower than that in control group. The differences between groups were statistically significant (P <0.05).The expression level of TGIF1 and Twist1 protein in TGIF1- silencing group was significantly lower than that in TGIF1-normal group, and E-cadherin was significantly higher than that in TGIF1-normal group (P < 0.05).The expression levels of TGIF1 and Twist1 proteins in the TGIF1-overexpression group were significantly higher than those in the TGIF1-normal group, and E-cadherin was significantly lower than that in the TGIF1-normal group. The differences between the groups were statistically significant (P <0.05). Pearson correlation analysis showed that the expression level of TGIF1 in breast cancer tissue was significantly negatively correlated with the expression level of E-cadherin protein, and was significantly positively correlated with the level of Twist1 protein (P <0.05). Conclusion: TGIF1 can affect the metastasis and invasion of breast cancer by regulating E-cadherin and Twist1 to interfere with the EMT pathway, which deserves further study. [ABSTRACT FROM AUTHOR]
- Published
- 2020
24. Breast cancer bone metastases are attenuated in a Tgif1-deficient bone microenvironment.
- Author
-
Haider, Marie-Therese, Saito, Hiroaki, Zarrer, Jennifer, Uzhunnumpuram, Kevin, Nagarajan, Sankari, Kari, Vijayalakshmi, Horn-Glander, Michael, Werner, Stefan, Hesse, Eric, and Taipaleenmäki, Hanna
- Subjects
METASTATIC breast cancer ,CANCER cell motility ,BONES ,BONE metastasis ,CANCER cell migration ,BONE marrow ,CALCIFICATIONS of the breast ,BONE metabolism ,BREAST tumor prevention ,PROTEIN metabolism ,OSTEOBLAST metabolism ,ANIMAL experimentation ,BIOLOGICAL models ,BONE tumors ,BREAST tumors ,CELL differentiation ,CELL lines ,CELL physiology ,COMPARATIVE studies ,RESEARCH methodology ,MEDICAL cooperation ,MICE ,PROTEINS ,RESEARCH ,EVALUATION research ,OSTEOBLASTS ,CHEMICAL inhibitors - Abstract
Background: Osteoclast activation is a hallmark of breast cancer-induced bone disease while little is known about the role of osteoblasts in this process. Recently, we identified the homeodomain protein TG-interacting factor-1 (Tgif1) as a crucial regulator of osteoblast function. In this study, we demonstrate that lack of Tgif1 also restricts the progression of breast cancer bone metastases.Methods: Transwell migration assays were used to investigate the osteoblast-breast cancer cell interaction in vitro. Molecular analyses included RNA sequencing, immunoblotting, and qRT-PCR. To determine the role of Tgif1 in metastatic bone disease, 4T1 breast cancer cells were injected intracardially into mice with a germ line deletion of Tgif1 (Tgif1-/-) or control littermates (Tgif1+/+). Progression of bone metastases and alterations in the bone microenvironment were assessed using bioluminescence imaging, immunofluorescence staining, confocal microscopy, and histomorphometry.Results: Medium conditioned by osteoblasts stimulated breast cancer cell migration, indicating a potential role of osteoblasts during bone metastasis progression. Tgif1 expression was strongly increased in osteoblasts upon stimulation by breast cancer cells, demonstrating the implication of Tgif1 in the osteoblast-breast cancer cell interaction. Indeed, conditioned medium from osteoblasts of Tgif1-/- mice failed to induce breast cancer cell migration compared to control, suggesting that Tgif1 in osteoblasts augments cancer cell motility. Semaphorin 3E (Sema3E), which is abundantly secreted by Tgif1-/- osteoblasts, dose-dependently reduced breast cancer cell migration while silencing of Sema3E expression in Tgif1-/- osteoblasts partially restored the impaired migration. In vivo, we observed a decreased number of breast cancer bone metastases in Tgif1-/- mice compared to control littermates. Consistently, the presence of single breast cancer cells or micro-metastases in the tibiae was reduced in Tgif1-/- mice. Breast cancer cells localized in close proximity to Endomucin-positive vascular cells as well as to osteoblasts. Although Tgif1 deficiency did not affect the bone marrow vasculature, the number and activity of osteoblasts were reduced compared to control. This suggests that the protective effect on bone metastases might be mediated by osteoblasts rather than by the bone marrow vasculature.Conclusion: We propose that the lack of Tgif1 in osteoblasts increases Sema3E expression and attenuates breast cancer cell migration as well as metastases formation. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
25. XTP8 stimulates migration and invasion of gastric carcinoma through interacting with TGIF1.
- Author
-
K. JIA, Q.-H. WEN, X. ZHAO, J.-M. CHENG, L. CHENG, and M. XI
- Abstract
OBJECTIVE: To determine expression characteristics of XTP8 and TGIF1 in gastric carcinoma (GC), and the potential roles of XTP8/TGIF1 axis in influencing the progression of GC. MATERIALS AND METHODS: The expression levels of XTP8 and TGIF1 in GC tissues and cells were detected. Their functions in prognosis in GC patients were evaluated by the Kaplan-Meier method. The correlation between the XTP8 level and the pathological indexes of the GC patients were analyzed. The changes in the proliferation, migration, and invasion capacities of MKN-45 and SGC-7901 cells affected by XTP8 and TGIF1 were assessed. The interaction between XTP8 and TGIF1 was determined through Dual-Luciferase reporter gene assay and rescue experiments. RESULTS: XTP8 was upregulated in GC tissues and cells. XTP8 level was positively correlated with lymphatic and distant metastasis, as well as poor prognosis of GC patients. The silence of XTP8 attenuated proliferation, migration, and invasion capacities of MKN-45 and SGC-7901 cells. TGIF1 was the downstream gene binding to XTP8, which was downregulated in GC, and XTP8 negatively regulated the TGIF1 level in GC tissues. Importantly, the knockdown of TGIF1 could abolish the regulatory effect of XTP8 on GC cell behaviors. CONCLUSIONS: XTP8 is upregulated in GC and is closely linked to lymphatic metastasis, distant metastasis, and poor prognosis of GC patients. Besides, it accelerates the malignant progression via negatively regulating TGIF1. [ABSTRACT FROM AUTHOR]
