1
|
Zhang S, He W, Li A, Zhao C, Chen Y, Xu C, Zhang Q, Zheng D, Chen M, Miao H, Huang Y. Involvement of the TNF-α/SATB2 axis in the induced apoptosis and inhibited autophagy of osteoblasts by the antipsychotic Risperidone. Mol Med 2022; 28:46. [PMID: 35505281 PMCID: PMC9066868 DOI: 10.1186/s10020-022-00466-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Risperidone, an atypical antipsychotic, impedes serotonin and dopamine receptor systems. Meanwhile, tumor necrosis factor-α (TNF-α) is known to participate in regulating osteoblast functions. Consequently, the current study aimed to investigate whether the influences of Risperidone on osteoblast functions are associated with TNF-α and special AT-rich sequence-binding protein (SATB2). METHODS Firstly, we searched the DGIdb, MEM and GeneCards databases to identify the critical factors involved in the effects of Risperidone on osteoblasts, as well as their interactions. Afterwards, osteoblast cell line MC3T3-E1 was transduced with lentivirus carrying si-TNF-α, si-SATB2 or both and subsequently treated with Risperidone. Various abilities including differentiation, autophagy and apoptosis of osteoblasts were examined after different treatments. Finally, animal experiments were performed with Risperidone alone or together with lentivirus to verify the function of Risperidone in vivo and the mechanism. RESULTS It was found that Risperidone might promote TNF-α expression, thereby inhibiting the expression of SATB2 to affect the autophagy and apoptosis in osteoblasts. Furthermore, as shown by our experimental findings, Risperidone treatment inhibited the differentiation and autophagy, and promoted the apoptosis of osteoblasts, as evidenced by elevated levels of OPG, p62, cleaved PARP1, cleaved caspase-3, cleaved caspase-8, and cleaved caspase-9, and reduced levels of LC3 II/I, Beclin1, collagen I, and RANKL. In addition, Risperidone was also found to elevate the expression of TNF-α to down-regulate SATB2, thereby inhibiting the differentiation and autophagy and enhancing the apoptosis of osteoblasts in vitro and in vivo. CONCLUSIONS Collectively, our findings indicated that Risperidone affects the differentiation of osteoblasts by inhibiting autophagy and enhancing apoptosis via TNF-α-mediated down-regulation of SATB2.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Wei He
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Aiguo Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Qiuzhen Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Danling Zheng
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Meini Chen
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Haixiong Miao
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pediatrics, Guangzhou Red Cross Hospital, Jinan University, No. 396, Tongfuzhong Road, Haizhu District, Guangzhou, 510220, Guangdong, China
| | - Yihui Huang
- Department of Pediatrics, Shantou University Medical College, Shantou, 515041, China
- Department of Pediatrics, Guangzhou Red Cross Hospital, Jinan University, No. 396, Tongfuzhong Road, Haizhu District, Guangzhou, 510220, Guangdong, China
| |
Collapse
|
2
|
Programmed suppression of oxidative phosphorylation and mitochondrial function by gestational alcohol exposure correlate with widespread increases in H3K9me2 that do not suppress transcription. Epigenetics Chromatin 2021; 14:27. [PMID: 34130715 PMCID: PMC8207718 DOI: 10.1186/s13072-021-00403-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 06/08/2021] [Indexed: 12/15/2022] Open
Abstract
Background A critical question emerging in the field of developmental toxicology is whether alterations in chromatin structure induced by toxicant exposure control patterns of gene expression or, instead, are structural changes that are part of a nuclear stress response. Previously, we used a mouse model to conduct a three-way comparison between control offspring, alcohol-exposed but phenotypically normal animals, and alcohol-exposed offspring exhibiting craniofacial and central nervous system structural defects. In the cerebral cortex of animals exhibiting alcohol-induced dysgenesis, we identified a dramatic increase in the enrichment of dimethylated histone H3, lysine 9 (H3K9me2) within the regulatory regions of key developmental factors driving histogenesis in the brain. However, whether this change in chromatin structure is causally involved in the development of structural defects remains unknown. Results Deep-sequencing analysis of the cortex transcriptome reveals that the emergence of alcohol-induced structural defects correlates with disruptions in the genetic pathways controlling oxidative phosphorylation and mitochondrial function. The majority of the affected pathways are downstream targets of the mammalian target of rapamycin complex 2 (mTORC2), indicating that this stress-responsive complex plays a role in propagating the epigenetic memory of alcohol exposure through gestation. Importantly, transcriptional disruptions of the pathways regulating oxidative homeostasis correlate with the emergence of increased H3K9me2 across genic, repetitive, and non-transcribed regions of the genome. However, although associated with gene silencing, none of the candidate genes displaying increased H3K9me2 become transcriptionally repressed, nor do they exhibit increased markers of canonical heterochromatin. Similar to studies in C. elegans, disruptions in oxidative homeostasis induce the chromatin looping factor SATB2, but in mammals, this protein does not appear to drive increased H3K9me2 or altered patterns of gene expression. Conclusions Our studies demonstrate that changes in H3K9me2 associate with alcohol-induced congenital defects, but that this epigenetic change does not correlate with transcriptional suppression. We speculate that the mobilization of SATB2 and increased enrichment of H3K9me2 may be components of a nuclear stress response that preserve chromatin integrity and interactions under prolonged oxidative stress. Further, we postulate that while this response may stabilize chromatin structure, it compromises the nuclear plasticity required for normal differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00403-w.
