1
|
Duan C, Yang L, Chen W, Zhou D, Cao S, Zhang Y, Li G, Chen H, Tian C. Long-term thermal stress reshapes the tolerance of head kidney of Hong Kong catfish (Clarias fuscus) to acute heat shock by regulating energy metabolism and immune response. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101437. [PMID: 39933313 DOI: 10.1016/j.cbd.2025.101437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Elevated water temperatures caused by climate warming can affect fish survival. However, fish can maintain normal physiological functions through physiological plasticity. When temperature fluctuations exceed their tolerance range, even stress-resistant species like Siluriformes are affected. It is known that fish have adaptive regulation mechanisms to reshape their tolerance to temperature stress, but the ability to respond to acute thermal shock and recover after adaptive remodeling remains unclear. This study investigated the effects of different culture temperatures on the ability of Hong Kong catfish (Clarias fuscus) to respond to acute heat stress and stress recovery. C. fuscus were cultured at normal temperature (NT, 26 °C) or high temperature (HT, 34 °C) for 90 days, and then their head kidney transcriptome was analyzed after acute heat stress (34 °C) and subsequent recovery (26 °C). The results revealed 8165 differentially expressed genes (DEGs) in the NT group and 8537 DEGs in the HT group during the entire temperature treatment process, with each group responding differently to various stages of temperature treatment. Enrichment analysis showed that both NT and HT groups had enriched pathways related to energy metabolism and immune response during acute heat stress. However, acute heat stress disrupted the energy supply and oxidative metabolism in the NT group, while enhancing the HT group's ability to respond to repeated heat stress. This experiment demonstrated that high-temperature culture reshaped the energy metabolism balance in the head kidney tissue, improving anti-stress and stress recovery abilities. These findings lay a foundation for further research on the plasticity of fish in coping with acute temperature changes.
Collapse
Affiliation(s)
- Cunyu Duan
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Lei Yang
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Wanying Chen
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Dayan Zhou
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China
| | - Shouxiong Cao
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China
| | - Yulei Zhang
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Guangli Li
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Huapu Chen
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China
| | - Changxu Tian
- Fisheries College, Guangdong Ocean University, Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Lab of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang 524088, China.
| |
Collapse
|
2
|
Chang YH, Tseng YH, Wang JM, Tsai YS, Liu XL, Huang HS. Phosphorylation of TG-interacting factor 1 at carboxyl-terminal sites in response to insulin regulates adipocyte differentiation. FEBS Lett 2024; 598:945-955. [PMID: 38472156 DOI: 10.1002/1873-3468.14849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 03/14/2024]
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.
Collapse
Affiliation(s)
- Yu-Hao Chang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Ju-Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Xin-Lei Liu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huei-Sheng Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
3
|
Parajuli P, Singh P, Wang Z, Li L, Eragamreddi S, Ozkan S, Ferrigno O, Prunier C, Razzaque MS, Xu K, Atfi A. TGIF1 functions as a tumor suppressor in pancreatic ductal adenocarcinoma. EMBO J 2019; 38:e101067. [PMID: 31268604 PMCID: PMC6601038 DOI: 10.15252/embj.2018101067] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/11/2019] [Accepted: 04/23/2019] [Indexed: 02/05/2023] Open
Abstract
A prominent function of TGIF1 is suppression of transforming growth factor beta (TGF-β) signaling, whose inactivation is deemed instrumental to the progression of pancreatic ductal adenocarcinoma (PDAC), as exemplified by the frequent loss of the tumor suppressor gene SMAD4 in this malignancy. Surprisingly, we found that genetic inactivation of Tgif1 in the context of oncogenic Kras, KrasG12D , culminated in the development of highly aggressive and metastatic PDAC despite de-repressing TGF-β signaling. Mechanistic experiments show that TGIF1 associates with Twist1 and inhibits Twist1 expression and activity, and this function is suppressed in the vast majority of human PDACs by KrasG12D /MAPK-mediated TGIF1 phosphorylation. Ablating Twist1 in KrasG12D ;Tgif1KO mice completely blunted PDAC formation, providing the proof-of-principle that TGIF1 restrains KrasG12D -driven PDAC through its ability to antagonize Twist1. Collectively, these findings pinpoint TGIF1 as a potential tumor suppressor in PDAC and further suggest that sustained activation of TGF-β signaling might act to accelerate PDAC progression rather than to suppress its initiation.
Collapse
Affiliation(s)
- Parash Parajuli
- Cellular and Molecular Pathogenesis DivisionDepartment of Pathology and Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
| | - Purba Singh
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Zhe Wang
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Lianna Li
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | | | - Seval Ozkan
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Olivier Ferrigno
- Centre de Recherche Saint‐Antoine, CRSAInsermSorbonne UniversitésParisFrance
| | - Celine Prunier
- Centre de Recherche Saint‐Antoine, CRSAInsermSorbonne UniversitésParisFrance
| | | | - Keli Xu
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Azeddine Atfi
- Cellular and Molecular Pathogenesis DivisionDepartment of Pathology and Massey Cancer CenterVirginia Commonwealth UniversityRichmondVAUSA
- Centre de Recherche Saint‐Antoine, CRSAInsermSorbonne UniversitésParisFrance
| |
Collapse
|
4
|
Yu X, Shen G, Ren H, Zhang Z, Shang Q, Zhao W, Huang J, Yang Z, Liang D, Jiang X. TGFβ-induced factor homeobox 2 blocks osteoblastic differentiation through targeting pSmad3/HDAC4/H4ac/Runx2 axis. J Cell Physiol 2019; 234:21284-21293. [PMID: 31066043 DOI: 10.1002/jcp.28733] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/02/2019] [Accepted: 04/11/2019] [Indexed: 01/22/2023]
Abstract
TGFβ-induced factor homeobox 2 (Tgif2) has been reported as a functional role in cell homeostasis and a key activator of osteoclastogenesis and bone loss, as well. In the present study, we aimed to investigate the potential role of Tgif2 on osteogenic differentiation. Tgif2 expression was assessed during the osteogenic differentiation process of bone marrow-derived mesenchymal stem cells (BMSCs) and primary calvarial osteoblasts (OBs). The expression of Tgif2 in BMSCs and OBs increased by using lentivirus-mediated gene overexpression (OE). The effect of Tgif2 on osteogenic differentiation was compared between Tgif2 negative control (Tgif2-NC) and Tgif2-OE group in BMSCs/OBs via performing alkaline phosphatase (ALP) assay, mineralization assay, and gene expression analysis of some osteogenic markers. To investigate the molecular mechanism, the direct interaction of histone deacetylase 4 (HDAC4) and pSmad3, acetylated histone H4 (H4ac), and Runx2-binding site of the Ocn promoter was confirmed by performing co-immunoprecipitation (CoIP) and chromatin immunoprecipitation (ChIP) assay, respectively. The results showed that Tgif2 abundantly expressed in BMSCs and primary calvarial OBs, but decreased after osteogenic induction. In vitro, osteogenic differentiation was significantly inhibited with Tgif2 overexpression in both BMSCs and OBs, as well as the expression levels of osteogenic markers (Runx2, Sp7, Alp, and Ocn). Moreover, we found that Tgif2 overexpression significantly promoted the interaction of pSmad3 with HDAC4 in differentiated OBs, and sequentially decreased the abundance of H4ac at the Runx2-binding site of the Ocn promoter. These findings indicated that Tgif2 might block osteoblastic differentiation in vitro through targeting pSmad3/HDAC4/H4ac/Runx2 axis.
