1
|
Yoshikawa K. Necdin: A purposive integrator of molecular interaction networks for mammalian neuron vitality. Genes Cells 2021; 26:641-683. [PMID: 34338396 PMCID: PMC9290590 DOI: 10.1111/gtc.12884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/29/2021] [Indexed: 12/29/2022]
Abstract
Necdin was originally found in 1991 as a hypothetical protein encoded by a neural differentiation‐specific gene transcript in murine embryonal carcinoma cells. Virtually all postmitotic neurons and their precursor cells express the necdin gene (Ndn) during neuronal development. Necdin mRNA is expressed only from the paternal allele through genomic imprinting, a placental mammal‐specific epigenetic mechanism. Necdin and its homologous MAGE (melanoma antigen) family, which have evolved presumedly from a subcomplex component of the SMC5/6 complex, are expressed exclusively in placental mammals. Paternal Ndn‐mutated mice totally lack necdin expression and exhibit various types of neuronal abnormalities throughout the nervous system. Ndn‐null neurons are vulnerable to detrimental stresses such as DNA damage. Necdin also suppresses both proliferation and apoptosis of neural stem/progenitor cells. Functional analyses using Ndn‐manipulated cells reveal that necdin consistently exerts antimitotic, anti‐apoptotic and prosurvival effects. Necdin interacts directly with a number of regulatory proteins including E2F1, p53, neurotrophin receptors, Sirt1 and PGC‐1α, which serve as major hubs of protein–protein interaction networks for mitosis, apoptosis, differentiation, neuroprotection and energy homeostasis. This review focuses on necdin as a pleiotropic protein that integrates molecular interaction networks to promote neuronal vitality in modern placental mammals.
Collapse
|
2
|
PGC1α: Friend or Foe in Cancer? Genes (Basel) 2018; 9:genes9010048. [PMID: 29361779 PMCID: PMC5793199 DOI: 10.3390/genes9010048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
The PGC1 family (Peroxisome proliferator-activated receptor γ (PPARγ) coactivators) of transcriptional coactivators are considered master regulators of mitochondrial biogenesis and function. The PGC1α isoform is expressed especially in metabolically active tissues, such as the liver, kidneys and brain, and responds to energy-demanding situations. Given the altered and highly adaptable metabolism of tumor cells, it is of interest to investigate PGC1α in cancer. Both high and low levels of PGC1α expression have been reported to be associated with cancer and worse prognosis, and PGC1α has been attributed with oncogenic as well as tumor suppressive features. Early in carcinogenesis PGC1α may be downregulated due to a protective anticancer role, and low levels likely reflect a glycolytic phenotype. We suggest mechanisms of PGC1α downregulation and how these might be connected to the increased cancer risk that obesity is now known to entail. Later in tumor progression PGC1α is often upregulated and is reported to contribute to increased lipid and fatty acid metabolism and/or a tumor cell phenotype with an overall metabolic plasticity that likely supports drug resistance as well as metastasis. We conclude that in cancer PGC1α is neither friend nor foe, but rather the obedient servant reacting to metabolic and environmental cues to benefit the tumor cell.
Collapse
|
3
|
Bharadwaj U, Eckols TK, Xu X, Kasembeli MM, Chen Y, Adachi M, Song Y, Mo Q, Lai SY, Tweardy DJ. Small-molecule inhibition of STAT3 in radioresistant head and neck squamous cell carcinoma. Oncotarget 2018; 7:26307-30. [PMID: 27027445 PMCID: PMC5041982 DOI: 10.18632/oncotarget.8368] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/14/2016] [Indexed: 12/17/2022] Open
Abstract
While STAT3 has been validated as a target for treatment of many cancers, including head and neck squamous cell carcinoma (HNSCC), a STAT3 inhibitor is yet to enter the clinic. We used the scaffold of C188, a small-molecule STAT3 inhibitor previously identified by us, in a hit-to-lead program to identify C188-9. C188-9 binds to STAT3 with high affinity and represents a substantial improvement over C188 in its ability to inhibit STAT3 binding to its pY-peptide ligand, to inhibit cytokine-stimulated pSTAT3, to reduce constitutive pSTAT3 activity in multiple HNSCC cell lines, and to inhibit anchorage dependent and independent growth of these cells. In addition, treatment of nude mice bearing xenografts of UM-SCC-17B, a radioresistant HNSCC line, with C188-9, but not C188, prevented tumor xenograft growth. C188-9 treatment modulated many STAT3-regulated genes involved in oncogenesis and radioresistance, as well as radioresistance genes regulated by STAT1, due to its potent activity against STAT1, in addition to STAT3. C188-9 was well tolerated in mice, showed good oral bioavailability, and was concentrated in tumors. Thus, C188-9, either alone or in combination with radiotherapy, has potential for use in treating HNSCC tumors that demonstrate increased STAT3 and/or STAT1 activation.