Collapse
|
3
|
Roy SK, Shrivastava A, Srivastav S, Shankar S, Srivastava RK. SATB2 is a novel biomarker and therapeutic target for cancer. J Cell Mol Med 2020; 24:11064-11069. [PMID: 32885593 PMCID: PMC7576221 DOI: 10.1111/jcmm.15755] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
Several studies have confirmed the involvement of cancer stem cells (CSC) in tumour progression, metastasis, drug resistance and cancer relapse. SATB2 (special AT-rich binding protein-2) acts as a transcriptional co-factor and modulates chromatin architecture to regulate gene expression. The purpose of this review was to discuss the pathophysiological roles of SATB2 and assess whether it could be used as a therapeutic target for cancer. SATB2 modulated the expression of those genes which regulated pluripotency and self-renewal. Overexpression of SATB2 gene in normal epithelial cells was shown to induce transformation, as a result transformed cells gained CSC's characteristics by expressing stem cell markers and pluripotency maintaining factors, suggesting its role as an oncogene. In addition, SATB2 induced epithelial-mesenchymal transition (EMT) and metastasis. Interestingly, the expression of SATB2 was positively correlated with the activation of β-catenin/TCF-LEF pathway. Furthermore, SATB2 silencing inhibited EMT and their positive regulators, and tumour growth, and suppressed the expression of stem cell markers, pluripotency maintaining factors, cell cycle and cell survival genes, and TCF/LEF targets. Based on the cancer genome atlas (TCGA) expression data and published papers, SATB2 alone or in combination with other proteins could be used a diagnostic biomarker for cancer. Although there is no pharmacological inhibitor of SATB2, studies using genetic approaches suggest that SATB2 could be a potential target for cancer treatment and prevention.
Collapse
Affiliation(s)
- Sanjit K. Roy
- Stanley S. Scott Cancer CenterLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | | | - Sudesh Srivastav
- Department of Biostatistics and Data ScienceSchool of Public Health and Tropical MedicineTulane University School of MedicineNew OrleansLAUSA
| | - Sharmila Shankar
- Stanley S. Scott Cancer CenterLouisiana State University Health Sciences CenterNew OrleansLAUSA
- Department of GeneticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
- John W. Deming Department of MedicineTulane University School of MedicineNew OrleansLAUSA
- Southeast Louisiana Veterans Health Care SystemNew OrleansLAUSA
| | - Rakesh K. Srivastava
- Stanley S. Scott Cancer CenterLouisiana State University Health Sciences CenterNew OrleansLAUSA
- Department of GeneticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| |
Collapse
|
4
|
Wu GJ, Chen JT, Cherng YG, Chang CC, Liu SH, Chen RM. Genistein Improves Bone Healing via Triggering Estrogen Receptor Alpha-Mediated Expressions of Osteogenesis-Associated Genes and Consequent Maturation of Osteoblasts. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:10639-10650. [PMID: 32897066 DOI: 10.1021/acs.jafc.0c02830] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Osteoporosis-associated fractures may cause higher morbidity and mortality. Our previous study showed the effects of genistein, a phytoestrogen, on the induction of estrogen receptor alpha (ERα) gene expression and stimulation of osteoblast mineralization. In this study, rat calvarial osteoblasts and an animal bone defect model were used to investigate the effects of genistein on bone healing. Treatment with genistein caused a time-dependent increase in alkaline phosphatase (ALP) activity in rat osteoblasts. Levels of cytosolic and nuclear ERα significantly augmented following exposure to genistein. Subsequently, genistein elevated levels of ALP mRNA and protein in rat osteoblasts. Moreover, genistein induced other osteogenesis-associated osteocalcin and Runx2 mRNA and protein expressions. Knocking-down ERα using RNA interference concurrently inhibited genistein-induced Runx2, osteocalcin, and ALP mRNA expression. Attractively, administration of ICR mice suffering bone defects with genistein caused significant increases in the callus width, chondrocyte proliferation, and ALP synthesis. Results of microcomputed tomography revealed that administration of genistein increased trabecular bone numbers and improved the bone thickness and volume. This study showed that genistein can improve bone healing via triggering ERα-mediated osteogenesis-associated gene expressions and subsequent osteoblast maturation.