Collapse
Affiliation(s)
- Xiang Yu
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shang
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenhua Zhao
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinjing Huang
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhidong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Wotton D, Taniguchi K. Functions of TGIF homeodomain proteins and their roles in normal brain development and holoprosencephaly. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 178:128-139. [PMID: 29749689 DOI: 10.1002/ajmg.c.31612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 01/08/2023]
Abstract
Holoprosencephaly (HPE) is a frequent human forebrain developmental disorder with both genetic and environmental causes. Multiple loci have been associated with HPE in humans, and potential causative genes at 14 of these loci have been identified. Although TGIF1 (originally TGIF, for Thymine Guanine-Interacting Factor) is among the most frequently screened genes in HPE patients, an understanding of how mutations in this gene contribute to the pathogenesis of HPE has remained elusive. However, mouse models based on loss of function of Tgif1, and the related Tgif2 gene, have shed some light on how human TGIF1 variants might cause HPE. Functional analyses of TGIF proteins and of TGIF1 single nucleotide variants from HPE patients, combined with analysis of forebrain development in mouse embryos lacking both Tgif1 and Tgif2, suggest that TGIFs regulate the transforming growth factor ß/Nodal signaling pathway and sonic hedgehog (SHH) signaling independently. Although, some developmental processes that are regulated by TGIFs may be Nodal-dependent, it appears that the forebrain patterning defects and HPE in Tgif mutant mouse embryos is primarily due to altered signaling via the Shh pathway.
Collapse
Affiliation(s)
- David Wotton
- Department of Biochemistry and Molecular Genetics, Center for Cell Signaling, University of Virginia, Charlottesville, Virginia
| | - Kenichiro Taniguchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
6
|
Tgif1 and Tgif2 Repress Expression of the RabGAP Evi5l. Mol Cell Biol 2017; 37:MCB.00527-16. [PMID: 27956704 DOI: 10.1128/mcb.00527-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/02/2016] [Indexed: 11/20/2022] Open
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, Evi5l, encodes a RabGAP that is known to block the formation of cilia when overexpressed. Evi5l 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 Evi5l 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 Evi5l 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.
Collapse
|
7
|
Stepwise reprogramming of liver cells to a pancreas progenitor state by the transcriptional regulator Tgif2. Nat Commun 2017; 8:14127. [PMID: 28193997 PMCID: PMC5316826 DOI: 10.1038/ncomms14127] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 11/11/2016] [Indexed: 12/12/2022] Open
Abstract
The development of a successful lineage reprogramming strategy of liver to pancreas holds promises for the treatment and potential cure of diabetes. The liver is an ideal tissue source for generating pancreatic cells, because of its close developmental origin with the pancreas and its regenerative ability. Yet, the molecular bases of hepatic and pancreatic cellular plasticity are still poorly understood. Here, we report that the TALE homeoprotein TGIF2 acts as a developmental regulator of the pancreas versus liver fate decision and is sufficient to elicit liver-to-pancreas fate conversion both ex vivo and in vivo. Hepatocytes expressing Tgif2 undergo extensive transcriptional remodelling, which represses the original hepatic identity and, over time, induces a pancreatic progenitor-like phenotype. Consistently, in vivo forced expression of Tgif2 activates pancreatic progenitor genes in adult mouse hepatocytes. This study uncovers the reprogramming activity of TGIF2 and suggests a stepwise reprogramming paradigm, whereby a 'lineage-restricted' dedifferentiation step precedes the identity switch.
Collapse
|
8
|
Taniguchi K, Anderson AE, Melhuish TA, Carlton AL, Manukyan A, Sutherland AE, Wotton D. Genetic and Molecular Analyses indicate independent effects of TGIFs on Nodal and Gli3 in neural tube patterning. Eur J Hum Genet 2016; 25:208-215. [PMID: 27924807 DOI: 10.1038/ejhg.2016.164] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/14/2016] [Accepted: 10/25/2016] [Indexed: 01/11/2023] Open
Abstract
Holoprosencephaly (HPE) is a prevalent craniofacial developmental disorder that has both genetic and environmental causes. The gene encoding TG-interacting factor 1 (TGIF1) is among those that are routinely screened in HPE patients. However, the mechanisms by which TGIF1 variants cause HPE are not fully understood. TGIF1 is a transcriptional repressor that limits the output of the Transforming Growth Factor ß (TGFß)/Nodal signaling pathway, and HPE in patients with TGIF1 variants has been suggested to be due to increased Nodal signaling. Mice lacking both Tgif1 and its paralog, Tgif2, have HPE, and embryos lacking Tgif function do not survive past mid-gestation. Here, we show that in the presence of a Nodal heterozygous mutation, proliferation defects are rescued and a proportion of embryos lacking all Tgif function survive to late gestation. However, these embryos have a classic HPE phenotype, suggesting that this is a Nodal-independent effect of Tgif loss of function. Further, we show that the Gli3 gene is a direct target for repression by Tgifs, independent of TGFß/Nodal signaling, consistent with Tgif mutations causing HPE via Nodal-independent effects on the Sonic Hedgehog (Shh) pathway. Based on this work, we propose a model for distinct functions of Tgifs in the Nodal and Shh/Gli3 pathways during forebrain development.
Collapse
Affiliation(s)
- Kenichiro Taniguchi
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| | - Anoush E Anderson
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| | - Tiffany A Melhuish
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| | - Anne L Carlton
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| | - Arkadi Manukyan
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| | - Ann E Sutherland
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - David Wotton
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
9
|
Wang Y, Pan T, Wang H, Li L, Li J, Zhang C, Yang H. Silencing of TGIF attenuates the tumorigenicity of A549 cells in vitro and in vivo. Tumour Biol 2016; 37:12725-12730. [PMID: 27448304 DOI: 10.1007/s13277-016-5222-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/14/2016] [Indexed: 12/29/2022] Open
Abstract
The aim of this study was to investigate the effects of the silencing of the TG-interacting factor (TGIF) on the tumorigenicity of A549 cells in vitro and in vivo. Stable TGIF-silenced A549 cells were established by infecting shRNA lentiviral particles. Western blotting analysis was used to detect the expression of proteins. Cell cycle was detected by flow cytometry. Soft agar assay and tumor formation assay in nude mice were applied. The silencing of TGIF inhibited A549 cell proliferation, colony formation in vitro, growth of tumor xenograft in vivo, and arrested the cell cycle in the G1 phase. The expression of CDK4, cyclin D1, and phospho-Rb was markedly decreased in the A549-shTGIF cells compared with the A549-shcon cells, and p21 was markedly increased in the A549-shTGIF cells compared with the A549-shcon cells. A lower level of β-Catenin protein expression was observed in the A549-shTGIF cells than that in the A549-shcon cells. The silencing of TGIF attenuates the tumorigenicity of A549 cells in vitro and in vivo.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China.
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Teng Pan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Congke Zhang
- Department of Toxicology, Henan Center for Disease Control and Prevention, No. 105 of South Nongye Road, Zhengzhou, 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
10
|
Abstract
Tgif1 and Tgif2 are transcriptional repressors that inhibit the transcriptional response to transforming growth factor β signaling, and can repress gene expression by direct binding to DNA. Loss of function mutations in TGIF1 are associated with holoprosencephaly (HPE) in humans. In mice, embryos lacking both Tgif1 and Tgif2 fail to complete gastrulation, and conditional double null embryos that survive past gastrulation have HPE and do not survive past mid-gestation. Here we show that in mice of a relatively pure C57BL/6 strain background, loss of Tgif1 alone results in defective axial patterning and altered expression of Hoxc6. The primary defects in Tgif1 null embryos are the presence of extra ribs on the C7 vertebra, consistent with a posterior transformation phenotype. In addition we observed defective cervical vertebrae, primarily C1-C5, in both adult mice and embryos that lacked Tgif1. The combination of Tgif1 and Tgif2 mutations increases the severity and penetrance of the posterior transformation phenotype, without altering the type of defects seen. Similarly, exposure of Tgif1 mutant embryos to retinoic acid at E8.5 increased the severity and penetrance of the Tgif1 phenotype. This suggests that Tgif1 and Tgif2 regulate axial patterning and that reduced TGIF function sensitizes embryos to the effects of retinoic acid.