- Published
- 2020
26. Loss of the transcriptional repressor TGIF1 results in enhanced Kras-driven development of pancreatic cancer.
- Author
-
Weng, Ching-Chieh, Hsieh, Mei-Jen, Wu, Chia-Chen, Lin, Yu-Chun, Shan, Yan-Shen, Hung, Wen-Chun, Chen, Li-Tzong, and Cheng, Kuang-Hung
- Subjects
RAS oncogenes ,PANCREATIC cancer ,GENE expression profiling ,PANCREATIC intraepithelial neoplasia ,PANCREATIC physiology ,IMMUNOSUPPRESSION ,GENE expression - Abstract
Background: The TG-interacting factor 1 (TGIF1) gene, which encodes a nuclear transcriptional corepressor of the TGFβ1/Smad signaling pathway, has been implicated in the pathogenesis of various types of human cancer; however, its role in pancreatic ductal adenocarcinoma (PDAC) has yet to be elucidated. Methods: The expression of TGIF1 in human and murine PDAC specimens were detected by IHC analysis. The functions of TGIF1 in in vivo PDAC growth, dissemination, and metastasis were assessed using conditional inactivation of TGIF1 in well-established autochthonous mouse models of PDAC. Primary cells from TGIF1 null or wild type PDAC mice were examined by assays for cell proliferation, migration, invasion, soft agar and xenograft tumorigenesis. Gene expression profiling, pathway analyses, epigenetic changes associated with TGIF1 loss, and in vitro and in vivo effects of 4-MU were assessed. Results: Conditional deletion of TGIF1 in the mouse pancreas had no discernible effect on pancreatic development or physiology. Notably, TGIF1 loss induced KrasG12D-driven PDAC models exhibited shorter latency and greater propensity for distant metastases. Deciphering the molecular mechanisms highlighted the TGIF1 loss-induced activation of the hyaluronan synthase 2 (HAS2)-CD44 signaling pathway and upregulation of the immune checkpoint regulator PD-L1 to facilitate the epithelial–mesenchymal transition (EMT) and tumor immune suppression. We also founded that TGIF1 might function as an epigenetic regulator and response for aberrant EMT gene expression during PDAC progression. Conclusions: Our results imply that targeting the HAS2 pathway in TGIF1 loss of PDAC could be a promising therapeutic strategy for improving the clinical efficacy against PDAC metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. Ghrelin alleviates 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y cells
- Author
-
Chunqing Yang, Fei Liu, Wei Yuan, Yixue Xue, Lu Zhu, Juan Feng, and Xin He
- Subjects
Small interfering RNA ,SH-SY5Y ,Parkinson's disease ,6-hydroxydopamine ,chemistry.chemical_compound ,α-synuclein ,Developmental Neuroscience ,Annexin ,neurotoxicity ,TGIF1 ,Propidium iodide ,lincRNA-p21 ,RC346-429 ,neuropeptide ,Gene knockdown ,STAU1-mediated mRNA decay ,apoptosis ,Cell biology ,Real-time polymerase chain reaction ,chemistry ,nervous system ,Apoptosis ,ghrelin ,Ghrelin ,Neurology. Diseases of the nervous system ,lincrna-p21 ,parkinson’s disease ,stau1-mediated mrna decay ,tgif1 ,Research Article - Abstract
Ghrelin is a neuropeptide that has various physiological functions and has been demonstrated to be neuroprotective in a number of neurological disease models. However, the underlying mechanisms of ghrelin in Parkinson's disease remain largely unexplored. The current study aimed to study the effects of ghrelin in a 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease model and evaluate the potential underlying mechanisms. In the present study, we treated an SH-SY5Y cell model with 6-OHDA, and observed that pretreatment with different concentrations of ghrelin (1, 10, and 100 nM) for 30 minutes relieved the neurotoxic effects of 6-OHDA, as revealed by Cell Counting Kit-8 and Annexin V/propidium iodide (PI) apoptosis assays. Reverse transcription quantitative polymerase chain reaction and western blot assay results demonstrated that 6-OHDA treatment upregulated α-synuclein and lincRNA-p21 and downregulated TG-interacting factor 1 (TGIF1), which was predicted as a potential transcription regulator of the gene encoding α-synuclein (SNCA). Ghrelin pretreatment was able to reverse the trends caused by 6-OHDA. The Annexin V/PI apoptosis assay results revealed that inhibiting either α-synuclein or lincRNA-p21 expression with small interfering RNA (siRNA) relieved 6-OHDA-induced cell apoptosis. Furthermore, inhibiting lincRNA-p21 also partially upregulated TGIF1. By retrieving information from a bioinformatics database and performing both double luciferase and RNA immunoprecipitation assays, we found that lincRNA-p21 and TGIF1 were able to form a double-stranded RNA-binding protein Staufen homolog 1 (STAU1) binding site and further activate the STAU1-mediated mRNA decay pathway. In addition, TGIF1 was able to transcriptionally regulate α-synuclein expression by binding to the promoter of SNCA. The Annexin V/PI apoptosis assay results showed that either knockdown of TGIF1 or overexpression of lincRNA-p21 notably abolished the neuroprotective effects of ghrelin against 6-OHDA-induced neurotoxicity. Collectively, these findings suggest that ghrelin exerts neuroprotective effects against 6-OHDA-induced neurotoxicity via the lincRNA-p21/TGIF1/α-synuclein pathway.
- Published
- 2022
28. TGIF1 Gene Silencing in Tendon-Derived Stem Cells Improves the Tendon-to-Bone Insertion Site Regeneration
- Author
-
Liyang Chen, Chaoyin Jiang, Shashi Ranjan Tiwari, Amrit Shrestha, Pengcheng Xu, Wenqing Liang, Yeqing Sun, Shisheng He, and Biao Cheng
- Subjects
Tendon-derived stem cells ,TGF-β ,TGIF1 ,Tendon-to-bone ,Regeneration ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background/Aims: The slow healing process of tendon-to-bone junctions can be accelerated via implanted tendon-derived stem cells (TDSCs) with silenced transforming growth interacting factor 1 (TGIF1) gene. Tendon-to-bone insertion site is the special form of connective tissues derivatives of common connective progenitors, where TGF-β plays bidirectional effects (chondrogenic or fibrogenic) through different signaling pathways at different stages. A recent study revealed that TGF-β directly induces the chondrogenic gene Sox9. However, TGIF1 represses the expression of the cartilage master Sox9 gene and changes its expression rate against the fibrogenesis gene Scleraxis (Scx). Methods: TGIF1 siRNA was transduced or TGIF1 was over-expressed in tendon-derived stem cells. Following suprapinatus tendon repair, rats were either treated with transduced TDSCs or nontransduced TDSCs. Histologic examination and Western blot were performed in both groups. Results: In this study, the silencing of TGIF1 significantly upregulated the chondrogenic genes and markers. Similarly, TGIF1 inhibited TDSC differentiation into cartilage via interactions with TGF-β-activated Smad2 and suppressed the phosphorylation of Smad2. The area of fibrocartilage at the tendon-bone interface was significantly increased in the TGIF1 (-) group compared with the control and TGIF1-overexpressing groups in the early stages of the animal model. The interface between the tendon and bone showed a increase of new bone and fibrocartilage in the TGIF1 (-) group at 4 weeks. Fibrovascular scar tissue was observed in the TGIF1-overexpressing group and the fibrin glue only group. Low levels of fibrocartilage and fibrovascular scar tissue were found in the TDSCs group. Conclusion: Collectively, this study shows that the tendon-derived stem cell modified with TGIF1 gene silencing has promising effects on tendon-to-bone healing which can be further explored as a therapeutic tool in regenerative medicine.
- Published
- 2015
- Full Text
- View/download PDF
29. Inhibition of microRNA-23b prevents polymicrobial sepsis-induced cardiac dysfunction by modulating TGIF1 and PTEN.
- Author
-
Zhang, Haiju, Caudle, Yi, Shaikh, Aamir, Yao, Baozhen, and Yin, Deling
- Subjects
- *
MICRORNA , *CARDIOVASCULAR disease treatment , *HEART fibrosis , *SEPTICEMIA treatment , *LUCIFERASES , *THERAPEUTICS - Abstract
Cardiovascular dysfunction is a major complication associated with sepsis induced mortality. Cardiac fibrosis plays a critical role in sepsis induced cardiac dysfunction. The mechanisms of the activation of cardiac fibrosis is unclarified. In this study, we found that microRNA-23b (miR-23b) was up-regulated in heart tissue during cecal ligation and puncture (CLP)-induced sepsis and transfection of miR-23b inhibitor improved survival in late sepsis. Inhibition of miR-23b in the myocardium protected against cardiac output and enhanced left ventricular systolic function. miR-23b inhibitor also alleviated cardiac fibrosis in late sepsis. MiR-23b mediates the activation of TGF-β1/Smad2/3 signaling to promote the differentiation of cardiac fibroblasts through suppression of 5′TG3′-interacting factor 1 (TGIF1). MiR-23b also induces AKT/N-Cadherin signaling to contribute to the deposition of extracellular matrix by inhibiting phosphatase and tensin homologue (PTEN). TGIF1 and PTEN were confirmed as the targets of miR-23b in vitro by Dual-Glo Luciferase assay. miR-23b inhibitor blocked the activation of adhesive molecules and restored the imbalance of pro-fibrotic and anti-fibrotic factors. These data provide direct evidence that miR-23b is a critical contributor to the activation of cardiac fibrosis to mediate the development of myocardial dysfunction in late sepsis. Blockade of miR-23b expression may be an effective approach for prevention sepsis-induced cardiac dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
30. Genetic Variants Contributing to Colistin Cytotoxicity: Identification of TGIF1 and HOXD10 Using a Population Genomics Approach.