Collapse
Affiliation(s)
- Uddalak Bharadwaj
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - T Kris Eckols
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xuejun Xu
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Moses M Kasembeli
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Makoto Adachi
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yongcheng Song
- Department of Pharmacology, Baylor College of Medicine, Houston, Texas, USA
| | - Qianxing Mo
- Department of Medicine, Division of Biostatistics, Dan L. Duncan Cancer Center, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David J Tweardy
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Wang Q, Wu W, Xu Z, Luo F, Zhou Q, Li P, Xie J. Copy number changes and methylation patterns in an isodicentric and a ring chromosome of 15q11-q13: report of two cases and review of literature. Mol Cytogenet 2015; 8:97. [PMID: 26697114 PMCID: PMC4687147 DOI: 10.1186/s13039-015-0198-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/10/2015] [Indexed: 01/27/2023] Open
Abstract
Background The low copy repeats (LCRs) in chromosome 15q11-q13 have been recognized as breakpoints (BP) for not only intrachromosomal deletions and duplications but also small supernumerary marker chromosomes 15, sSMC(15)s, in the forms of isodicentric chromosome or small ring chromosome. Further characterization of copy number changes and methylation patterns in these sSMC(15)s could lead to better understanding of their phenotypic consequences. Methods Routine G-band karyotyping, fluorescence in situ hybridization (FISH), array comparative genomic hybridization (aCGH) analysis and methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) assay were performed on two Chinese patients with a sSMC(15). Results Patient 1 showed an isodicentric 15, idic(15)(q13), containing symmetrically two copies of a 7.7 Mb segment of the 15q11-q13 region by a BP3::BP3 fusion. Patient 2 showed a ring chromosome 15, r(15)(q13), with alternative one-copy and two-copy segments spanning a 12.3 Mb region. The defined methylation pattern indicated that the idic(15)(q13) and the r(15)(q13) were maternally derived. Conclusions Results from these two cases and other reported cases from literature indicated that combined karyotyping, aCGH and MS-MLPA analyses are effective to define the copy number changes and methylation patterns for sSMC(15)s in a clinical setting. The characterized genomic structure and epigenetic pattern of sSMC(15)s could lead to further gene expression profiling for better phenotype correlation.
Collapse
Affiliation(s)
- Qin Wang
- Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen, Guangdong China
| | - Weiqing Wu
- Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen, Guangdong China ; Department of Genetics, Yale School of Medicine, New Haven, CT USA
| | - Zhiyong Xu
- Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen, Guangdong China
| | - Fuwei Luo
- Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen, Guangdong China
| | - Qinghua Zhou
- Department of Genetics, Yale School of Medicine, New Haven, CT USA ; First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong China
| | - Peining Li
- Department of Genetics, Yale School of Medicine, New Haven, CT USA
| | - Jiansheng Xie
- Shenzhen Maternity and Child Healthcare Hospital, 3012 Fuqiang Road, Shenzhen, Guangdong China
| |
Collapse
|
5
|
Virani S, Light E, Peterson LA, Sartor MA, Taylor JMG, McHugh JB, Wolf GT, Rozek LS. Stability of methylation markers in head and neck squamous cell carcinoma. Head Neck 2015; 38 Suppl 1:E1325-31. [PMID: 26566081 DOI: 10.1002/hed.24223] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND As cancer progresses, methylation patterns change to promote the tumorigenic phenotype. However, stability of methylation markers over time and the extent that biopsy samples are representative of larger tumor specimens are unknown. This information is critical for clinical use of such biomarkers. METHODS Ninety-eight patients with tumor specimens from 2 timepoints were measured for DNA methylation in the promoter regions across 4 genes. RESULTS There were no significant differences in overall methylation of CCNA1 (cyclin A1), NDN (necdin), deleted in colorectal carcinoma (DCC), and cluster of differentiation 1a (CD1A) within paired specimens (p values = .56, .17, .66, and .58, respectively). All genes showed strong correlations between paired specimens across time. Methylation was most consistent for CCNA1 and NDN over time. CONCLUSION This report provides the first evidence that methylation markers measured in biopsy samples are representative of gene methylation in later specimens and suggests that biopsy markers could be representative biomarkers for use in defining personalized treatment utilizing epigenetic changes. © 2015 Wiley Periodicals, Head Neck 38: E1325-E1331, 2016.