Collapse
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jui-Tai Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yih-Giun Cherng
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chuen-Chau Chang
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Ruei-Ming Chen
- Anesthesiology and Health Policy Research Center, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
5
|
Zeng X, Tsui JCC, Shi M, Peng J, Cao CY, Kan LLY, Lau CPY, Liang Y, Wang L, Liu L, Chen Z, Tsui SKW. Genome-Wide Characterization of Host Transcriptional and Epigenetic Alterations During HIV Infection of T Lymphocytes. Front Immunol 2020; 11:2131. [PMID: 33013899 PMCID: PMC7511662 DOI: 10.3389/fimmu.2020.02131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background and methods: Host genomic alterations are closely related to dysfunction of CD4+ T lymphocytes in the HIV-host interplay. However, the roles of aberrant DNA methylation and gene expression in the response to HIV infection are not fully understood. We investigated the genome-wide DNA methylation and transcriptomic profiles in two HIV-infected T lymphocyte cell lines using high-throughput sequencing. Results: Based on DNA methylation data, we identified 3,060 hypomethylated differentially methylated regions (DMRs) and 2,659 hypermethylated DMRs in HIV-infected cells. Transcription-factor-binding motifs were significantly associated with methylation alterations, suggesting that DNA methylation modulates gene expression by affecting the binding to transcription factors during HIV infection. In support of this hypothesis, genes with promoters overlapping with DMRs were enriched in the biological function related to transcription factor activities. Furthermore, the analysis of gene expression data identified 1,633 upregulated genes and 2,142 downregulated genes on average in HIV-infected cells. These differentially expressed genes (DEGs) were significantly enriched in apoptosis-related pathways. Our results suggest alternative splicing as an additional mechanism that may contribute to T-cell apoptosis during HIV infection. We also demonstrated a genome-scale correlation between DNA methylation and gene expression in HIV-infected cells. We identified 831 genes with alterations in both DNA methylation and gene expression, which were enriched in apoptosis. Our results were validated using various experimental methods. In addition, consistent with our in silico results, a luciferase assay showed that the activity of the PDX1 and SMAD3 promoters was significantly decreased in the presence of HIV proteins, indicating the potential of these genes as genetic markers of HIV infection. Conclusions: Our results suggest important roles for DNA methylation and gene expression regulation in T-cell apoptosis during HIV infection. We propose a list of novel genes related to these processes for further investigation. This study also provides a comprehensive characterization of changes occurring at the transcriptional and epigenetic levels in T cells in response to HIV infection.