- Author
-
Eadon, Michael T., Hause, Ronald J., Stark, Amy L., Ying-Hua Cheng, Wheeler, Heather E., Burgess, Kimberly S., Benson, Eric A., Cunningham, Patrick N., Bacallao, Robert L., Dagher, Pierre C., Skaar, Todd C., and Dolan, M. Eileen
- Subjects
- *
METAGENOMICS , *GRAM-negative bacterial diseases , *COLISTIN , *GENETIC markers , *NEPHROTOXICOLOGY , *THERAPEUTICS - Abstract
Colistin sulfate (polymixin E) is an antibiotic prescribed with increasing frequency for severe Gram-negative bacterial infections. As nephrotoxicity is a common side effect, the discovery of pharmacogenomic markers associated with toxicity would benefit the utility of this drug. Our objective was to identify genetic markers of colistin cytotoxicity that were also associated with expression of key proteins using an unbiased, whole genome approach and further evaluate the functional significance in renal cell lines. To this end, we employed International HapMap lymphoblastoid cell lines (LCLs) of Yoruban ancestry with known genetic information to perform a genome-wide association study (GWAS) with cellular sensitivity to colistin. Further association studies revealed that single nucleotide polymorphisms (SNPs) associated with gene expression and protein expression were significantly enriched in SNPs associated with cytotoxicity (p ≤ 0.001 for gene and p = 0.015 for protein expression). The most highly associated SNP, chr18:3417240 (p = 6.49 x 10-8), was nominally a cis-expression quantitative trait locus (eQTL) of the gene TGIF1 (transforming growth factor β (TGFβ)-induced factor-1; p = 0.021) and was associated with expression of the protein HOXD10 (homeobox protein D10; p = 7.17 x 10-5). To demonstrate functional relevance in a murine colistin nephrotoxicity model, HOXD10 immunohistochemistry revealed upregulated protein expression independent of mRNA expression in response to colistin administration. Knockdown of TGIF1 resulted in decreased protein expression of HOXD10 and increased resistance to colistin cytotoxicity. Furthermore, knockdown of HOXD10 in renal cells also resulted in increased resistance to colistin cytotoxicity, supporting the physiological relevance of the initial genomic associations. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
31. Tgif1 and Tgif2 Repress Expression of the RabGAP Evi5I.
- Author
-
Anderson, Anoush E., Yi Hao, Melhuish, Tiffany A., Shah, Anant, Wotton, David, Kenichiro Taniguchi, Turner, Stephen D., and Sutherland, Ann E.
- Subjects
- *
GENE expression , *GENETIC repressors , *TRANSFORMING growth factors , *FIBROBLASTS , *TRANSCRIPTION factors - Abstract
Mouse embryos conditionally lacking Tgif1 and Tgif2 have holoprosencephaly and defects in left-right asymmetry. To identify pathways affected by loss of Tgif function during embryogenesis, we performed transcriptome profiling on whole mouse embryos. Among the genes with altered expression in embryos lacking Tgifs were a number with links to cilium function. One of these, Evi5I, encodes a RabGAP that is known to block the formation of cilia when overexpressed. Evi5I expression is increased in Tgif1; Tgif2-null embryos and in double-null mouse embryo fibroblasts (MEFs). Knockdown of Tgifs in a human retinal pigment epithelial cell line also increased EVI5L expression. We show that TGIF1 binds to a conserved consensus TGIF site 5' of the human and mouse Evi5I genes and represses Evi5l expression. In primary MEFs lacking both Tgifs, the number of cells with primary cilia was significantly decreased, and we observed a reduction in the transcriptional response to Shh pathway activation. Reducing Evi5I expression in MEFs lacking Tgifs resulted in a partial restoration of cilium numbers and in the transcriptional response to activation of the Shh pathway. In summary, this work shows that Tgifs regulate ciliogenesis and suggests that Evi5l mediates at least part of this effect. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
32. Upregulation of miR-101a Suppresses Chronic Renal Fibrosis by Regulating KDM3A via Blockade of the YAP-TGF-β-Smad Signaling Pathway
- Author
-
Yanyan Xu, Hong Ding, and Nan Jiang
- Subjects
0301 basic medicine ,urologic and male genital diseases ,Article ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,chronic renal fibrosis ,Drug Discovery ,microRNA ,Renal fibrosis ,TGIF1 ,Medicine ,YAP-TGF-β-Smad signaling pathway ,YAP1 ,biology ,business.industry ,lcsh:RM1-950 ,microRNA-101a ,Fibronectin ,Reverse transcription polymerase chain reaction ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,KDM3A ,biology.protein ,Cancer research ,Molecular Medicine ,enhancer ,Signal transduction ,business ,Transforming growth factor - Abstract
Renal fibrosis denotes a common complication of diabetic nephropathy and is a predominant cause of end-stage renal disease. Despite the association between microRNAs (miRNAs or miRs) and renal fibrosis, miRNAs have been reported to play a vital role in the development of chronic renal fibrosis. Therefore, the aim of the present study was to investigate the possible function of miR-101a in chronic renal fibrosis. Initially, microarray-based gene expression profiling of renal fibrosis was employed to screen the differentially expressed genes. An in vivo mouse model of chronic renal fibrosis induced by a unilateral ureteral obstruction (UUO) and an in vitro cell model induced by aristolochic acid (AA) were constructed. miR-101a expression was examined using a fluorescence in situ hybridization (FISH) assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Then, the interaction between miR-101a and KDM3A was identified using an online website combined with a dual-luciferase reporter assay. Finally, gain- and loss-of-function experiments were conducted to elucidate the effect of miR-101a on the expression of Col1a1, fibronectin, α-smooth muscle actin (α-SMA), and YAP-TGF-β (transforming growth factor β)-Smad signaling pathway-related genes, as well as the degree of renal fibrosis. miR-101a was poorly expressed while KDM3A was robustly induced in chronic renal fibrosis tissues and cells. In addition, miR-101a could target and downregulate KDM3A expression, which led to elevated TGIF1, inhibited expression of Collagen I (Col1a1), fibronectin, α-SMA, YAP1, and TGF-β2 along with the extent of Smad2/3 phosphorylation, as well as delayed renal fibrosis degree. Besides, overexpressed YAP/TGF-β2 or inhibited TGIF1 partially restored the inhibitory effect of miR-101a on chronic renal fibrosis. Taken together, miR-101a could potentially slow down chronic renal fibrosis by the inactivation of the YAP-TGF-β-Smad signaling pathway via KDM3A, highlighting the potential of miR-101a as a therapeutic target for chronic renal fibrosis treatment. Keywords: chronic renal fibrosis, microRNA-101a, KDM3A, YAP-TGF-β-Smad signaling pathway, enhancer, TGIF1
- Published
- 2020
33. TGIF1-Twist1 axis in pancreatic ductal adenocarcinoma
- Author
-
Azeddine Atfi and Mohammed S. Razzaque
- Subjects
TGF-β ,lcsh:Biotechnology ,Biophysics ,Context (language use) ,SMAD ,Review Article ,Biology ,medicine.disease_cause ,Biochemistry ,law.invention ,Pathogenesis ,Pancreatic ductal adenocarcinoma ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Structural Biology ,law ,lcsh:TP248.13-248.65 ,Genetics ,medicine ,TGIF1 ,Transcription factor ,030304 developmental biology ,0303 health sciences ,Tumor suppressor ,medicine.disease ,Computer Science Applications ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,Signal transduction ,Carcinogenesis ,Biotechnology ,Twist1 - Abstract
TG-interacting factor 1 (TGIF1) exerts inhibitory effects on transforming growth factor-beta (TGF-β) signaling by suppressing Smad signaling pathway at multiple levels. TGIF1 activity is important for normal embryogenesis and organogenesis, yet its dysregulation can culminate in tumorigenesis. For instance, increased expression of TGIF1 correlates with poor prognosis in triple-negative breast cancer patients, and enforced expression of TGIF1 facilitates Wnt-driven mammary tumorigenesis, suggesting that TGIF1 might function as an oncoprotein. Quite surprisingly, TGIF1 has recently been shown to function as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC), possibly owing to its ability to antagonize the pro-malignant transcription factor Twist1. In this article, we will briefly elaborate on the biological and clinical significance of the unique tumor-suppressive function of TGIF1 and its functional interaction with Twist1 in the context of PDAC pathogenesis and progression.