Collapse
Affiliation(s)
- Shama Virani
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Emily Light
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, Michigan
| | - Lisa A Peterson
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jeremy M G Taylor
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, Michigan
| | - Jonathan B McHugh
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Laura S Rozek
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.,Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
6
|
Virani S, Bellile E, Bradford CR, Carey TE, Chepeha DB, Colacino JA, Helman JI, McHugh JB, Peterson LA, Sartor MA, Taylor JM, Walline HM, Wolf GT, Rozek LS. NDN and CD1A are novel prognostic methylation markers in patients with head and neck squamous carcinomas. BMC Cancer 2015; 15:825. [PMID: 26518708 PMCID: PMC4628358 DOI: 10.1186/s12885-015-1806-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/16/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND HPV-associated HNSCCs have a distinct etiologic mechanism and better prognosis than those with non-HPV associated HNSCCs. However, even within the each group, there is heterogeneity in survival time. Here, we test the hypothesis that specific candidate gene methylation markers (CCNA1, NDN, CD1A, DCC, p16, GADD45A) are associated with tumor recurrence and survival, in a well-characterized, prospective, cohort of 346 HNSCC patients. METHODS Kaplan-Meier curves were used to estimate survival time distributions. Multivariable Cox Proportional Hazards models were used to test associations between each methylation marker and OST/RPFT after adjusting for known or identified prognostic factors. Stratified Cox models included an interaction term between HPV and methylation marker to test for differences in the associations of the biomarker with OST or RPFT across HPV status. RESULTS Methylation markers were differentially associated with patient characteristics. DNA hypermethylation of NDN and CD1A was found to be significantly associated with overall survival time (OST) in all HNSCC patients (NDN hazard ratio (HR): 2.35, 95% CI: 1.40-3.94; CD1A HR: 1.31, 95% CI: 1.01-1.71). Stratification by HPV status revealed hypermethylation of CD1A was associated with better OST and recurrence/persistence-free time (RPFT) (OST HR: 3.34, 95% CI: 1.88-5.93; RPFT HR: 2.06, 95% CI: 1.21-3.49), while hypomethylation of CCNA1 was associated with increased RPFT in HPV (+) patients only (HR: 0.31, 95% CI: 0.13-0.74). CONCLUSIONS This study is the first to describe novel epigenetic alterations associated with survival in an unselected, prospectively collected, consecutive cohort of patients with HNSCC. DNA hypermethylation of NDN and CD1A was found to be significantly associated with increased overall survival time in all HNSCC patients. However, stratification by the important prognostic factor of HPV status revealed the immune marker, CD1A, and the cell cycle regulator, CCNA1 to be associated with prognosis in HPV (+) patients, specifically. Here, we identified novel methylation markers and specific, epigenetic molecular differences associated with HPV status, which warrant further investigation.
Collapse
Affiliation(s)
- Shama Virani
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Emily Bellile
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA.
| | - Carol R Bradford
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Thomas E Carey
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Douglas B Chepeha
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Joseph I Helman
- Department of Oral-Maxillofacial Surgery, University of Michigan Dental School, Ann Arbor, MI, USA.
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Jonathan B McHugh
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Lisa A Peterson
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| | - Jeremy Mg Taylor
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA.
| | - Heather M Walline
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Greg T Wolf
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Laura S Rozek
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
- Department of Otolaryngology, University of Michigan Medical School, Ann Arbor, MI, USA.