Collapse
|
11
|
Lee BK, Shen W, Lee J, Rhee C, Chung H, Kim KY, Park IH, Kim J. Tgif1 Counterbalances the Activity of Core Pluripotency Factors in Mouse Embryonic Stem Cells. Cell Rep 2015; 13:52-60. [PMID: 26411691 DOI: 10.1016/j.celrep.2015.08.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 07/24/2015] [Accepted: 08/24/2015] [Indexed: 11/16/2022] Open
Abstract
Core pluripotency factors, such as Oct4, Sox2, and Nanog, play important roles in maintaining embryonic stem cell (ESC) identity by autoregulatory feedforward loops. Nevertheless, the mechanism that provides precise control of the levels of the ESC core factors without indefinite amplification has remained elusive. Here, we report the direct repression of core pluripotency factors by Tgif1, a previously known terminal repressor of TGFβ/activin/nodal signaling. Overexpression of Tgif1 reduces the levels of ESC core factors, whereas its depletion leads to the induction of the pluripotency factors. We confirm the existence of physical associations between Tgif1 and Oct4, Nanog, and HDAC1/2 and further show the level of Tgif1 is not significantly altered by treatment with an activator/inhibitor of the TGFβ/activin/nodal signaling. Collectively, our findings establish Tgif1 as an integral member of the core regulatory circuitry of mouse ESCs that counterbalances the levels of the core pluripotency factors in a TGFβ/activin/nodal-independent manner.
Collapse
Affiliation(s)
- Bum-Kyu Lee
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, the University of Texas at Austin, Austin, TX 78712, USA
| | - Wenwen Shen
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA
| | - Jiwoon Lee
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, the University of Texas at Austin, Austin, TX 78712, USA
| | - Catherine Rhee
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, the University of Texas at Austin, Austin, TX 78712, USA
| | - Haewon Chung
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, the University of Texas at Austin, Austin, TX 78712, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, 10 Amistad, 201B, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, 10 Amistad, 201B, New Haven, CT 06520, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, Center for Systems and Synthetic Biology, the University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
12
|
Sharma A, Sinha NR, Siddiqui S, Mohan RR. Role of 5'TG3'-interacting factors (TGIFs) in Vorinostat (HDAC inhibitor)-mediated Corneal Fibrosis Inhibition. Mol Vis 2015; 21:974-84. [PMID: 26330748 PMCID: PMC4551282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/26/2015] [Indexed: 11/25/2022] Open
Abstract
PURPOSE We have previously reported that vorinostat, an FDA-approved, clinically used histone deacetylase (HDAC) inhibitor, attenuates corneal fibrosis in vivo in rabbits by blocking transforming growth factor β (TGFβ). The 5'TG3'-interacting factors (TGIFs) are transcriptional repressors of TGFβ1 signaling via the Smad pathway. The present study was designed to explore the expression of TGIFs in human corneal fibroblasts and to investigate their role in mediating the antifibrotic effect of vorinostat. METHODS Human corneal fibroblast cultures were generated from donor corneas. RNA isolation, cDNA preparation, and PCR were performed to detect the presence of TGIF1 and TGIF2 transcripts. The cultures were exposed to vorinostat (2.5 µM) to test its effect on TGIF mRNA and protein levels using qPCR and immunoblotting. Myofibroblast formation was induced with TGFβ1 (5 ng/ml) treatment under serum-free conditions. The changes in fibrosis parameters were quantified by measuring fibrosis marker α-smooth muscle actin (αSMA) mRNA and protein levels with qPCR, immunostaining, and immunoblotting. Smad2/3/4 and TGIF knockdowns were performed using pre-validated RNAi/siRNAs and a commercially available transfection reagent. RESULTS Human corneal fibroblasts showed the expression of TGIF1 and TGIF2. Vorinostat (2.5 µM) caused a 2.8-3.3-fold increase in TGIF1 and TGIF2 mRNA levels and a 1.4-1.8-fold increase in TGIF1 and TGIF2 protein levels. Vorinostat treatment also caused a significant increase in acetylhistone H3 and acetylhistone H4. Vorinostat-induced increases in TGIF1 and TGIF2 were accompanied by a concurrent decrease in corneal fibrosis, as indicated by a decrease in αSMA mRNA by 83±7.7% and protein levels by 97±5%. The RNAi-mediated knockdown of Smad2, Smad3, and Smad4 markedly attenuated TGFβ1-evoked transdifferentiation of fibroblasts to myofibroblasts. The siRNA-mediated knockdown of TGIF1 and TGIF2 neutralized vorinostat-evoked decreases in αSMA mRNA by 31%-45% and protein levels by 12%-23%. CONCLUSIONS Human corneal fibroblasts demonstrate the expression of TGIF1 and TGIF2 transcription factors. These transcriptional repressors are critical, at least partially, in mediating the antifibrotic effect of vorinostat in the cornea.
Collapse
Affiliation(s)
- Ajay Sharma
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO,Veterinary Medicine and Surgery, University of Missouri, Columbia, MO
| | - Nishant R. Sinha
- Veterinary Medicine and Surgery, University of Missouri, Columbia, MO
| | - Saad Siddiqui
- Veterinary Medicine and Surgery, University of Missouri, Columbia, MO
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO,Mason Eye Institute, School of Medicine, Columbia, MO,Veterinary Medicine and Surgery, University of Missouri, Columbia, MO
| |
Collapse
|
13
|
Chio CC, Chang CP, Lin MT, Su FC, Yang CZ, Tseng HY, Liu ZM, Huang HS. Involvement of TG-interacting factor in microglial activation during experimental traumatic brain injury. J Neurochem 2014; 131:816-24. [PMID: 25319900 DOI: 10.1111/jnc.12971] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 09/13/2014] [Accepted: 10/06/2014] [Indexed: 11/28/2022]
Abstract
Traumatic brain injury (TBI) is a complex injury involving several physiological alterations, potentially leading to neurological impairment. Previous mouse studies using high-density oligonucleotide array analysis have confirmed the upregulation of transforming growth-interacting factor (TGIF) mRNA in TBI. TGIF is a transcriptional corepressor of transforming growth factor beta (TGF-β) signaling which plays a protective role in TBI. However, the functional roles of TGIF in TBI are not well understood. In this study, we used confocal microscopy after immunofluorescence staining to demonstrate the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex significantly downregulated TGIF expression, attenuated microglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. Collectively, our results indicate that TGIF is involved in TBI-induced microglial activation, resulting in secondary brain injury and motor dysfunction. This study investigated the roles of transforming growth-interacting factor (TGIF) in a traumatic brain injury (TBI)-rat model. We demonstrated the increase of TGIF levels in the activated microglia of the pericontusional cortex of rats with TBI. Intracerebral knockdown of TGIF in the pericontusional cortex of the TBI rats significantly attenuated micoglial activation, reduced the volume of damaged brain tissue, and facilitated recovery of limb motor function. We suggest that inhibition of TGIF might provide a promising therapeutic strategy for TBI.
Collapse
|
14
|
Pramfalk C, Eriksson M, Parini P. Role of TG-interacting factor (Tgif) in lipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:9-12. [PMID: 25088698 DOI: 10.1016/j.bbalip.2014.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/17/2014] [Accepted: 07/24/2014] [Indexed: 11/18/2022]
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 (LXRs) and peroxisome proliferator-activated receptors (PPARs)], 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.