- Published
- 2020
34. Allelic Variants in Established Hypopituitarism Genes Expand Our Knowledge of the Phenotypic Spectrum
- Author
-
Juliana Moreira Silva, Amanda de Moraes Narcizo, Anna Flavia Figueredo Benedetti, Alexander A. L. Jorge, Luciani R. Carvalho, Nathalia Garcia Bianchi Pereira Ferreira, Ivo J.P. Arnhold, Qing Fang, Mariana Cotarelli Madi, Mirian Yumie Nishi, Berenice B. Mendonca, Marilena Nakaguma, Ayse Bilge Ozel, Luciana Ribeiro Montenegro, Lais Cavalca Cardoso, Mariana F A Funari, Sally A. Camper, Jun Li, and Qianyi Ma
- Subjects
0301 basic medicine ,Male ,Genotype ,030209 endocrinology & metabolism ,Hypopituitarism ,Biology ,QH426-470 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Holoprosencephaly ,Anterior pituitary ,medicine ,Genetics ,TGIF1 ,Missense mutation ,Humans ,Allele ,Child ,Genetics (clinical) ,Alleles ,Homeodomain Proteins ,GH1 ,Human Growth Hormone ,SOXB1 Transcription Factors ,Infant ,Aplasia ,medicine.disease ,Penetrance ,Magnetic Resonance Imaging ,Pedigree ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,allelic variants ,hypopituitarism ,Child, Preschool ,Mutation ,IGHD ,SOX3 ,Female - Abstract
We report four allelic variants (three novel) in three genes previously established as causal for hypopituitarism or related disorders. A novel homozygous variant in the growth hormone gene, GH1 c.171delT (p.Phe 57Leufs*43), was found in a male patient with severe isolated growth hormone deficiency (IGHD) born to consanguineous parents. A hemizygous SOX3 allelic variant (p.Met304Ile) was found in a male patient with IGHD and hypoplastic anterior pituitary. YASARA, a tool to evaluate protein stability, suggests that p.Met304Ile destabilizes the SOX3 protein (ΔΔG = 2.49 kcal/mol). A rare, heterozygous missense variant in the TALE homeobox protein gene, TGIF1 (c.268C>, T:p.Arg90Cys) was found in a patient with combined pituitary hormone deficiency (CPHD), diabetes insipidus, and syndromic features of holoprosencephaly (HPE). This variant was previously reported in a patient with severe holoprosencephaly and shown to affect TGIF1 function. A novel heterozygous TGIF1 variant (c.82T>, C:p.Ser28Pro) was identified in a patient with CPHD, pituitary aplasia and ectopic posterior lobe. Both TGIF1 variants have an autosomal dominant pattern of inheritance with incomplete penetrance. In conclusion, we have found allelic variants in three genes in hypopituitarism patients. We discuss these variants and associated patient phenotypes in relation to previously reported variants in these genes, expanding our knowledge of the phenotypic spectrum in patient populations.
- Published
- 2021
- Full Text
- View/download PDF
35. TGIF1 promoted the growth and migration of cancer cells in nonsmall cell lung cancer.
- Author
-
Xiang, Guo, Yi, Yang, Weiwei, He, and Weiming, Wu
- Abstract
Transforming growth factor beta-inducing factor 1 (TGIF1) was reported to be dysregulated in several types of cancer. However, its expression pattern and functions in nonsmall cell lung cancer (NSCLC) remained unknown. In the present study, the expression of TGIF1 was found to be elevated in the clinical NSCLC tissues. TGIF1 promoted the growth and migration of NSCLC cells, while knocking down the expression of TGIF1 inhibited the growth and migration of NSCLC cells. Moreover, downregulation of TGIF1 impaired the metastasis of NSCLC cells. In the study for the molecular mechanisms, it was found that TGIF1 positively regulated beta-catenin/TCF signaling. In summary, our study demonstrated the oncogenic role of TGIF1 in NSCLC, and TGIF1 might be a therapeutic target for NSCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
36. TGIF1 Gene Silencing in Tendon-Derived Stem Cells Improves the Tendon-to-Bone Insertion Site Regeneration.
- Author
-
Chen, Liyang, Jiang, Chaoyin, Tiwari, Shashi Ranjan, Shrestha, amrit, Xu, Pengcheng, Liang, Wenqing, Sun, Yeqing, He, Shisheng, and Cheng, Biao
- Subjects
REGENERATION (Biology) ,FIBRIN tissue adhesive ,CONNECTIVE tissues ,MUSCULOSKELETAL system ,PROGENITOR cells - Abstract
Background/Aims: The slow healing process of tendon-to-bone junctions can be accelerated via implanted tendon-derived stem cells (TDSCs) with silenced transforming growth interacting factor 1 (TGIF1) gene. Tendon-to-bone insertion site is the special form of connective tissues derivatives of common connective progenitors, where TGF-β plays bidirectional effects (chondrogenic or fibrogenic) through different signaling pathways at different stages. A recent study revealed that TGF-β directly induces the chondrogenic gene Sox9. However, TGIF1 represses the expression of the cartilage master Sox9 gene and changes its expression rate against the fibrogenesis gene Scleraxis (Scx). Methods: TGIF1 siRNA was transduced or TGIF1 was over-expressed in tendon-derived stem cells. Following suprapinatus tendon repair, rats were either treated with transduced TDSCs or nontransduced TDSCs. Histologic examination and Western blot were performed in both groups. Results: In this study, the silencing of TGIF1 significantly upregulated the chondrogenic genes and markers. Similarly, TGIF1 inhibited TDSC differentiation into cartilage via interactions with TGF-β-activated Smad2 and suppressed the phosphorylation of Smad2. The area of fibrocartilage at the tendon-bone interface was significantly increased in the TGIF1 (-) group compared with the control and TGIFl-overexpressing groups in the early stages of the animal model. The interface between the tendon and bone showed a increase of new bone and fibrocartilage in the TGIF1 (-) group at 4 weeks. Fibrovascular scar tissue was observed in the TGIFl-overexpressing group and the fibrin glue only group. Low levels of fibrocartilage and fibrovascular scar tissue were found in the TDSCs group. Conclusion: Collectively, this study shows that the tendon-derived stem cell modified with TGIF1 gene silencing has promising effects on tendon-to-bone healing which can be further explored as a therapeutic tool in regenerative medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
37. Silencing of TGIF1 in bone mesenchymal stem cells applied to the post-operative rotator cuff improves both functional and histologic outcomes.