- 1415 Washington Heights, Environmental Health Sciences 6630 SPH, Ann Arbor, MI, 48109-2029, USA.
| |
Collapse
|
7
|
Lu Z, Jiao D, Qiao J, Yang S, Yan M, Cui S, Liu Z. Restin suppressed epithelial-mesenchymal transition and tumor metastasis in breast cancer cells through upregulating mir-200a/b expression via association with p73. Mol Cancer 2015; 14:102. [PMID: 25972084 PMCID: PMC4429374 DOI: 10.1186/s12943-015-0370-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 04/21/2015] [Indexed: 12/31/2022] Open
Abstract
Background Restin belongs to MAGE superfamily and is known as MAGE H1. Restin was firstly cloned from HL-60 cells treated with all-trans retinoic acid (ATRA). Previous studies showed a pro-apoptotic role of Restin in several cell lines. However, little information is available on its expression patterns and functions in vivo. Our study was performed to detect if Restin plays a role in breast cancer cells in vitro and in vivo. Methods and results Real-time PCR and western blot were conducted to detect Restin expression in multiple breast cancer cell lines and Restin level was negatively related with cell motility. Restin overexpression and knockdown stable cell lines were established by transducing lentivirus into MCF-7 and MDA-MB-231 cells. Cell morphology, wound closure assay, transwell migration and invasion assays were performed to detect if Restin inhibited EMT. Our data showed that Restin overexpressed cells exhibited classical epithelial cell morphology, and Restin overexpression resulted in activation of epithelial markers and suppression of mesenchymal markers, and inhibition of cell migration and invasion. Tumor xenograft model was used to characterize the biological functions of Restin in vivo. We found that Restin overexpression led to reduced lung metastasis. Real-time PCR, western blot, luciferase assay and ChIP assay were performed to identify the potential targets of Restin and the underlying molecular mechanisms. Among several master regulators of EMT, only ZEB1/2 levels were dramatically inhibited by Restin. Unexpectedly, Restin indirectly regulated ZEB1/2 expression at post-transcriptional level. We further identified mir-200a/b, well-characterized mediators controlling ZEB1/2 expression, were transcriptionally activated by Restin and the regulation was dependent on the p53 binding site in mir-200b/a/429 promoter. Further mechanical studies demonstrated Restin interacted with p73, one of p53 family members, which contributed to Restin-mediated activation of mir-200a/b and suppression of ZEB1/2. Conclusions Taken together, our results suggest that Restin inhibits EMT and tumor metastasis by controlling the expression of the tumor metastasis suppressor mir-200a/b via association with p73. Our findings not only establish a mechanistic link between Restin, EMT and tumor metastasis, but also provide strong evidence supporting the notion that MAGE Group II proteins may exert a tumor suppressive effect in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0370-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenduo Lu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Dechuang Jiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Jianghua Qiao
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Sen Yang
- Department of Pathogen Biology, Basic Medical College of Zhengzhou University, #100 Science Road, Zhengzhou, 450001, People's Republic of China.
| | - Min Yan
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Shude Cui
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| | - Zhenzhen Liu
- Department of Breast surgery, Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, #127 Dongming Road, Zhengzhou, Henan, 450008, People's Republic of China.