Collapse
Affiliation(s)
- Camilla Pramfalk
- Division of Clinical Chemistry, Department of Laboratory Medicine, Centre for Nutrition and Toxicology, NOVUM, Karolinska Institutet at Karolinska University Hospital Huddinge, Sweden; Molecular Nutrition Unit, Department of Biosciences and Nutrition, Centre for Nutrition and Toxicology, NOVUM, Karolinska Institutet at Karolinska University Hospital Huddinge, Sweden
| | - Mats Eriksson
- Molecular Nutrition Unit, Department of Biosciences and Nutrition, Centre for Nutrition and Toxicology, NOVUM, Karolinska Institutet at Karolinska University Hospital Huddinge, Sweden; Metabolism Unit, Department of Endocrinology, Metabolism and Diabetes, and Department of Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, S-141 86 Stockholm, Sweden
| | - Paolo Parini
- Division of Clinical Chemistry, Department of Laboratory Medicine, Centre for Nutrition and Toxicology, NOVUM, Karolinska Institutet at Karolinska University Hospital Huddinge, Sweden; Molecular Nutrition Unit, Department of Biosciences and Nutrition, Centre for Nutrition and Toxicology, NOVUM, Karolinska Institutet at Karolinska University Hospital Huddinge, Sweden.
| |
Collapse
|
15
|
Cerdá-Esteban N, Spagnoli FM. Glimpse into Hox and tale regulation of cell differentiation and reprogramming. Dev Dyn 2013; 243:76-87. [PMID: 24123411 DOI: 10.1002/dvdy.24075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 09/15/2013] [Accepted: 10/04/2013] [Indexed: 12/20/2022] Open
Abstract
During embryonic development, cells become gradually restricted in their developmental potential and start elaborating lineage-specific transcriptional networks to ultimately acquire a unique differentiated state. Hox genes play a central role in specifying regional identities, thereby providing the cell with critical information on positional value along its differentiation path. The exquisite DNA-binding specificity of the Hox proteins is frequently dependent upon their interaction with members of the TALE family of homeodomain proteins. In addition to their function as Hox-cofactors, TALE homeoproteins control multiple crucial developmental processes through Hox-independent mechanisms. Here, we will review recent findings on the function of both Hox and TALE proteins in cell differentiation, referring mostly to vertebrate species. In addition, we will discuss the direct implications of this knowledge on cell plasticity and cell reprogramming.
Collapse
Affiliation(s)
- Nuria Cerdá-Esteban
- Laboratory of Molecular and Cellular Basis of Embryonic Development, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
16
|
Hneino M, Blirando K, Buard V, Tarlet G, Benderitter M, Hoodless P, François A, Milliat F. The TG-interacting factor TGIF1 regulates stress-induced proinflammatory phenotype of endothelial cells. J Biol Chem 2012; 287:38913-21. [PMID: 22995913 DOI: 10.1074/jbc.m112.388389] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The endothelium contributes to the control of the tissue inflammatory response following stress and in particular after exposure to ionizing radiation. We previously showed that the TG-interacting factor 1 (TGIF1) plays a role in radiation-induced normal tissue injury. In this study we hypothesized that this protein could play a role in inflammation. The role of TGIF1 in the stress-induced proinflammatory phenotype was investigated in human endothelial cells. In HUVECs ionizing radiation induces TGIF1 expression as well as a proinflammatory phenotype associated with up-regulation of IL-6, IL-8, CXCL1, MIP-2, and MCP-1. TGIF1 overexpression enhances the radiation-induced proinflammatory phenotype whereas TGIF1 silencing limits both the TNF-α- and radiation-induced overexpression of proinflammatory cytokines. Interestingly, in vivo, in radiation-induced intestinal inflammation in mice, TGIF1 genetic deficiency is associated with a reduced radiation-induced overexpression of proinflammatory molecules. In HUVECs, TNF-α- and radiation-induced NF-κB pathway activation is not influenced by TGIF1 expression, whereas TGIF1 knockdown inhibits both TNF-α- and radiation-induced p38 MAPK pathway activation. This study demonstrates that TGIF1 plays a role in TNF-α- and radiation-induced inflammation and suggests that it could be a target in limiting this event in the vascular compartment.
Collapse
Affiliation(s)
- Mohammad Hneino
- Laboratory of Radiopathology and Experimental Therapeutics, Institute for Radiological Protection and Nuclear Safety, 92262 Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Overexpression of TG-interacting factor is associated with worse prognosis in upper urinary tract urothelial carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1044-55. [PMID: 22771156 DOI: 10.1016/j.ajpath.2012.05.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/21/2012] [Accepted: 05/17/2012] [Indexed: 12/23/2022]
Abstract
Prognostic outcome prediction would be useful for the treatment of patients with upper urinary tract urothelial carcinoma (UC). However, its prognostic biomarkers are not well established so far. According to the results of analysis of 168 human upper urinary tract UC specimens, overexpressed TG-interacting factor (TGIF) in nuclei of tumor tissues is significantly correlated with poor progression-free survival and higher cancer-related death. When both TGIF and p21 expression are altered, these patients had an even worse prognosis than those with one or no marker altered. Furthermore, to elucidate the role of TGIF in the progression of UC, overexpression of TGIF in RT4 or TSGH8301 cells was performed, and the results revealed that TGIF can significantly increase migration/invasion ability, matrix metalloproteinase expression, and invadopodia formation via the phosphatidylinositol 3-kinase-AKT pathway. In contrast, knockdown of TGIF with its specific short hairpin RNA inhibited the invasion ability of T24 cells. Besides, TGIF could inhibit p21(WAF/CIP1) expression, up-regulate cyclin D1 expression, and phosphorylate retinoblastoma to promote G1-S transition and cellular proliferation. In conclusion, we demonstrated that TGIF contributes to the progression of urothelial carcinoma via the phosphatidylinositol 3-kinase-AKT pathway. It may serve as an attractive therapeutic or prognostic target for selected patients with upper urinary tract UC.
Collapse
|
18
|
Hneino M, François A, Buard V, Tarlet G, Abderrahmani R, Blirando K, Hoodless PA, Benderitter M, Milliat F. The TGF-β/Smad repressor TG-interacting factor 1 (TGIF1) plays a role in radiation-induced intestinal injury independently of a Smad signaling pathway. PLoS One 2012; 7:e35672. [PMID: 22567107 PMCID: PMC3342305 DOI: 10.1371/journal.pone.0035672] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/22/2012] [Indexed: 01/17/2023] Open
Abstract
Despite advances in radiation delivery protocols, exposure of normal tissues during the course of radiation therapy remains a limiting factor of cancer treatment. If the canonical TGF-β/Smad pathway has been extensively studied and implicated in the development of radiation damage in various organs, the precise modalities of its activation following radiation exposure remain elusive. In the present study, we hypothesized that TGF-β1 signaling and target genes expression may depend on radiation-induced modifications in Smad transcriptional co-repressors/inhibitors expressions (TGIF1, SnoN, Ski and Smad7). In endothelial cells (HUVECs) and in a model of experimental radiation enteropathy in mice, radiation exposure increases expression of TGF-β/Smad pathway and of its target gene PAI-1, together with the overexpression of Smad co-repressor TGIF1. In mice, TGIF1 deficiency is not associated with changes in the expression of radiation-induced TGF-β pathway-related transcripts following localized small intestinal irradiation. In HUVECs, TGIF1 overexpression or silencing has no influence either on the radiation-induced Smad activation or the Smad3-dependent PAI-1 overexpression. However, TGIF1 genetic deficiency sensitizes mice to radiation-induced intestinal damage after total body or localized small intestinal radiation exposure, demonstrating that TGIF1 plays a role in radiation-induced intestinal injury. In conclusion, the TGF-β/Smad co-repressor TGIF1 plays a role in radiation-induced normal tissue damage by a Smad-independent mechanism.