Collapse
Affiliation(s)
- Xi Zeng
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, College of Informatics, Huazhong Agricultural University, Wuhan, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Joseph Chi-Ching Tsui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Mai Shi
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jie Peng
- Acquired Immunodeficiency Syndrome (AIDS) Institute, The University of Hong Kong, Hong Kong, China
| | - Cyanne Ye Cao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lea Ling-Yu Kan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Carol Po-Ying Lau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yonghao Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lingyi Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Liu
- Acquired Immunodeficiency Syndrome (AIDS) Institute, The University of Hong Kong, Hong Kong, China
| | - Zhiwei Chen
- Acquired Immunodeficiency Syndrome (AIDS) Institute, The University of Hong Kong, Hong Kong, China
| | - Stephen Kwok-Wing Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
6
|
Yu W, Roy SK, Ma Y, LaVeist TA, Shankar S, Srivastava RK. Higher expression of SATB2 in hepatocellular carcinoma of African Americans determines more aggressive phenotypes than those of Caucasian Americans. J Cell Mol Med 2019; 23:7999-8009. [PMID: 31602781 PMCID: PMC6850930 DOI: 10.1111/jcmm.14652] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/16/2019] [Indexed: 12/20/2022] Open
Abstract
In the United States, Hepatocellular Carcinoma (HCC) incidence has tripled over the past two decades. The disease has disproportionately affected minority and disadvantaged populations. The purpose of this study was to examine the expression of SATB2 gene in HCC cells derived from African Americans (AA) and Caucasian Americans (CA) and assess its oncogenic potential by measuring cell viability, spheroid formation, epithelial‐mesenchymal transition (EMT), stem cell markers and pluripotency maintaining factors in cancer stem cells (CSCs). We compared the expression of SATB2 in human primary hepatocytes, HCC cells derived from AA and CA, and HCC CSCs. Hepatocellular carcinoma cells derived from AA expressed the higher level of SATB2 than those from CA. By comparison, normal human hepatocytes did not express SATB2. Higher expression of SATB2 in HCC cells from AA was associated with greater growth rate, cell viability, colony formation and EMT characteristics than those from CA. Knockout of SATB2 in CSCs by Crispr/Cas9 technique significantly inhibited the expression of SATB2 gene, stem cell markers (CD24, CD44 and CD133), pluripotency maintaining factors (c‐Myc, KLF4, SOX2 and OCT4), and EMT compared with non‐targeting control group. The expression of SATB2 was negatively correlated with miR34a. SATB2 rescued the miR‐34a‐mediated inhibition of CSC's viability. These data suggest that SATB2 is an oncogenic factor, and its higher expression may explain the disparity in HCC outcomes among AA.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Sanjit K Roy
- Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, USA
| | - Yiming Ma
- Kansas City VA Medical Center, Kansas City, MO, USA
| | - Thomas A LaVeist
- Department of Health Policy and Management, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, MO, USA.,Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, USA.,Department of Genetics, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, MO, USA.,Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health-New Orleans, New Orleans, LA, USA.,Department of Genetics, Louisiana State University Health Sciences Center-New Orleans, New Orleans, LA, USA
| |
Collapse
|
7
|
Dowrey T, Schwager EE, Duong J, Merkuri F, Zarate YA, Fish JL. Satb2 regulates proliferation and nuclear integrity of pre-osteoblasts. Bone 2019; 127:488-498. [PMID: 31325654 PMCID: PMC6708767 DOI: 10.1016/j.bone.2019.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/08/2019] [Accepted: 07/14/2019] [Indexed: 12/25/2022]
Abstract
Special AT-rich sequence binding protein 2 (Satb2) is a matrix attachment region (MAR) binding protein. Satb2 impacts skeletal development by regulating gene transcription required for osteogenic differentiation. Although its role as a high-order transcription factor is well supported, other roles for Satb2 in skeletal development remain unclear. In particular, the impact of dosage sensitivity (heterozygous mutations) and variance on phenotypic severity is still not well understood. To further investigate molecular and cellular mechanisms of Satb2-mediated skeletal defects, we used the CRISPR/Cas9 system to generate Satb2 mutations in MC3T3-E1 cells. Our data suggest that, in addition to its role in differentiation, Satb2 regulates progenitor proliferation. We also find that mutations in Satb2 cause chromatin defects including nuclear blebbing and donut-shaped nuclei. These defects may contribute to a slight increase in apoptosis in mutant cells, but apoptosis is insufficient to explain the proliferation defects. Satb2 expression exhibits population-level variation and is most highly expressed from late G1 to late G2. Based on these data, we hypothesize that Satb2 may regulate proliferation through two separate mechanisms. First, Satb2 may regulate the expression of genes necessary for cell cycle progression in pre-osteoblasts. Second, similar to other MAR-binding proteins, Satb2 may participate in DNA replication. We also hypothesize that variation in the severity or penetrance of Satb2-mediated proliferation defects is due to stochastic variation in Satb2 binding to DNA, which may be buffered in some genetic backgrounds. Further elucidation of the role of Satb2 in proliferation has potential impacts on our understanding of both skeletal defects and cancer.