- Author
-
Li, Jie, Chen, Liyang, Sun, Lin, Chen, Hua, Sun, Yeqing, Jiang, Chaoyin, and Cheng, Biao
- Abstract
Stem cells have long been hypothesized to improve outcomes following rotator cuff repair. However, these cells must be signaled in order to do so. TGIF1 is a transcription factor that has been found to be down-regulated in cells involved in chondrogenesis. We therefore wished to examine whether stem cells expressing lower levels of TGIF1 could better improve outcomes following rotator cuff repair than stem cells expressing normal levels of TGIF1. Bone mesenchymal stem cells (BMSCs) were transduced with TGIF1 siRNA to suppress native TGIF1. Nontransduced BMSCs were also obtained for the control group. Following suprapinatus tendon repair, rats were either treated with transduced BMSCs or nontransduced BMSCs. Histologic and functional testing were performed on both groups. Rats treated with transduced TGIF1 siRNA BMSCs following suprapinatus repair expressed significantly higher levels of chondrogenic proteins at 4 weeks than rats treated with nontransduced BMSCs. Further, rats treated with BMSCs transduced with TGIF1 siRNA had both a significantly greater maximum load at failure and stiffness. Rats treated with transduced TGIF1 siRNA BMSCs following supraspinatus repair perform better both histologically and functionally at 4 weeks. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
38. PSPC1 is a new contextual determinant of aberrant subcellular translocation of oncogenes in tumor progression
- Author
-
Yuh-Shan Jou and Yaw-Dong Lang
- Subjects
0301 basic medicine ,NPM1 ,Carcinoma, Hepatocellular ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,NPM ,Review ,Biology ,Mortalin and EBP50 ,Translocation, Genetic ,03 medical and health sciences ,XPO1 ,0302 clinical medicine ,Neoplasms ,medicine ,TGIF1 ,Humans ,Pharmacology (medical) ,Nuclear export signal ,Molecular Biology ,Nucleophosmin ,Biochemistry (medical) ,Liver Neoplasms ,Cancer ,RNA-Binding Proteins ,Cell Biology ,General Medicine ,Oncogenes ,medicine.disease ,PSPC1 ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,Disease Progression ,Medicine ,Nucleocytoplasmic shuttling ,PTK6 ,Selective inhibitor of nucleocytoplasmic shuttling ,Transforming growth factor ,Oncogenic switch - Abstract
Dysregulation of nucleocytoplasmic shuttling is commonly observed in cancers and emerging as a cancer hallmark for the development of anticancer therapeutic strategies. Despite its severe adverse effects, selinexor, a selective first-in-class inhibitor of the common nuclear export receptor XPO1, was developed to target nucleocytoplasmic protein shuttling and received accelerated FDA approval in 2019 in combination with dexamethasone as a fifth-line therapeutic option for adults with relapsed refractory multiple myeloma (RRMM). To explore innovative targets in nucleocytoplasmic shuttling, we propose that the aberrant contextual determinants of nucleocytoplasmic shuttling, such as PSPC1 (Paraspeckle component 1), TGIF1 (TGF-β Induced Factor Homeobox 1), NPM1 (Nucleophosmin), Mortalin and EBP50, that modulate shuttling (or cargo) proteins with opposite tumorigenic functions in different subcellular locations could be theranostic targets for developing anticancer strategies. For instance, PSPC1 was recently shown to be the contextual determinant of the TGF-β prometastatic switch and PTK6/β-catenin reciprocal oncogenic nucleocytoplasmic shuttling during hepatocellular carcinoma (HCC) progression. The innovative nucleocytoplasmic shuttling inhibitor PSPC1 C-terminal 131 polypeptide (PSPC1-CT131), which was developed to target both the shuttling determinant PSPC1 and the shuttling protein PTK6, maintained their tumor-suppressive characteristics and exhibited synergistic effects on tumor suppression in HCC cells and mouse models. In summary, targeting the contextual determinants of nucleocytoplasmic shuttling with cargo proteins having opposite tumorigenic functions in different subcellular locations could be an innovative strategy for developing new therapeutic biomarkers and agents to improve cancer therapy.
- Published
- 2021
39. Role of TG-interacting factor (Tgif) in lipid metabolism.
- Author
-
Pramfalk, Camilla, Eriksson, Mats, and Parini, Paolo
- Subjects
- *
HOMEOBOX proteins , *LIPID metabolism , *NUCLEOTIDE sequence , *C-terminal binding proteins , *GENETIC mutation , *HOLOPROSENCEPHALY , *LIPID analysis - Abstract
TG interacting factors (Tgifs) 1 and 2 are members of the TALE (three-amino-acid loop extension) superfamily of homeodomain proteins. These two proteins bind to the same DNA sequence and share a conserved C-terminal repression domain. Mutations in TGIF1 have been linked to holoprosencephaly, which is a human genetic disease that affects craniofacial development. As these proteins can interact with the ligand binding domain of retinoid X receptor α, a common heterodimeric partner of several nuclear receptors [e.g., liver X receptors ( LXR s) and peroxisome proliferator-activated receptors ( PPAR s)], Tgif1 and Tgif2 might repress other transcriptional pathways activated by lipids. In line with this, Tgif1 interacts with LXRα and Tgif1 null mice have increased expression of the two Lxrα target genes apolipoproteins ( Apo ) c2 and a4 . Also, we have recently identified Tgif1 to function as a transcriptional repressor of the cholesterol esterifying enzyme acyl-coenzyme A:cholesterol acyltransferase 2 (gene name SOAT2 ). As no studies yet have shown involvement of Tgif2 in the lipid metabolism, this review will focus on the role of Tgif1 in lipid and cholesterol metabolism. This article is part of a Special Issue entitled: Linking transcription to physiology in lipodomics. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
40. The Chinese Medicine, Shezhi Huangling Decoction, Inhibits the Growth and Metastasis of Glioma Cells via the Regulation of miR-1298-5p/TGIF1 Axis
- Author
-
Xiaoqian Liu, Zongmin Zhu, Qun Liu, Jianfeng Ju, and Chunxia Liu
- Subjects
0301 basic medicine ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Western blot ,glioma ,Glioma ,TGIF1 ,medicine ,Original Research ,miR-1298-5p ,Chinese medicine ,medicine.diagnostic_test ,Cell growth ,Chemistry ,Transfection ,medicine.disease ,030104 developmental biology ,Shezhi Huangling decoction ,Oncology ,Apoptosis ,Cancer Management and Research ,030220 oncology & carcinogenesis ,Cancer research ,Immunohistochemistry ,Transforming growth factor - Abstract
Xiaoqian Liu,1 Jianfeng Ju,1 Qun Liu,1 Zongmin Zhu,1 Chunxia Liu2 1Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, People’s Republic of China; 2Department of Pharmacy, Caoxian People’s Hospital of Heze City, Heze 274400, Shandong, People’s Republic of ChinaCorrespondence: Jianfeng JuDepartment of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369, Jingshi Road, Jinan 250011, Shandong, People’s Republic of ChinaEmail yxbgongyou@163.comPurpose: In recent years, traditional Chinese medicine has achieved good results in treating gliomas. This research aimed to reveal the effect of Shezhi Huangling decoction (SD) on glioma cell process.Methods: U87 and U251 cells were treated with different concentrations (10, 30 and 50 μg/mL) of SD or transfected with miR-1298-5p mimic, inhibitor and siRNA targeting TGIF1. Cell proliferation, migration, invasion and apoptosis were detected. The expression of miR-1298-5p was measured by qRT-PCR, while TGIF1 expression was examined by immunohistochemical analysis and Western blot.Results: SD treatment inhibited the proliferation, migration and invasion of glioma cells and induced the apoptosis. In addition, SD treatment induced the expression of miR-1298-5p in glioma cells. The low expression of miR-1298-5p was examined in glioma tissues and was significantly related to the high histological grade of glioma patients and predicted a poor prognosis. MiR-1298-5p directly targeted the 3ʹ-UTR of transforming growth factor β induced factor 1 (TGIF1) and reduced TGIF1 protein expression. MiR-1298-5p restricted the proliferation, migration and invasion of glioma cells and induced cell apoptosis by targeting TGIF1.Conclusion: Our data reveal that SD acts as a cancer-inhibiting agent in glioma via miR-1298-5p/TGIF1 axis, suggesting a potential therapeutic application of SD in glioma.Keywords: glioma, Chinese medicine, Shezhi Huangling decoction, miR-1298-5p, TGIF1
- Published
- 2020