Collapse
Affiliation(s)
- Mohammad Hneino
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Agnes François
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Valerie Buard
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Georges Tarlet
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Rym Abderrahmani
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Karl Blirando
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Pamela A. Hoodless
- Terry Fox Laboratory, British Columbia Cancer Agency and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Marc Benderitter
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Fabien Milliat
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
- * E-mail:
| |
Collapse
|
19
|
Zerlanko BJ, Bartholin L, Melhuish TA, Wotton D. Premature senescence and increased TGFβ signaling in the absence of Tgif1. PLoS One 2012; 7:e35460. [PMID: 22514746 PMCID: PMC3325954 DOI: 10.1371/journal.pone.0035460] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 03/19/2012] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor β (TGFβ) signaling regulates cell cycle progression in several cell types, primarily by inducing a G1 cell cycle arrest. Tgif1 is a transcriptional corepressor that limits TGFβ responsive gene expression. Here we demonstrate that primary mouse embryo fibroblasts (MEFs) lacking Tgif1 proliferate slowly, accumulate increased levels of DNA damage, and senesce prematurely. We also provide evidence that the effects of loss of Tgif1 on proliferation and senescence are not limited to primary cells. The increased DNA damage in Tgif1 null MEFs can be partially reversed by culturing cells at physiological oxygen levels, and growth in normoxic conditions also partially rescues the proliferation defect, suggesting that in the absence of Tgif1 primary MEFs are less able to cope with elevated levels of oxidative stress. Additionally, we show that Tgif1 null MEFs are more sensitive to TGFβ-mediated growth inhibition, and that treatment with a TGFβ receptor kinase inhibitor increases proliferation of Tgif1 null MEFs. Conversely, persistent treatment of wild type cells with low levels of TGFβ slows proliferation and induces senescence, suggesting that TGFβ signaling also contributes to cellular senescence. We suggest that in the absence of Tgif1, a persistent increase in TGFβ responsive transcription and a reduced ability to deal with hyperoxic stress result in premature senescence in primary MEFs.
Collapse
Affiliation(s)
| | | | | | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
20
|
Taniguchi K, Anderson AE, Sutherland AE, Wotton D. Loss of Tgif function causes holoprosencephaly by disrupting the SHH signaling pathway. PLoS Genet 2012; 8:e1002524. [PMID: 22383895 PMCID: PMC3285584 DOI: 10.1371/journal.pgen.1002524] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 12/21/2011] [Indexed: 01/23/2023] Open
Abstract
Holoprosencephaly (HPE) is a severe human genetic disease affecting craniofacial development, with an incidence of up to 1/250 human conceptions and 1.3 per 10,000 live births. Mutations in the Sonic Hedgehog (SHH) gene result in HPE in humans and mice, and the Shh pathway is targeted by other mutations that cause HPE. However, at least 12 loci are associated with HPE in humans, suggesting that defects in other pathways contribute to this disease. Although the TGIF1 (TG-interacting factor) gene maps to the HPE4 locus, and heterozygous loss of function TGIF1 mutations are associated with HPE, mouse models have not yet explained how loss of Tgif1 causes HPE. Using a conditional Tgif1 allele, we show that mouse embryos lacking both Tgif1 and the related Tgif2 have HPE-like phenotypes reminiscent of Shh null embryos. Eye and nasal field separation is defective, and forebrain patterning is disrupted in embryos lacking both Tgifs. Early anterior patterning is relatively normal, but expression of Shh is reduced in the forebrain, and Gli3 expression is up-regulated throughout the neural tube. Gli3 acts primarily as an antagonist of Shh function, and the introduction of a heterozygous Gli3 mutation into embryos lacking both Tgif genes partially rescues Shh signaling, nasal field separation, and HPE. Tgif1 and Tgif2 are transcriptional repressors that limit Transforming Growth Factor β/Nodal signaling, and we show that reducing Nodal signaling in embryos lacking both Tgifs reduces the severity of HPE and partially restores the output of Shh signaling. Together, these results support a model in which Tgif function limits Nodal signaling to maintain the appropriate output of the Shh pathway in the forebrain. These data show for the first time that Tgif1 mutation in mouse contributes to HPE pathogenesis and provide evidence that this is due to disruption of the Shh pathway. Holoprosencephaly (HPE) is a devastating genetic disease affecting human brain development. HPE affects more than 1/8,000 live births and up to 1/250 conceptions. Several genetic loci are associated with HPE, and the mutated genes have been identified at some. We have analyzed the role of the TGIF1 gene, which is present at one of these loci (the HPE4 locus) and is mutated in a subset of human HPE patients. We show that Tgif1 mutations in mice cause HPE when combined with a mutation in the closely related Tgif2 gene. This provides the first evidence from model organisms that TGIF1 is in fact the gene at the HPE4 locus that causes HPE when mutated. The Sonic Hedgehog signaling pathway is the best understood pathway in the pathogenesis of HPE, and mutation of the Sonic Hedgehog gene in both humans and mice causes HPE. We show that mutations in Tgif1 and Tgif2 in mice cause HPE by disrupting the Sonic Hedgehog signaling pathway, further emphasizing the importance of this pathway for normal brain development. Thus we confirm TGIF1 as an HPE gene and provide genetic evidence that Tgif1 mutations cause HPE by disrupting the interplay of the Nodal and Sonic Hedgehog pathways.
Collapse
Affiliation(s)
- Kenichiro Taniguchi
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anoush E. Anderson
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ann E. Sutherland
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
21
|
Gongal PA, French CR, Waskiewicz AJ. Aberrant forebrain signaling during early development underlies the generation of holoprosencephaly and coloboma. Biochim Biophys Acta Mol Basis Dis 2010; 1812:390-401. [PMID: 20850526 DOI: 10.1016/j.bbadis.2010.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 09/08/2010] [Indexed: 01/10/2023]
Abstract
In this review, we highlight recent literature concerning the signaling mechanisms underlying the development of two neural birth defects, holoprosencephaly and coloboma. Holoprosencephaly, the most common forebrain defect, occurs when the cerebral hemispheres fail to separate and is typically associated with mispatterning of embryonic midline tissue. Coloboma results when the choroid fissure in the eye fails to close. It is clear that Sonic hedgehog (Shh) signaling regulates both forebrain and eye development, with defects in Shh, or components of the Shh signaling cascade leading to the generation of both birth defects. In addition, other intercellular signaling pathways are known factors in the incidence of holoprosencephaly and coloboma. This review will outline recent advances in our understanding of forebrain and eye embryonic pattern formation, with a focus on zebrafish studies of Shh and retinoic acid pathways. Given the clear overlap in the mechanisms that generate both diseases, we propose that holoprosencephaly and coloboma can represent mild and severe aspects of single phenotypic spectrum resulting from aberrant forebrain development. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.
Collapse
Affiliation(s)
- Patricia A Gongal
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
22
|
Powers SE, Taniguchi K, Yen W, Melhuish TA, Shen J, Walsh CA, Sutherland AE, Wotton D. Tgif1 and Tgif2 regulate Nodal signaling and are required for gastrulation. Development 2010; 137:249-59. [PMID: 20040491 DOI: 10.1242/dev.040782] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tgif1 and Tgif2 are transcriptional co-repressors that limit the response to TGFbeta signaling and play a role in regulating retinoic-acid-mediated gene expression. Mutations in human TGIF1 are associated with holoprosencephaly, but it is unclear whether this is a result of deregulation of TGFbeta/Nodal signaling, or of effects on other pathways. Surprisingly, mutation of Tgif1 in mice results in only relatively mild developmental phenotypes in most strain backgrounds. Here, we show that loss-of-function mutations in both Tgif1 and Tgif2 result in a failure of gastrulation. By conditionally deleting Tgif1 in the epiblast, we demonstrate that a single wild-type allele of Tgif1 in the extra-embryonic tissue allows the double null embryos to gastrulate and begin organogenesis, suggesting that extra-embryonic Tgif function is required for patterning the epiblast. Genetically reducing the dose of Nodal in embryos lacking all Tgif function results in partial rescue of the gastrulation defects. Conditional double null embryos have defects in left-right asymmetry, which are also alleviated by reducing the dose of Nodal. Together, these data show that Tgif function is required for gastrulation, and provide the first clear evidence that Tgifs limit the transcriptional response to Nodal signaling during early embryogenesis.