Collapse
Affiliation(s)
- Todd Dowrey
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States of America
| | - Evelyn E Schwager
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States of America
| | - Julieann Duong
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States of America
| | - Fjodor Merkuri
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States of America
| | - Yuri A Zarate
- Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Jennifer L Fish
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States of America.
| |
Collapse
|
8
|
Chen QY, Costa M. Oncogenic and tumor suppressive roles of special AT-rich sequence-binding protein. J Carcinog 2018; 17:2. [PMID: 30123095 PMCID: PMC6071479 DOI: 10.4103/jcar.jcar_8_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/28/2017] [Indexed: 12/28/2022] Open
Abstract
In recent years, research efforts have been centered on the functional roles of special AT-rich sequence-binding protein (SATB2) in cancer development. Existing studies differ in the types of tumor tissues and cell lines used, resulting in mixed results, which hinder the clear understanding of whether SATB2 acts as a tumor suppressor or promoter. Literature search for this review consisted of a basic search on PubMed using keywords "SATB2" and "special AT-rich sequence-binding protein 2." Each article was then selected for further examination based on relevance of the title. In consideration to possible missing data from a primary PubMed search, after coding for relevant information, articles listed in the references section were filtered for further review. The current literature suggests that SATB2 can act both as a tumor suppressor and as a promoter since it can be regulated by multiple factors and is able to target different downstream genes in various types of cancer cell lines as well as tissues. Future studies should focus on its contradictory roles in different types of tumors. This paper provides a comprehensive review of currently available research on the role of SATB2 in different cancer cells and tissues and may provide some insight into the contradictory roles of SATB2 in cancer development.
Collapse
Affiliation(s)
- Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, NY, USA
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, NY, USA
| |
Collapse
|
9
|
Zuo C, Zhao X, Shi Y, Wu W, Zhang N, Xu J, Wang C, Hu G, Zhang X. TNF-α inhibits SATB2 expression and osteoblast differentiation through NF-κB and MAPK pathways. Oncotarget 2017; 9:4833-4850. [PMID: 29435145 PMCID: PMC5797016 DOI: 10.18632/oncotarget.23373] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/04/2017] [Indexed: 11/25/2022] Open
Abstract
Although the mechanisms of Tumor necrosis factor alpha (TNF-α) on facilitating osteoclast differentiation and bone resorption is well known, the mechanisms behind the suppression of the osteoblast differentiation from mesenchymal stem cells (MSCs) are still poorly understood. In this study, we observed a negative correlation between TNF-α levels and the expression of special AT-rich sequence-binding protein 2 (SATB2), a critical osteoblastogenesis transcription factor, in ovariectomy (OVX)-induced bone loss and IL-1-induced arthritis animal model. We found that TNF-α treatment inhibited mesenchymal cell line C2C12 osteoblast differentiation and sharply decreased BMP2-induced SATB2 expression. Upon TNF-α treatment, the activity of smad1/5/8 was inhibited, by contrast, extracellular signal-regulated kinase-1/2 (ERK1/2) and P38 was increased in C2C12 cells, the inhibitor of ERK1/2 (U0126) was found to abrogate the TNF-α inhibition of SATB2 expression. Furthermore, the NF-κB signaling pathway in C2C12 cells was significantly activated by the treatment of TNF-α, and TNF-α induced NF-κB directly binds to SATB2 promoter to suppress its expression. These results suggest that TNF-α suppresses SATB2 expression through activating NF-κB and MAPK signaling and depressing smad1/5/8 signaling, which contributes to the inhibition of osteoblast differentiation and might be potential therapeutic targets for inflammation-induced bone loss.