41. TGIF1 Knockdown Inhibits the Proliferation and Invasion of Gastric Cancer via AKT Signaling Pathway
- Author
-
Jiye Fan, Jing Zhang, Feiyan Zhang, and Bin Feng
- Subjects
0301 basic medicine ,Gene knockdown ,Chemistry ,Akt/PKB signaling pathway ,gastric cancer ,proliferation ,apoptosis ,Cancer ,Cell cycle ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,AKT pathway ,Oncology ,Cancer Management and Research ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Cancer research ,TGIF1 ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Original Research - Abstract
Jing Zhang,1 Feiyan Zhang,2 Jiye Fan,3,4 Bin Feng5 1Pharmacy Department, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei 050026, People’s Republic of China; 2Department of Outpatient Operating Room, Heze Municipal Hospital, Heze City, Shandong Province, 274000, People’s Republic of China; 3Department of Pharmaceutical Engineering, Hebei Chemical and Pharmaceutical College, Shijiazhuang, Hebei 050026, People’s Republic of China; 4College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, People’s Republic of China; 5Department of Gastrointestinal Surgery, Heze Municipal Hospital, Heze City, 274000 Shandong Province, People’s Republic of ChinaCorrespondence: Bin FengDepartment of Gastrointestinal Surgery, Heze Municipal Hospital, No. 2888, Caozhou Road, Mudan District, Heze City 274000, Shandong Province, People’s Republic of ChinaEmail 13020511011@163.comIntroduction: Gastric cancer is a kind of cancer with high mortality. TGIF1, as a transcription inhibitor, can inhibit the transcription of specific genes. The purpose of this study was to investigate the role of TGIF1 in gastric cancer by knocking down TGIF1.Methods: The expression of TGIF1 was detected by qPCR and Western blotting; CCK8 assay, colony formation assay, transwell, and wound-healing assay were used to evaluate the proliferation, migration, and invasion of gastric cancer cells; cell apoptosis was analyzed by flow cytometry and Hoechst-PI double staining; cell cycle was detected by flow cytometry. Gelatinase experiment was performed to detect the expression level of MMP-2; apoptosis related proteins and AKT singling pathway were assessed by Western blotting.Results: Knockdown of TGIF1 inhibited the proliferation, migration, and invasion of gastric cancer cells and promoted apoptosis. TGIF1 knockdown down-regulated the expression levels of MMP-2, Bcl2, CyclinD1, and p-Akt, and up-regulated the expression levels of Bax and Caspase3. These data suggested that knockdown of TGIF1 inhibited the development of gastric cancer via AKT signaling pathway.Conclusion: TGIF1 knockdown inhibited the proliferation, migration, and invasion and promoted apoptosis of gastric cancer cells via the AKT signaling pathway, suggesting that TGIF1 is considered a potential inhibitor in gastric cancer.Keywords: TGIF1, gastric cancer, AKT pathway, apoptosis, proliferation
- Published
- 2020
42. TGIF1 functions as a tumor suppressor in pancreatic ductal adenocarcinoma
- Author
-
Parajuli, Parash, Singh, Purba, Wang, Zhe, Li, Lianna, Eragamreddi, Sailaja, Ozkan, Seval, Ferrigno, Olivier, Prunier, Celine, Razzaque, Mohammed S, Xu, Keli, and Atfi, Azeddine
- Published
- 2019
- Full Text
- View/download PDF
43. TGFβ induced factor homeobox 1 promotes colorectal cancer development through activating Wnt/β-catenin signaling
- Author
-
Ye-Guang Chen, Jilian Wang, Hong-Mei Zhao, Zhen Qi, Wei Fu, and Yehua Li
- Subjects
TGF-β ,0301 basic medicine ,Colorectal cancer ,business.industry ,Wnt signaling pathway ,Cancer ,colorectal cancer ,β-catenin ,medicine.disease ,medicine.disease_cause ,Wnt signaling ,03 medical and health sciences ,030104 developmental biology ,Oncology ,Downregulation and upregulation ,Tumor progression ,Immunology ,TGIF1 ,medicine ,Cancer research ,Homeobox ,Carcinogenesis ,business ,Research Paper ,Transforming growth factor - Abstract
// Ji-Lian Wang 1 , Zhen Qi 2 , Ye-Hua Li 2 , Hong-Mei Zhao 1 , Ye-Guang Chen 2 and Wei Fu 1 1 Department of General Surgery, Peking University Third Hospital, Beijing 100191, China 2 The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China Correspondence to: Wei Fu, email: fuwei@bjmu.edu.cn Keywords: colorectal cancer, TGIF1, Wnt signaling, β-catenin, TGF-β Received: March 24, 2017 Accepted: June 20, 2017 Published: July 26, 2017 ABSTRACT Colorectal cancer (CRC) is one of the most common cancers, but the mechanisms underlying its initiation and progression are largely unknown. TGIF1 (TGFB induced factor homeobox 1) is a transcriptional corepressor that belongs to the three-amino acid loop extension (TALE) superclass of atypical homeodomains. It has been reported that TGIF1 is highly expressed in mammary cancer and non-small cell lung cancer and can enhance tumor progression. However, the role of TGIF1 in colorectal cancer remains unknown. Here, we report that TGIF1 is significantly upregulated in colorectal cancers, and its high expression predicts poor prognosis. Overexpression of TGIF1 markedly promotes the proliferation of colorectal cancer cells both in vivo and in vitro . In addition, TGIF1 activates Wnt/β-catenin signaling, and the homeodomain is indispensable for Wnt activation and β-catenin interaction. Taken together, our results suggest that TGIF1 is a novel colorectal tumor promoter and indicate that TGIF1 enhances colorectal cancer tumorigenesis through activating Wnt signaling.
- Published
- 2017
- Full Text
- View/download PDF
44. Targeted interference of SIN3A-TGIF1 function by SID decoy treatment inhibits Wnt signaling and invasion in triple negative breast cancer cells
- Author
-
Boris A. Leibovitch, Arthur Zelent, Eduardo F. Farias, Yeon-Jin Kwon, Samuel Waxman, Nidhi Bansal, Lutecia Pereira, Edgardo V. Ariztia, and Chi-Yeh Chung
- Subjects
0301 basic medicine ,Metastasis ,03 medical and health sciences ,Wnt ,TGIF1 ,Medicine ,metastasis ,Epithelial–mesenchymal transition ,Triple-negative breast cancer ,SIN3A ,business.industry ,Actin cytoskeleton reorganization ,Wnt signaling pathway ,Cancer ,medicine.disease ,invasion ,3. Good health ,030104 developmental biology ,Oncology ,triple negative breast cancer ,Cancer cell ,Immunology ,Cancer research ,business ,Decoy ,Research Paper - Abstract
// Yeon-Jin Kwon 1 , Boris A. Leibovitch 1 , Nidhi Bansal 1 , Lutecia Pereira 2 , Chi-Yeh Chung 1 , Edgardo V. Ariztia 1 , Arthur Zelent 2 , Eduardo F. Farias 1 and Samuel Waxman 1 1 Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA 2 University of Miami, Sylvester Comprehensive Cancer Center, Florida MI, USA Correspondence to: Samuel Waxman, email: samuel.waxman@mssm.edu Eduardo F. Farias, email: eduardo.farias@mssm.edu Keywords: triple negative breast cancer, invasion, SIN3A, TGIF1, Wnt, metastasis Received: May 20, 2016 Accepted: July 23, 2016 Published: August 19, 2016 ABSTRACT Cancer cell invasion is an obligatory step for metastatic dissemination that contributes to rapid relapse and a poorer survival in triple negative breast cancer (TNBC) patients. Development of novel therapeutic strategies to block tumor invasion is an unmet need in the treatment of cancer. We reported that the selective inhibition of the PAH2 domain of SIN3A protein function markedly suppressed metastatic dissemination to the lungs in TNBC xenograft bearing mice. Here, we show that TNBC cell lines treated with Sin3 interaction domain (SID) decoy peptides that bind to PAH2 display a strong in vitro inhibition of transwell invasion. This is accompanied by actin cytoskeleton reorganization with increased cortical actin deposition and downregulation of known Wnt target genes that are associated with epithelial to mesenchymal transition (EMT) and cancer cell invasion. Wnt pathway inhibition by SID decoy peptide was confirmed by decreased Wnt reporter activity and altered cytoplasmic localization of nuclear β-catenin. TGIF1, a transcription factor that modulates Wnt signaling and known to interact with the PAH2 domain of SIN3A, can be dissociated from the SIN3A complex by SID decoys. TGIF1 knockdown inhibits WNT target genes and in vitro cell invasion suggesting that TGIF1 might be a key target of the SID decoys to block tumor invasion. Taken together, targeting SIN3 function using SID decoys is a novel strategy to reverse invasion and the EMT program in TNBC translating into the inhibition of metastasis dissemination and eradication of residual disease.