Collapse
Affiliation(s)
- Shannon E Powers
- Department of Biochemistry and Molecular Genetics, and Center for Cell Signaling, University of Virginia, Box 800577, HSC, Charlottesville VA 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Huang HS, Liu ZM, Hong DY. Blockage of JNK pathway enhances arsenic trioxide-induced apoptosis in human keratinocytes. Toxicol Appl Pharmacol 2010; 244:234-41. [PMID: 20074581 DOI: 10.1016/j.taap.2009.12.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 12/25/2009] [Accepted: 12/30/2009] [Indexed: 12/12/2022]
Abstract
Arsenic is well known as a carcinogen predisposing humans to some severe diseases and also as an effective medicine for treating acute promyelocytic leukemia, syphilis, and psoriasis. Multiple active mechanisms, including cell cycle arrest and apoptosis, have been proposed in therapy; however, the opposing effects of arsenic remain controversial. Our previous study found that arsenic trioxide (ATO)-induced activation of p21(WAF1/CIP1) (p21) led to A431 cell death through the antagonistic effects of the signaling of ERK1/2 and JNK1. In the current study, the inhibitory effects of JNK1 on ATO-induced p21 expression were explored. Over-expression of JNK1 in A431 cells could inhibit p21 expression, which was associated with HDAC1 and TGIF. Using the GST pull-down assay and fluorescence resonance energy transfer analysis, N-terminal domain (amino acids 1-108) of TGIF, critical to its binding with c-Jun, was found. Using reporter assays, requirement of the C-terminal domain (amino acids 138-272) of TGIF to suppress ATO-induced p21 expression was observed. Thus, the domains of TGIF that carried out its inhibitory effects on p21 were identified. Finally, treatment with JNK inhibitor SP600125 could enhance ATO-induced apoptosis of HaCaT keratinocytes by using flow cytometry.
Collapse
Affiliation(s)
- Huei-Sheng Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| | | | | |
Collapse
|
24
|
Hamid R, Brandt SJ. Transforming growth-interacting factor (TGIF) regulates proliferation and differentiation of human myeloid leukemia cells. Mol Oncol 2009; 3:451-63. [PMID: 19699159 PMCID: PMC5527533 DOI: 10.1016/j.molonc.2009.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 06/29/2009] [Accepted: 07/13/2009] [Indexed: 11/26/2022] Open
Abstract
Transforming growth-interacting factor (TGIF) is a homeobox transcriptional repressor that has been implicated in holoprosencephaly and various types of cancer. TGIF is expressed in hematopoietic stem cells and modulates TGF-beta and retinoic acid (RA) signaling, both of which play an important role in hematopoiesis. We recently reported that TGIF's levels correlate inversely with survival in patients with acute myelogenous leukemia. Here we present the first direct evidence of a role for TGIF in myelopoiesis. We used short hairpin RNA interference to define the effects of TGIF knockdown on proliferation and differentiation of myeloid leukemia-derived cell lines. Decreased TGIF expression resulted in reduced proliferation and differentiation and lower expression of CEBPbeta, CEBPepsilon, PU.1 and RUNX1, key myeloid transcription factors. Furthermore, TGF-beta signaling was increased and RA signaling was decreased. Further insights into the molecular basis of TGIF's effects were provided by a genome-wide chromatin immunoprecipitation-based elucidation of TGIF target genes. Together, these data suggest that TGIF has an important role myelopoiesis and may regulate the balance between proliferation and differentiation. Reduced TGIF expression could tip the balance toward quiescence thus providing progenitor as well as hematopoietic stem cells protection from anti-cycle agents.
Collapse
Affiliation(s)
- Rizwan Hamid
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, United States.
| | | |
Collapse
|
25
|
Kerr TC, Cuykendall TN, Luettjohann LC, Houston DW. Maternal Tgif1 regulates nodal gene expression in Xenopus. Dev Dyn 2008; 237:2862-73. [PMID: 18816846 DOI: 10.1002/dvdy.21707] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In Xenopus, the maternal transcription factor VegT is necessary and sufficient to initiate the expression of nodal-related genes, which are central to many aspects of early development. However, little is known about regulation of VegT activity. Using maternal loss-of-function experiments, we show that the maternal homeoprotein, Tgif1, antagonizes VegT and plays a central role in anteroposterior patterning by negatively regulating a subset of nodal-related genes. Depletion of Tgif1 causes the anteriorization of embryos and the up-regulation of nodal paralogues nr5 and nr6. Furthermore, Tgif1 inhibits activation of nr5 by VegT in a manner that requires a C-terminal Sin3 corepressor-interacting domain. Tgif1 has been implicated in the transcriptional corepression of transforming growth factor-beta (TGFbeta) and retinoid signaling. However, we show that Tgif1 does not inhibit these pathways in early development. These results identify an essential role for Tgif1 in the control of nodal expression and provide insight into Tgif1 function and mechanisms controlling VegT activity.
Collapse
Affiliation(s)
- Tyler C Kerr
- University of Iowa, Department of Biology, Iowa City, Iowa 52246-1324, USA
| | | | | | | |
Collapse
|
26
|
Satoh S, Watanabe S. TGIF, a homeodomain transcription factor, regulates retinal progenitor cell differentiation. Exp Eye Res 2008; 87:571-9. [PMID: 18926818 DOI: 10.1016/j.exer.2008.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2008] [Revised: 09/09/2008] [Accepted: 09/12/2008] [Indexed: 12/29/2022]
Abstract
TG-interacting factor (TGIF) is a TALE homeodomain protein expressed predominantly in the central nervous system and functions as a transcriptional repressor. Several mutations in TGIF have been identified in patients with holoprosencephaly, the most common congenital malformation of the developing human forebrain. However, the precise role of TGIF in neural development is not well understood. We found that TGIF was expressed strongly in the mouse retina during early stages of development, and that its expression gradually decreased as retinal development progressed. In vitro explant cultures of mouse retina mimic the in vivo development of retinal subtypes. Forced expression of TGIF using a retrovirus in explant culture induced the differentiation of amacrine cells from retinal progenitor cells. A TGIF paralog, TGIF2, showed a similar transition in expression during retinal development, and TGIF2 also promoted amacrine cell differentiation in a retinal explant culture system. However, no apparent difference between wild-type and TGIF-knockout mouse retina was observed, suggesting that TGIF and TGIF2 function redundantly in that tissue. Forced expression of TGIF homeodomain (HD)-EnR (repressing) rather than TGIF HD-VP16 (activating) resulted in a phenotype similar to that induced by wild-type TGIF, suggesting that TGIFs may act as transcriptional repressors to induce amacrine genesis.
Collapse
Affiliation(s)
- Shinya Satoh
- Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | |
Collapse
|
27
|
Hsu LCL, Chen HY, Lin YW, Chu WC, Lin MJ, Yan YT, Yen PH. DAZAP1, an hnRNP protein, is required for normal growth and spermatogenesis in mice. RNA (NEW YORK, N.Y.) 2008; 14:1814-1822. [PMID: 18669443 PMCID: PMC2525968 DOI: 10.1261/rna.1152808] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Accepted: 05/23/2008] [Indexed: 05/26/2023]
Abstract
DAZAP1 (Deleted in Azoospermia Associated Protein 1) is a ubiquitous hnRNP protein that is expressed most abundantly in the testis. Its ability to shuttle between the nucleus and the cytoplasm and its exclusion from the transcriptionally inactive XY body in pachytene spermatocytes implicate it in mRNA transcription and transport. We generated Dazap1 mutant alleles to study the role of DAZAP1 in mouse development. Most mice homozygous for the null allele as well as a hypomorphic Fn allele died soon after birth. The few Dazap1(Fn/Fn) mice that survived could nonetheless live for more than a year. They appeared and behaved normally but were much smaller in size compared to their wild-type and heterozygous littermates. Both male and female Dazap1(Fn/Fn) mice were sterile. Males had small testes, and the seminiferous tubules were atrophic with increased numbers of apoptotic cells. The tubules contained many germ cells, including pachytene spermatocytes with visible XY-bodies and diplotene spermatocytes, but no post-meiotic cells. FACS analyses confirmed the absence of haploid germ cells, indicating spermatogenesis arrested right before the meiotic division. Female Dazap1(Fn/Fn) mice had small ovaries that contained normal-appearing follicles, yet their pregnancy produced no progeny due to failure in embryonic development. The phenotypes of Dazap1 mutant mice indicate that DAZAP1 is not only essential for spermatogenesis, but also required for the normal growth and development of mice.