| |
Collapse
|
8
|
Bharadwaj U, Eckols TK, Kolosov M, Kasembeli MM, Adam A, Torres D, Zhang X, Dobrolecki LE, Wei W, Lewis MT, Dave B, Chang JC, Landis MD, Creighton CJ, Mancini MA, Tweardy DJ. Drug-repositioning screening identified piperlongumine as a direct STAT3 inhibitor with potent activity against breast cancer. Oncogene 2015; 34:1341-53. [PMID: 24681959 PMCID: PMC4182178 DOI: 10.1038/onc.2014.72] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 12/12/2022]
Abstract
Signal transducer and activator of transcription (STAT) 3 regulates many cardinal features of cancer including cancer cell growth, apoptosis resistance, DNA damage response, metastasis, immune escape, tumor angiogenesis, the Warburg effect and oncogene addiction and has been validated as a drug target for cancer therapy. Several strategies have been used to identify agents that target Stat3 in breast cancer but none has yet entered into clinical use. We used a high-throughput fluorescence microscopy search strategy to identify compounds in a drug-repositioning library (Prestwick library) that block ligand-induced nuclear translocation of Stat3 and identified piperlongumine (PL), a natural product isolated from the fruit of the pepper Piper longum. PL inhibited Stat3 nuclear translocation, inhibited ligand-induced and constitutive Stat3 phosphorylation, and modulated expression of multiple Stat3-regulated genes. Surface plasmon resonance assay revealed that PL directly inhibited binding of Stat3 to its phosphotyrosyl peptide ligand. Phosphoprotein antibody array analysis revealed that PL does not modulate kinases known to activate Stat3 such as Janus kinases, Src kinase family members or receptor tyrosine kinases. PL inhibited anchorage-independent and anchorage-dependent growth of multiple breast cancer cell lines having increased pStat3 or total Stat3, and induced apoptosis. PL also inhibited mammosphere formation by tumor cells from patient-derived xenografts. PL's antitumorigenic function was causally linked to its Stat3-inhibitory effect. PL was non-toxic in mice up to a dose of 30 mg/kg/day for 14 days and caused regression of breast cancer cell line xenografts in nude mice. Thus, PL represents a promising new agent for rapid entry into the clinic for use in treating breast cancer, as well as other cancers in which Stat3 has a role.
Collapse
Affiliation(s)
- U Bharadwaj
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - T K Eckols
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - M Kolosov
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - M M Kasembeli
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - A Adam
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - D Torres
- Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - X Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - L E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - W Wei
- 1] Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - M T Lewis
- 1] Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA [3] Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA [4] Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - B Dave
- The Methodist Cancer Center, The Methodist Hospital Research Institute, Houston, TX, USA
| | - J C Chang
- The Methodist Cancer Center, The Methodist Hospital Research Institute, Houston, TX, USA
| | - M D Landis
- The Methodist Cancer Center, The Methodist Hospital Research Institute, Houston, TX, USA
| | - C J Creighton
- Section of Hematology-Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - M A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - D J Tweardy
- 1] Section of Infectious Disease, Department of Medicine, Baylor College of Medicine, Houston, TX, USA [2] Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA [3] Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
Li X, Hughes SC, Wevrick R. Evaluation of melanoma antigen (MAGE) gene expression in human cancers using The Cancer Genome Atlas. Cancer Genet 2015; 208:25-34. [DOI: 10.1016/j.cancergen.2014.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/11/2014] [Accepted: 11/18/2014] [Indexed: 12/20/2022]
|
10
|
Ju H, Lee S, Kang S, Kim SS, Ghil S. The alpha subunit of Go modulates cell proliferation and differentiation through interactions with Necdin. Cell Commun Signal 2014; 12:39. [PMID: 25012566 PMCID: PMC4227020 DOI: 10.1186/s12964-014-0039-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/12/2014] [Indexed: 11/23/2022] Open
Abstract
Background Heterotrimeric GTP-binding proteins (G-proteins) play an important role in mediating signal transduction generated by neurotransmitters or hormones. Go, a member of the Gi/Go subfamily, is the most abundant G-protein found in the brain. Recently, the alpha subunit of Go (Gαo) was characterized as an inducer of neuronal differentiation. However, its underlying molecular mechanisms have remained unclear to date, since the downstream effectors of Gαo are ambiguous. Results A neurally differentiated embryonal carcinoma-derived protein (Necdin) was isolated as an interacting partner for Gαo from a mouse brain cDNA library using yeast two-hybrid screening. Interactions between the proteins were confirmed with several affinity binding assays, both in vitro and in vivo. Necdin interacted directly and preferentially with activated Gαo, compared to wild-type protein. Interestingly, Gαo did not interact with Gαi, despite high sequence homology between the two proteins. We subsequently analyzed whether Gαo modulates the cellular activities of Necdin. Notably, expression of Gαo significantly augmented Necdin-mediated cellular responses, such as proliferation and differentiation. Moreover, activation of type 1 cannabinoid receptor (CB1R), a Gi/oα-coupled receptor, augmented cell growth suppression, which was mediated by Gαo and Necdin in U87MG cells containing CB1R, Gαo, and Necdin as normal components. Conclusions These results collectively suggest that Necdin is a candidate downstream effector for Gαo. Our findings provide novel insights into the cellular roles of Gαo and its coupled receptor.