- Published
- 2016
45. Allelic Variants in Established Hypopituitarism Genes Expand Our Knowledge of the Phenotypic Spectrum.
- Author
-
Nakaguma, Marilena, Ferreira, Nathalia Garcia Bianchi Pereira, Benedetti, Anna Flavia Figueredo, Madi, Mariana Cotarelli, Silva, Juliana Moreira, Li, Jun Z., Ma, Qianyi, Bilge Ozel, Ayse, Fang, Qing, Narcizo, Amanda de Moraes, Cardoso, Laís Cavalca, Montenegro, Luciana Ribeiro, Funari, Mariana Ferreira de Assis, Nishi, Mirian Yumie, Arnhold, Ivo Jorge Prado, Jorge, Alexander Augusto de Lima, Mendonca, Berenice Bilharinho de, Camper, Sally Ann, and Carvalho, Luciani R.
- Subjects
PHENOTYPES ,HYPOPITUITARISM ,GENETIC variation ,MISSENSE mutation ,HOMEOBOX proteins ,PITUITARY dwarfism ,PROTEIN stability - Abstract
We report four allelic variants (three novel) in three genes previously established as causal for hypopituitarism or related disorders. A novel homozygous variant in the growth hormone gene, GH1 c.171delT (p.Phe 57Leufs*43), was found in a male patient with severe isolated growth hormone deficiency (IGHD) born to consanguineous parents. A hemizygous SOX3 allelic variant (p.Met304Ile) was found in a male patient with IGHD and hypoplastic anterior pituitary. YASARA, a tool to evaluate protein stability, suggests that p.Met304Ile destabilizes the SOX3 protein (ΔΔG = 2.49 kcal/mol). A rare, heterozygous missense variant in the TALE homeobox protein gene, TGIF1 (c.268C>T:p.Arg90Cys) was found in a patient with combined pituitary hormone deficiency (CPHD), diabetes insipidus, and syndromic features of holoprosencephaly (HPE). This variant was previously reported in a patient with severe holoprosencephaly and shown to affect TGIF1 function. A novel heterozygous TGIF1 variant (c.82T>C:p.Ser28Pro) was identified in a patient with CPHD, pituitary aplasia and ectopic posterior lobe. Both TGIF1 variants have an autosomal dominant pattern of inheritance with incomplete penetrance. In conclusion, we have found allelic variants in three genes in hypopituitarism patients. We discuss these variants and associated patient phenotypes in relation to previously reported variants in these genes, expanding our knowledge of the phenotypic spectrum in patient populations. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
46. Effects of TG interaction factor 1 on synthesis of estradiol and progesterone in granulosa cells of goats through SMAD2/3-SP1 signaling pathway.
- Author
-
An, Xiaopeng, Cao, Heran, Liu, Shujuan, and Cao, Binyun
- Subjects
- *
GRANULOSA cells , *GOATS , *SMAD proteins , *GENE silencing , *ESTRADIOL , *MESSENGER RNA , *PROGESTERONE , *PROGESTERONE receptors - Abstract
The TG interaction factor 1 (TGIF1) is of the TALE homologue domain protein family and is considered as a transcriptional repressor of SMAD protein that interacts with DNA through a specific consensus-binding site for TG and recruits mSin3A and histone deacetylases to the SMAD complex. In this study, there is the first detailed description of TGIF1 on steroidogenesis in goat granulosa cells. When there is a relatively greater expression of the TGIF1 gene, there is a lesser abundance of CYP11A1 , CYP19A1 , and StAR mRNA transcript and protein and 3β-HSD mRNA transcript in granulosa cells of goats. Furthermore, there were lesser concentrations of 17β-estradiol (E 2) and progesterone (P 4) in culture medium when there was greater TGIF1 gene expression and there were greater concentrations of these hormones in the culture medium when there was lesser TGIF1 gene expression. There may be functions of TGIF1, therefore, in suppression of SMAD-induced E 2 and P 4 production and in decreasing the phosphorylation of SMAD2/3 in granulosa cells of goats and relative abundance of the SMAD2/3 protein transcription factor, SP1. With suppression of TGIF1 gene expression, there was a reversal of SP1-induced suppression of steroidogenesis-related genes. Results of the present study provide insights about the potential mechanism underlying the regulation of granulosa cell steroidogenesis of goats by TGIF1. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Genetic Variants Contributing to Colistin Cytotoxicity: Identification of TGIF1 and HOXD10 Using a Population Genomics Approach
- Author
-
Ronald J. Hause, Pierre C. Dagher, Eric A. Benson, Kimberly S. Burgess, Robert L. Bacallao, Amy L. Stark, Heather E. Wheeler, Todd C. Skaar, Ying Hua Cheng, Michael T. Eadon, Patrick N. Cunningham, and M. Eileen Dolan
- Subjects
0301 basic medicine ,Quantitative Trait Loci ,Drug Resistance ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Catalysis ,Article ,Cell Line ,Inorganic Chemistry ,lcsh:Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Gene expression ,medicine ,TGIF1 ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Gene ,lcsh:QH301-705.5 ,Spectroscopy ,Genetics ,Homeodomain Proteins ,Gene knockdown ,Colistin ,nephrotoxicity ,Organic Chemistry ,General Medicine ,Molecular biology ,HOXD10 ,3. Good health ,Computer Science Applications ,Anti-Bacterial Agents ,Repressor Proteins ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,lymphoblastoid cell line ,030220 oncology & carcinogenesis ,Expression quantitative trait loci ,colistin ,medicine.drug ,Genome-Wide Association Study ,Transcription Factors - Abstract
Colistin sulfate (polymixin E) is an antibiotic prescribed with increasing frequency for severe Gram-negative bacterial infections. As nephrotoxicity is a common side effect, the discovery of pharmacogenomic markers associated with toxicity would benefit the utility of this drug. Our objective was to identify genetic markers of colistin cytotoxicity that were also associated with expression of key proteins using an unbiased, whole genome approach and further evaluate the functional significance in renal cell lines. To this end, we employed International HapMap lymphoblastoid cell lines (LCLs) of Yoruban ancestry with known genetic information to perform a genome-wide association study (GWAS) with cellular sensitivity to colistin. Further association studies revealed that single nucleotide polymorphisms (SNPs) associated with gene expression and protein expression were significantly enriched in SNPs associated with cytotoxicity (p ≤ 0.001 for gene and p = 0.015 for protein expression). The most highly associated SNP, chr18:3417240 (p = 6.49 × 10-8), was nominally a cis-expression quantitative trait locus (eQTL) of the gene TGIF1 (transforming growth factor β (TGFβ)-induced factor-1; p = 0.021) and was associated with expression of the protein HOXD10 (homeobox protein D10; p = 7.17 × 10-5). To demonstrate functional relevance in a murine colistin nephrotoxicity model, HOXD10 immunohistochemistry revealed upregulated protein expression independent of mRNA expression in response to colistin administration. Knockdown of TGIF1 resulted in decreased protein expression of HOXD10 and increased resistance to colistin cytotoxicity. Furthermore, knockdown of HOXD10 in renal cells also resulted in increased resistance to colistin cytotoxicity, supporting the physiological relevance of the initial genomic associations.