Collapse
Affiliation(s)
- Lea Chia-Ling Hsu
- 1Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | | | | | | | | | | | | |
Collapse
|
28
|
Bartholin L, Melhuish TA, Powers SE, Goddard-Léon S, Treilleux I, Sutherland AE, Wotton D. Maternal Tgif is required for vascularization of the embryonic placenta. Dev Biol 2008; 319:285-97. [PMID: 18508043 PMCID: PMC2517231 DOI: 10.1016/j.ydbio.2008.04.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 04/15/2008] [Accepted: 04/16/2008] [Indexed: 11/25/2022]
Abstract
The mammalian placenta is the site of exchange of nutrients and waste between mother and embryo. In humans, placental insufficiency can result in intrauterine growth retardation, perinatal death and spontaneous abortion. We show that in C57BL/6J mice a null mutation in the gene encoding the transcriptional corepressor, Tgif, causes placental defects. The major defects are decreased vascularization of the placenta, due to a decrease in the fetal blood vessels, and decreased expression of the gap junction protein Gjb2 (Cx26). These defects result in severe growth retardation in a proportion of Tgif null embryos in Tgif heterozygous mothers, and an overall growth delay in Tgif null animals. Placental defects are much more severe if the mother also completely lacks Tgif function, and placentas from heterozygous Tgif embryos are defective in a Tgif null mother. Embryo transfer experiments show that even the placenta from a wild type embryo is compromised in the absence of maternal Tgif. These results demonstrate that Tgif functions in the normal development of the placenta, and suggest a role for maternal factors in regulating the morphogenesis of embryonically-derived placental tissues.
Collapse
Affiliation(s)
- Laurent Bartholin
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | - Tiffany A. Melhuish
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | - Shannon E. Powers
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | | | | | | | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| |
Collapse
|
29
|
Faresse N, Colland F, Ferrand N, Prunier C, Bourgeade MF, Atfi A. Identification of PCTA, a TGIF antagonist that promotes PML function in TGF-beta signalling. EMBO J 2008; 27:1804-15. [PMID: 18511908 DOI: 10.1038/emboj.2008.109] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Accepted: 05/07/2008] [Indexed: 11/09/2022] Open
Abstract
The TGIF homoeodomain protein functions as an important negative regulator in the TGF-beta signalling pathway. The inhibitory function of TGIF is executed in part through its ability to sequester the tumour suppressor cytoplasmic promyelocytic leukaemia (cPML) in the nucleus, thereby preventing the phosphorylation of Smad2 by the activated TGF-beta type I receptor. Here, we report on the identification of PCTA (PML competitor for TGIF association), a TGIF antagonist that promotes TGF-beta-induced transcriptional and cytostatic responses. We provide evidence that PCTA functions in TGF-beta signalling by relieving the suppression of Smad2 phosphorylation by TGIF. Furthermore, we demonstrate that PCTA selectively competes with cPML for TGIF association, resulting in the accumulation of cPML in the cytoplasm, where it associates with SARA and coordinates the access of Smad2 for phosphorylation by the activated TGF-beta type I receptor. Thus, our findings on the mode of action of PCTA provide new and important insights into the molecular mechanism underlying the antagonistic interplay between TGIF and cPML in the TGF-beta signalling network.
Collapse
Affiliation(s)
- Nourdine Faresse
- Laboratory of Cell Signaling and Carcinogenesis, INSERM U673, Paris, France
| | | | | | | | | | | |
Collapse
|
30
|
Schweikl H, Hiller KA, Eckhardt A, Bolay C, Spagnuolo G, Stempfl T, Schmalz G. Differential gene expression involved in oxidative stress response caused by triethylene glycol dimethacrylate. Biomaterials 2008; 29:1377-87. [DOI: 10.1016/j.biomaterials.2007.11.049] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Accepted: 11/26/2007] [Indexed: 12/31/2022]
|
31
|
Abstract
Holoprosencephaly (HPE), the most common developmental defect of the forebrain and midface, is caused by a failure to delineate the midline in these structures. Both genetic and environmental etiologies exist for HPE, and clinical presentation is highly variable. HPE occurs in sporadic and inherited forms, and even HPE in pedigrees is characterized by incomplete penetrance and variable expressivity. Heterozygous mutations in eight different genes have been identified in human HPE, and disruption of Sonic hedgehog expression and/or signaling in the rostroventral region of the embryo is a major common effect of these mutations. An understanding of the mechanisms whereby genetic defects and teratogenic exposures become manifest as developmental anomalies of varying severity requires experimental models that accurately reproduce the spectrum of defects seen in human HPE. The mouse has emerged as such a model, because of its ease of genetic manipulation and similarity to humans in development of the forebrain and face. HPE is generally observed in mice homozygous for mutations in orthologs of human HPE genes though, unlike humans, rarely in mice with heterozygous mutations. Moreover, reverse genetics in the mouse has provided a wealth of new candidate human HPE genes. Construction of hypomorphic alleles, interbreeding to produce double mutants, and analysis of these mutations on different genetic backgrounds has generated multiple models of HPE and begun to provide insight into the conundrum of the HPE spectrum. Here, we review forebrain development with an emphasis on the pathways known to be defective in HPE and describe the strengths and weaknesses of various murine models of HPE.
Collapse
Affiliation(s)
- Karen A Schachter
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York 10029, USA
| | | |
Collapse
|
32
|
Gongal PA, Waskiewicz AJ. Zebrafish model of holoprosencephaly demonstrates a key role for TGIF in regulating retinoic acid metabolism. Hum Mol Genet 2007; 17:525-38. [PMID: 17998248 DOI: 10.1093/hmg/ddm328] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Holoprosencephaly (HPE) is the most common human congenital forebrain defect, affecting specification of forebrain tissue and subsequent division of the cerebral hemispheres. The causes of HPE are multivariate and heterogeneous, and include exposure to teratogens, such as retinoic acid (RA), and mutations in forebrain patterning genes. Many of the defects in HPE patients resemble animal models with aberrant RA levels, which also show severe forebrain abnormalities. RA plays an important role in early neural patterning of the vertebrate embryo: expression of RA-synthesizing enzymes initiates high RA levels in the trunk, which are required for proper anterior-posterior patterning of the hindbrain and spinal cord. In the forebrain and midbrain, RA-degrading enzymes are expressed, protecting these regions from the effects of RA. However, the mechanisms that regulate RA-synthesizing and RA-degrading enzymes are poorly understood. Mutations in the gene TGIF are associated with incidence of HPE. We demonstrate in zebrafish that Tgif plays a key role in regulating RA signaling, and is essential to properly pattern the forebrain. Tgif is necessary for normal initiation of genes that control RA synthesis and degradation, resulting in defects in RA-dependent central nervous system patterning in Tgif-depleted embryos. The loss of the forebrain-specific RA-degrading enzyme cyp26a1 causes a forebrain phenotype that mimics tgif morphants. We propose a model in which Tgif controls forebrain patterning by regulating RA degradation. The consequences of abnormal RA levels for forebrain patterning are profound, and imply that in human patients with TGIF deficiencies, increased forebrain RA levels contribute to the development of HPE.