Collapse
Affiliation(s)
- Chijian Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.,The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Xiaoying Zhao
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yu Shi
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Wen Wu
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ning Zhang
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Chuandong Wang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Guoli Hu
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.,The Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200025, China
| |
Collapse
|
10
|
SATB2/β-catenin/TCF-LEF pathway induces cellular transformation by generating cancer stem cells in colorectal cancer. Sci Rep 2017. [PMID: 28887549 DOI: 10.1038/s41598‐017‐05458‐y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent studies have demonstrated the involvement of colorectal cancer (CRC) stem cells (CSC) in transformation, cancer progression and metastasis. The main goal of this paper was to examine the molecular mechanisms by which SATB2 induced malignant transformation of colorectal epithelial cells. SATB2 induced malignant transformation and these transformed cells gained the characteristics of CSCs by expressing stem cell markers (CD44, CD133, LGR5 and DCLK1) and transcription factors (c-Myc, Nanog and Sox2). Overexpression of SATB2 in normal colorectal epithelial cells increased cell motility, migration and invasion, which were associated with an increase in N-cadherin and Zeb1, and decrease in E-cadherin expression. SATB2 overexpression also upregulated XIAP and cyclin D1, suggesting its role in cell survival and cell cycle. Furthermore, the expression of SATB2 was positively correlated with β-catenin expression in CRC. In contrary, depletion of SATB2 inhibited cell proliferation, colony formation, cell motility and expression of β-catenin, Snail, Slug, Zeb1 and N-cadherin, and upregulated E-cadherin. Furthermore, SATB2 silencing inhibited the expression of stem cell markers, pluripotency maintaining transcription factors, cell cycle and cell proliferation/survival genes and TCF/LEF targets. Finally, β-catenin/TCF-LEF pathway mediated the biological effects of SATB2 in CSCs. These studies support the role of SATB2/β-catenin/TCF-LEF pathway in transformation and carcinogenesis.
Collapse
|
11
|
Yu W, Ma Y, Shankar S, Srivastava RK. SATB2/β-catenin/TCF-LEF pathway induces cellular transformation by generating cancer stem cells in colorectal cancer. Sci Rep 2017; 7:10939. [PMID: 28887549 PMCID: PMC5591219 DOI: 10.1038/s41598-017-05458-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
Recent studies have demonstrated the involvement of colorectal cancer (CRC) stem cells (CSC) in transformation, cancer progression and metastasis. The main goal of this paper was to examine the molecular mechanisms by which SATB2 induced malignant transformation of colorectal epithelial cells. SATB2 induced malignant transformation and these transformed cells gained the characteristics of CSCs by expressing stem cell markers (CD44, CD133, LGR5 and DCLK1) and transcription factors (c-Myc, Nanog and Sox2). Overexpression of SATB2 in normal colorectal epithelial cells increased cell motility, migration and invasion, which were associated with an increase in N-cadherin and Zeb1, and decrease in E-cadherin expression. SATB2 overexpression also upregulated XIAP and cyclin D1, suggesting its role in cell survival and cell cycle. Furthermore, the expression of SATB2 was positively correlated with β-catenin expression in CRC. In contrary, depletion of SATB2 inhibited cell proliferation, colony formation, cell motility and expression of β-catenin, Snail, Slug, Zeb1 and N-cadherin, and upregulated E-cadherin. Furthermore, SATB2 silencing inhibited the expression of stem cell markers, pluripotency maintaining transcription factors, cell cycle and cell proliferation/survival genes and TCF/LEF targets. Finally, β-catenin/TCF-LEF pathway mediated the biological effects of SATB2 in CSCs. These studies support the role of SATB2/β-catenin/TCF-LEF pathway in transformation and carcinogenesis.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Yiming Ma
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA.,Department of Pathology, University of Missouri-School of Medicine, Kansas City, MO, 64108, USA.,Stanley S. Scott Cancer Center, Department of Genetics, Louisiana State University Health Sciences Center, 1700 Tulane Avenue, New Orleans, LA 70112, United States
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO, 66128, USA. .,Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, MO, 64108, USA. .,Stanley S. Scott Cancer Center, Department of Genetics, Louisiana State University Health Sciences Center, 1700 Tulane Avenue, New Orleans, LA 70112, United States.