- Published
- 2017
48. New SHH and Known SIX3 Variants in a Series of Latin American Patients with Holoprosencephaly.
- Author
-
de Castro VF, Mattos D, de Carvalho FM, Cavalcanti DP, Duenas-Roque MM, Llerena J Jr, Cosentino VR, Honjo RS, Leite JCL, Sanseverino MT, de Souza MPA, Bernardi P, Bolognese AM, Santana da Silva LC, Barbero P, Correia PS, Bueno LSM, Savastano CP, and Orioli IM
- Abstract
Holoprosencephaly (HPE) is the failure of the embryonic forebrain to develop into 2 hemispheres promoting midline cerebral and facial defects. The wide phenotypic variability and causal heterogeneity make genetic counseling difficult. Heterozygous variants with incomplete penetrance and variable expressivity in the SHH , SIX3 , ZIC2 , and TGIF1 genes explain ∼25% of the known causes of nonchromosomal HPE. We studied these 4 genes and clinically described 27 Latin American families presenting with nonchromosomal HPE. Three new SHH variants and a third known SIX3 likely pathogenic variant found by Sanger sequencing explained 15% of our cases. Genotype-phenotype correlation in these 4 families and published families with identical or similar driver gene, mutated domain, conservation of residue in other species, and the type of variant explain the pathogenicity but not the phenotypic variability. Nine patients, including 2 with SHH pathogenic variants, presented benign variants of the SHH , SIX3 , ZIC2 , and TGIF1 genes with potential alteration of splicing, a causal proposition in need of further studies. Finding more families with the same SIX3 variant may allow further identification of genetic or environmental modifiers explaining its variable phenotypic expression., Competing Interests: The authors have no conflicts of interest to declare., (Copyright © 2021 by S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
49. Upregulation of miR-101a Suppresses Chronic Renal Fibrosis by Regulating KDM3A via Blockade of the YAP-TGF-β-Smad Signaling Pathway.
- Author
-
Ding H, Xu Y, and Jiang N
- Abstract
Renal fibrosis denotes a common complication of diabetic nephropathy and is a predominant cause of end-stage renal disease. Despite the association between microRNAs (miRNAs or miRs) and renal fibrosis, miRNAs have been reported to play a vital role in the development of chronic renal fibrosis. Therefore, the aim of the present study was to investigate the possible function of miR-101a in chronic renal fibrosis. Initially, microarray-based gene expression profiling of renal fibrosis was employed to screen the differentially expressed genes. An in vivo mouse model of chronic renal fibrosis induced by a unilateral ureteral obstruction (UUO) and an in vitro cell model induced by aristolochic acid (AA) were constructed. miR-101a expression was examined using a fluorescence in situ hybridization (FISH) assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Then, the interaction between miR-101a and KDM3A was identified using an online website combined with a dual-luciferase reporter assay. Finally, gain- and loss-of-function experiments were conducted to elucidate the effect of miR-101a on the expression of Col1a1, fibronectin, α-smooth muscle actin (α-SMA), and YAP-TGF-β (transforming growth factor β)-Smad signaling pathway-related genes, as well as the degree of renal fibrosis. miR-101a was poorly expressed while KDM3A was robustly induced in chronic renal fibrosis tissues and cells. In addition, miR-101a could target and downregulate KDM3A expression, which led to elevated TGIF1, inhibited expression of Collagen I (Col1a1), fibronectin, α-SMA, YAP1, and TGF-β2 along with the extent of Smad2/3 phosphorylation, as well as delayed renal fibrosis degree. Besides, overexpressed YAP/TGF-β2 or inhibited TGIF1 partially restored the inhibitory effect of miR-101a on chronic renal fibrosis. Taken together, miR-101a could potentially slow down chronic renal fibrosis by the inactivation of the YAP-TGF-β-Smad signaling pathway via KDM3A, highlighting the potential of miR-101a as a therapeutic target for chronic renal fibrosis treatment., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
50. Die Rolle von TGIF1 in der peripheren T-Zellaktivierung und Differenzierung
- Author
-
Krüger, Melanie
- Subjects
TGIF1 ,T cell differentiation ,Foxp3 induction ,hemic and immune systems ,chemical and pharmacologic phenomena ,regulatory T cells - Abstract
Regulatorische T-Zellen (Treg-Zellen), die den Transkriptionsfaktor Foxp3 (Forkehead protein P3) exprimieren sind maßgeblich für die Aufrechterhaltung der peripheren Immuntoleranz verantwortlich. Für die Induktion der Treg-Zellen in der Peripherie ist die Stimulation des TGF-β-Signalweges zusammen mit einer suboptimalen T-Zell-Rezeptor (TCR)-Stimulation essentiell. In dieser Arbeit wurde untersucht, ob und wie der transkriptionelle Repressor des TGF-β-Signalweges TGIF1 (TGF-β interacting factor) die Induktion von peripheren Treg Zellen beeinflusst. Es konnte gezeigt werden, dass TGIF1 in T-Zellen exprimiert wird und seine Expressionshöhe von der TCR- Stimulationsstärke, nicht aber vom TGF-β-Signalweg abhängig ist. Mit Hilfe von TGIF1-KO-Mäusen wurde nachgewiesen, dass die Expressionshöhe von TGIF1 in T-Zellen einen entscheidenden Einfluss auf die Induktion von peripheren Treg- Zellen hat. Eine geringe oder fehlende Expression von TGIF1 fördert die in vitro Differenzierung von naiven T-Zellen zu Treg-Zellen. Im Gegensatz dazu ist die Differenzierung von TGIF1-KO-Zellen zu IFN-gamma-produzierenden Th1-Zellen im Vergleich zu WT-T-Zellen eingeschränkt. Zur Identifizierung der molekularen Wirkmechanismen von TGIF1 wurden ChIP (Chromatin Immunopräzipitation) -Sequenzierungen sowie globale Genexpressionanalysen durchgeführt. Dabei konnten verschiedene Zielgene von TGIF1, wie z. B. die Transkriptionsfaktoren Foxp3, KLF10, T-bet, STAT3 und Foxo3 sowie die Rezeptoren IL12R, und TGF-βRI identifiziert werden. Die Bindung und Wirkung von TGIF1 auf die Gene foxp3 und klf10 konnten bereits verifiziert werden. Damit erweist sich TGIF1 nicht nur als Repressor der Induktion peripherer Treg-Zellen, sondern auch als Aktivator der IFN-gamma Produktion in differenzierten Th1-Zellen. Weitere in vivo Experimente sollen zeigen, ob die Hemmung dieser beiden Effekte durch die Inhibierung von TGIF1 einen geeigneten therapeutischen Ansatz zur Behandlung von Autoimmunerkrankungen und Allergien bietet., Regulatory T-cells (Tregs-cells), which are characterized by expression of the transcription factor Foxp3 (Forkehead protein P3), are pivotal for the maintainence of peripheral immuntolerance. The induction of Treg cells from naïve T cells in the periphery critically depends on sub-optimal T-cell receptor (TCR) stimulation and TGF-β signaling. In this study the significance of TGIF1, a transcriptional repressor of TGF-β signaling, for the peripheral induction of Treg cells was investigated. It was shown that TGIF1 is expressed in T cells and that TGIF1 expression levels are dependent on TCR stimulation strength, but independent of TGF-β stimulation. By using T cells isolated from TGIF-KO-mice it was demonstrated that expression levels of TGIF1 have a significant impact on the induction of peripheral Treg cells. Little or missing expression of TGIF1 protein correlate with an increased induction of Foxp3+Tregs cells from naive T-cells in vitro. To identify the molecular mechanisms of TGIF1 action ChIP (Chromatin Immunoprecipitation)-sequencing and microarray analysis have been carried out. In this context different target genes of TGIF1 were identified, such as the transcription factors Foxp3, KLF10, T-bet, STAT3 and Foxo3 but also receptors such as IL-12R and TGF-βRI. In fact, binding of TGIF1 to foxp3 and klf10 genes as well as its impact on transcription of these genes could already be verified. Apart from effects on Treg induction, a major role of TGIF1 signaling for the differentiation of Th1 effector cells was revealed. It was shown that stimulation of TGIF1-KO-T cells under Th1 polarising conditions results in a significant reduction of IFN- gamma-producing cells in comparison to WT-T-cells. Taken together, the results of this work demonstrate that TGIF1 does not only act as a repressor of peripheral Treg cell induction but also as an activator of IFN-gamma production in Th1-cells. Future experiments will clarify whether inhibition of TGIF1-dependent effects on T cell differentiation has potential for the therapy of autoimmune diseases and allergies.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.