Collapse
Affiliation(s)
- Patricia A Gongal
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
33
|
Abstract
Holoprosencephaly (HPE) is a common congenital malformation that is characterised by a failure to divide the forebrain into left and right hemispheres and is usually accompanied by defects in patterning of the midline of the face. HPE exists in inherited, autosomal dominant (familial) forms and mutation-associated sporadic forms, but environmental factors are also implicated. There are several features of HPE that are not well understood, including the extremely variable clinical presentation, even among obligate carriers of familial mutations, and the restriction of structural anomalies to the ventral anterior midline, despite association with defects in signal transduction pathways that regulate development of many additional body structures. The new animal models described in this review may help unravel these puzzles. Furthermore, these model systems suggest that human HPE arises from a complex interaction between the timing and strength of developmental signalling pathways, genetic variation and exposure to environmental agents.
Collapse
Affiliation(s)
- Robert S Krauss
- Department of Molecular, Cell and Developmental Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
34
|
Gilner JB, Walton WG, Gush K, Kirby SL. Antibodies to Stem Cell Marker Antigens Reduce Engraftment of Hematopoietic Stem Cells. Stem Cells 2007; 25:279-88. [PMID: 17008427 DOI: 10.1634/stemcells.2006-0076] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cells (HSCs) have enormous potential for use in transplantation and gene therapy. However, the frequency of repopulating HSCs is often very low; thus, highly effective techniques for cell enrichment and maintenance are required to obtain sufficient cell numbers for therapeutic use and for studies of HSC physiology. Common methods of HSC enrichment use antibodies recognizing HSC surface marker antigens. Because antibodies are known to alter the physiology of other cell types, we investigated the effect of such enrichment strategies on the physiology and lineage commitment of HSCs. We sorted HSCs using a method that does not require antibodies: exclusion of Hoechst 33342 to isolate side population (SP) cells. To elucidate the effect of antibody binding on this HSC population, we compared untreated SP cells with SP cells treated with the Sca-1(+)c-Kit(+)Lin(-) (SKL) antibody cocktail prior to SP sorting. Our findings revealed that HSCs incubated with the antibody cocktail had decreased expression of the stem cell-associated genes c-Kit, Cd34, Tal-1, and Slamf1 relative to untreated SP cells or to cells treated with polyclonal isotype control antibodies. Moreover, SKL antibodies induced cycling in SP cells and diminished their ability to confer long-term hematopoietic engraftment in lethally irradiated mice. Taken together, these data suggest that antibody-based stem cell isolation procedures can have negative effects on HSC physiology.
Collapse
Affiliation(s)
- Jennifer B Gilner
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
35
|
El-Jaick KB, Powers SE, Bartholin L, Myers KR, Hahn J, Orioli IM, Ouspenskaia M, Lacbawan F, Roessler E, Wotton D, Muenke M. Functional analysis of mutations in TGIF associated with holoprosencephaly. Mol Genet Metab 2007; 90:97-111. [PMID: 16962354 PMCID: PMC1820763 DOI: 10.1016/j.ymgme.2006.07.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 07/26/2006] [Indexed: 11/19/2022]
Abstract
Holoprosencephaly (HPE) is the most common structural malformation of the forebrain and face in humans. Our current understanding of the pathogenesis of HPE attempts to integrate genetic susceptibility, evidenced by mutations in the known HPE genes, with the epigenetic influence of environmental factors. Mutations or deletions of the human TGIF gene have been associated with HPE in multiple population cohorts. Here we examine the functional effects of all previously reported mutations, and describe four additional variants. Of the eleven sequence variations in TGIF, all but four can be demonstrated to be functionally abnormal. In contrast, no potentially pathogenic sequence alterations were detected in the related gene TGIF2. These results provide further evidence of a role for TGIF in HPE and demonstrate the importance of functional analysis of putative disease-associated alleles.
Collapse
Affiliation(s)
- Kenia B. El-Jaick
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda 20892-3717 MD USA
| | - Shannon E. Powers
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | - Laurent Bartholin
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | - Kenneth R. Myers
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
- Cell and Developmental Biology Program, University of Virginia
| | - Jin Hahn
- Stanford University Medical School, Stanford, CA
| | - Ieda M. Orioli
- Laboratory of Congenital Malformations, University of Rio de Janeiro, Brazil
| | - Maia Ouspenskaia
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda 20892-3717 MD USA
| | - Felicitas Lacbawan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda 20892-3717 MD USA
| | - Erich Roessler
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda 20892-3717 MD USA
| | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda 20892-3717 MD USA
- Corresponding author: *Maximilian Muenke, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, 35 Convent Drive - MSC 3717, Building 35, Room 1B-203, Bethesda, MD 20892-3717, Tel.: (301) 402-8167, Fax.: (301) 480-7876,
| |
Collapse
|
36
|
Ferrand N, Demange C, Prunier C, Seo SR, Atfi A. A mechanism for mutational inactivation of the homeodomain protein TGIF in holoprosencephaly. FASEB J 2006; 21:488-96. [PMID: 17158784 DOI: 10.1096/fj.06-6423com] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The homeodomain protein TGIF functions as a negative modulator for multiple classes of transcription factors. Loss of function mutations in a single copy of TGIF result in holoprosencephaly, a developmental anomaly leading to severe forebrain and craniofacial malformations. However, the mechanisms by which these mutations disrupt the functions of TGIF remain to be elucidated. Here we show that a holoprosencephaly mutation (P63R) interferes with the ability of TGIF to act as a corepressor for c-Jun and Smad2, suggesting that this holoprosencephaly mutation may lead to a general defect in the TGIF protein. In fact, we observed that the P63R mutation affects folding of the TGIF protein, resulting in the disruption of the diffuse nuclear staining pattern characteristic of wild-type (WT) TGIF and the accumulation of TGIF in nuclear aggregates. We also show that the mutant TGIF.P63R is degraded more rapidly when compared with WT TGIF and that this degradation occurs through the ubiquitin-proteasome pathway. Furthermore, we observed that TGIF.P63R homodimerizes with WT TGIF to sequester it into nuclear aggregates and to enhance its ubiquitin-dependent degradation. These results reveal an important mechanism for the degradation of TGIF through the ubiquitin-proteasome pathway, whose deregulation might contribute to the development of human holoprosencephaly.
Collapse
Affiliation(s)
- Nathalie Ferrand
- INSERM U 673, Hôpital St-Antoine, 184 Rue du Faubourg St-Antoine, 75571, Paris Cedex 12, France
| | | | | | | | | |
Collapse
|
37
|
Kuang C, Xiao Y, Yang L, Chen Q, Wang Z, Conway SJ, Chen Y. Intragenic deletion of Tgif causes defectsin brain development. Hum Mol Genet 2006; 15:3508-19. [PMID: 17082251 DOI: 10.1093/hmg/ddl427] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
TG-interacting factor (TGIF) is a homeodomain-containing protein and functions as a transcriptional repressor within the TGF-beta and retinoic acid signaling pathways. Heterozygous mutations of TGIF have been found in patients with holoprosencephaly (HPE), which is the most common congenital brain malformation in humans. However, targeted null deletions of the entire Tgif gene in mice surprisingly revealed no apparent brain defects. We report here that deletion of the third exon of Tgif gene resulted in a defined spectrum of brain developmental defects including exencephaly, microcephaly, HPE, and abnormalities in embryonic brain ventricle formation and cleavage. These defects could be detected in mice both heterozygous and homozygous for the targeted Tgif deletion. Moreover, expression of dorsal-ventral patterning genes including Shh, Pax6 and Nkx2.2 was altered. The ventricular neuroepithelium exhibited focalized increase of cell proliferation rate and resultant tissue expansion. The incidence of brain abnormalities within the mutant mice was dependent on its genetic background, suggesting that additional genetic modifiers functionally interact with Tgif during embryonic brain development. The intragenic Tgif deletion mouse, therefore, would serve as a useful model that can be used to unravel the genetic complexity implicated in the pathogenesis of HPE.
Collapse
Affiliation(s)
- Chenzhong Kuang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine and the Walther Cancer Institute, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|