| |
Collapse
|
12
|
Chio CC, Wei L, Chen TG, Lin CM, Shieh JP, Yeh PS, Chen RM. Neuron-derived orphan receptor 1 transduces survival signals in neuronal cells in response to hypoxia-induced apoptotic insults. J Neurosurg 2015; 124:1654-64. [PMID: 26613168 DOI: 10.3171/2015.6.jns1535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECT Hypoxia can induce cell death or trigger adaptive mechanisms to guarantee cell survival. Neuron-derived orphan receptor 1 (NOR-1) works as an early-response protein in response to a variety of environmental stresses. In this study, the authors evaluated the roles of NOR-1 in hypoxia-induced neuronal insults. METHODS Neuro-2a cells were exposed to oxygen/glucose deprivation (OGD). Cell viability, cell morphology, cas-pase-3 activity, DNA fragmentation, and cell apoptosis were assayed to determine the mechanisms of OGD-induced neuronal insults. RNA and protein analyses were carried out to evaluate the effects of OGD on expressions of NOR-1, cAMP response element-binding (CREB), and cellular inhibitor of apoptosis protein 2 (cIAP2) genes. Translations of these gene expressions were knocked down using RNA interference. Mice subjected to traumatic brain injury (TBI) and NOR-1 was immunodetected. RESULTS Exposure of neuro-2a cells to OGD decreased cell viability in a time-dependent manner. Additionally, OGD led to cell shrinkage, DNA fragmentation, and cell apoptosis. In parallel, treatment of neuro-2a cells with OGD time dependently increased cellular NOR-1 mRNA and protein expressions. Interestingly, administration of TBI also augmented NOR-1 levels in the impacted regions of mice. As to the mechanism, exposure to OGD increased nuclear levels of the transcription factor CREB protein. Downregulating CREB expression using RNA interference simultaneously inhibited OGD-induced NOR-1 mRNA expression. Also, levels of cIAP2 mRNA and protein in neuro-2a cells were augmented by OGD. After reducing cIAP2 translation, OGD-induced cell death was reduced. Sequentially, application of NOR-1 small interfering RNA to neuro-2a cells significantly inhibited OGD-induced cIAP2 mRNA expression and concurrently alleviated hypoxia-induced alterations in cell viability, caspase-3 activation, DNA damage, and cell apoptosis. CONCLUSIONS This study shows that NOR-1 can transduce survival signals in neuronal cells responsible for hypoxiainduced apoptotic insults through activation of a CREB/cIAP2-dependent mechanism.
Collapse
Affiliation(s)
| | - Li Wei
- Departments of 3 Neurosugery and
| | | | | | | | | | - Ruei-Ming Chen
- Comprehensive Cancer Center.,Graduate Institute of Medical Sciences, Taipei Medical University; ,Brain Disease Research Center, Wan-Fang Hospital, Taipei Medical University; and.,Anesthetics and Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
13
|
Lee YE, Hong CY, Lin YL, Chen RM. MicroRNA-1 participates in nitric oxide-induced apoptotic insults to MC3T3-E1 cells by targeting heat-shock protein-70. Int J Biol Sci 2015; 11:246-55. [PMID: 25678843 PMCID: PMC4323364 DOI: 10.7150/ijbs.11138] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 01/06/2015] [Indexed: 12/25/2022] Open
Abstract
Our previous studies showed that nitric oxide (NO) could induce osteoblast apoptosis. MicroRNA-1 (miR-1), a skeletal- and cardiac muscle-specific small non-coding RNA, contributes to the regulation of multiple cell activities. In this study, we evaluated the roles of miR-1 in NO-induced insults to osteoblasts and the possible mechanisms. Exposure of mouse MC3T3-E1 cells to sodium nitroprusside (SNP) increased amounts of cellular NO and intracellular reactive oxygen species. Sequentially, SNP decreased cell survival but induced caspase-3 activation, DNA fragmentation, and cell apoptosis. In parallel, treatment with SNP induced miR-1 expression in a time-dependent manner. Application of miR-1 antisense inhibitors to osteoblasts caused significant inhibition of SNP-induced miR-1 expression. Knocking down miR-1 concurrently attenuated SNP-induced alterations in cell morphology and survival. Consecutively, SNP time-dependently inhibited heat-shock protein (HSP)-70 messenger (m)RNA and protein expressions. A bioinformatic search predicted the existence of miR-1-specific binding elements in the 3'-untranslational region of HSP-70 mRNA. Downregulation of miR-1 expression simultaneously lessened SNP-induced inhibition of HSP-70 mRNA and protein expressions. Consequently, SNP-induced modifications in the mitochondrial membrane potential, caspase-3 activation, DNA fragmentation, and apoptotic insults were significantly alleviated by miR-1 antisense inhibitors. Therefore, this study showed that miR-1 participates in NO-induced apoptotic insults through targeting HSP-70 gene expression.
Collapse
Affiliation(s)
- Yong-Eng Lee
- 1. Department of Orthopedic Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan ; 2. Cell Biology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Chung-Ye Hong
- 3. Department of Internal Medicine, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Yi-Ling Lin
- 2. Cell Biology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| | - Ruei-Ming Chen
- 2. Cell Biology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan ; 4. Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. ; 5. Anesthetics and Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Fang HW, Kao WY, Lin PI, Chang GW, Hung YJ, Chen RM. Effects of Polypropylene Carbonate/Poly(d,l-lactic) Acid/Tricalcium Phosphate Elastic Composites on Improving Osteoblast Maturation. Ann Biomed Eng 2014; 43:1999-2009. [DOI: 10.1007/s10439-014-1236-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 12/22/2014] [Indexed: 12/11/2022]
|
15
|
Roles of microRNA-1 in hypoxia-induced apoptotic insults to neuronal cells. Arch Toxicol 2014; 90:191-202. [DOI: 10.1007/s00204-014-1364-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
|
16
|
Zhao X, Qu Z, Tickner J, Xu J, Dai K, Zhang X. The role of SATB2 in skeletogenesis and human disease. Cytokine Growth Factor Rev 2013; 25:35-44. [PMID: 24411565 DOI: 10.1016/j.cytogfr.2013.12.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/15/2013] [Indexed: 02/06/2023]
Abstract
Since the discovery of SATB2 (special AT-rich sequence binding protein 2) a decade ago, its pivotal roles in development and tissue regeneration have emerged, particularly in craniofacial patterning and development, palate formation, and osteoblast differentiation and maturation. As a member of the special AT-rich binding proteins family that bind to nuclear matrix-attachment regions (MAR), it also displays functional versatility in central nervous development, especially corpus callosum and pons formation, cancer development and prognosis, as well as in immune regulation. At the molecular level, Satb2 gene expression appears to be tissue and stage-specific, and is regulated by several cytokines and growth factors, such as BMP2/4/7, insulin, CNTF, and LIF via ligand receptor signaling pathways. SATB2 mainly performs a twofold role as a transcription regulator by directly binding to AT-rich sequences in MARs to modulate chromatin remodeling, or through association with other transcription factors to modulate the cis-regulation elements and thus to regulate the expression of down-stream target genes and a wide range of biological processes. This contemporary review provides an exploration of the molecular characteristics and function of SATB2; including its expression and cytokine regulation, its involvement in human disease, and its potential roles in skeletogenesis.
Collapse
Affiliation(s)
- Xiaoying Zhao
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China
| | - Zhihu Qu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 200031, China
| | - Jennifer Tickner
- School of Pathology and Laboratory Medicine, The University of Western Australia (M504), 35 Stirling Highway, Crawley WA 6009, Australia
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, The University of Western Australia (M504), 35 Stirling Highway, Crawley WA 6009, Australia.
| | - Kerong Dai
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China; Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaoling Zhang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200025, China; Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
17
|
Wang H, Dong D, Tang S, Chen X, Gao Q. PPE38 of Mycobacterium marinum triggers the cross-talk of multiple pathways involved in the host response, as revealed by subcellular quantitative proteomics. J Proteome Res 2013; 12:2055-66. [PMID: 23514422 PMCID: PMC3646403 DOI: 10.1021/pr301017e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
The
PE/PPE family of proteins which are in high abundance in pathogenic
species such as Mycobacterium tuberculosis and M. marinum, play the critical
role in generating antigenic variation and evasion of host immune
responses. However, little is known about their functional roles in
mycobacterial pathogenesis. Previously, we found that PPE38 is associated
with the virulence of mycobacteria, presumably by modulating the host
immune response. To clarify the link between PPE38 and host response,
we employed a subcellular, amino acid-coded mass tagging (AACT)/SILAC-based
quantitative proteomic approach to determine the proteome changes
during host response to M. marinum PPE38.
As a result, 291 or 290 proteins were found respectively to be up-
or down-regulated in the nucleus. Meanwhile, 576 upregulated and 272
downregulated proteins were respectively detected in the cytosol.
The data of quantitative proteomic changes and concurrent biological
validations revealed that M. marinum PPE38 could trigger extensive inflammatory responses in macrophages,
probably through interacting with toll-like receptor 2 (TLR2). We
also found that PPE38 may arrest MHC-1 processing and presentation
in infected macrophages. Using bioinformatics tools to analyze global
changes in the host proteome, we obtained a PPE38-respondor network involved in various transcriptional factors (TFs) and TF-associated
proteins. The results of our systems investigation now indicate that there is cross-talk involving a broad range of diverse biological pathways/processes that coordinate the host response to M. marinum PPE38.
Collapse
Affiliation(s)
- Hui Wang
- Key Laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|