Collapse
|
11
|
STAT3 Target Genes Relevant to Human Cancers. Cancers (Basel) 2014; 6:897-925. [PMID: 24743777 PMCID: PMC4074809 DOI: 10.3390/cancers6020897] [Citation(s) in RCA: 383] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/22/2014] [Accepted: 03/28/2014] [Indexed: 12/29/2022] Open
Abstract
Since its discovery, the STAT3 transcription factor has been extensively studied for its function as a transcriptional regulator and its role as a mediator of development, normal physiology, and pathology of many diseases, including cancers. These efforts have uncovered an array of genes that can be positively and negatively regulated by STAT3, alone and in cooperation with other transcription factors. Through regulating gene expression, STAT3 has been demonstrated to play a pivotal role in many cellular processes including oncogenesis, tumor growth and progression, and stemness. Interestingly, recent studies suggest that STAT3 may behave as a tumor suppressor by activating expression of genes known to inhibit tumorigenesis. Additional evidence suggested that STAT3 may elicit opposing effects depending on cellular context and tumor types. These mixed results signify the need for a deeper understanding of STAT3, including its upstream regulators, parallel transcription co-regulators, and downstream target genes. To help facilitate fulfilling this unmet need, this review will be primarily focused on STAT3 downstream target genes that have been validated to associate with tumorigenesis and/or malignant biology of human cancers.
Collapse
|
12
|
Sheng WJ, Jiang H, Wu DL, Zheng JH. Early responses of the STAT3 pathway to platinum drugs are associated with cisplatin resistance in epithelial ovarian cancer. Braz J Med Biol Res 2013; 46:650-8. [PMID: 23969971 PMCID: PMC3854422 DOI: 10.1590/1414-431x20133003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 05/23/2013] [Indexed: 01/05/2023] Open
Abstract
Cisplatin resistance remains one of the major obstacles when treating epithelial
ovarian cancer. Because oxaliplatin and nedaplatin are effective against
cisplatin-resistant ovarian cancer in clinical trials and signal transducer and
activator of transcription 3 (STAT3) is associated with cisplatin resistance, we
investigated whether overcoming cisplatin resistance by oxaliplatin and nedaplatin
was associated with the STAT3 pathway in ovarian cancer. Alamar blue, clonogenic, and
wound healing assays, and Western blot analysis were used to compare the effects of
platinum drugs in SKOV-3 cells. At an equitoxic dose, oxaliplatin and nedaplatin
exhibited similar inhibitory effects on colony-forming ability and greater inhibition
on cell motility than cisplatin in ovarian cancer. Early in the time course of drug
administration, cisplatin increased the expression of pSTAT3 (Tyr705), STAT3α, VEGF,
survivin, and Bcl-XL, while oxaliplatin and nedaplatin exhibited the
opposite effects, and upregulated pSTAT3 (Ser727) and STAT3β. The STAT3 pathway
responded early to platinum drugs associated with cisplatin resistance in epithelial
ovarian cancer and provided a rationale for new therapeutic strategies to reverse
cisplatin resistance.
Collapse
Affiliation(s)
- W J Sheng
- The First Affiliated Hospital of Harbin Medical University, Department of Obstetrics and Gynecology, Harbin, China
| | | | | | | |
Collapse
|
13
|
Selective STAT3-α or -β expression reveals spliceform-specific phosphorylation kinetics, nuclear retention and distinct gene expression outcomes. Biochem J 2012; 447:125-36. [PMID: 22799634 PMCID: PMC3441131 DOI: 10.1042/bj20120941] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphorylation of STAT3 (signal transducer and activator of transcription 3) is critical for its nuclear import and transcriptional activity. Although a shorter STAT3β spliceform was initially described as a negative regulator of STAT3α, gene knockout studies have revealed that both forms play critical roles. We have expressed STAT3α and STAT3β at comparable levels to facilitate a direct comparison of their functional effects, and have shown their different cytokine-stimulated kinetics of phosphorylation and nuclear translocation. Notably, the sustained nuclear translocation and phosphorylation of STAT3β following cytokine exposure contrasted with a transient nuclear translocation and phosphorylation of STAT3α. Importantly, co-expression of the spliceforms revealed that STAT3β enhanced and prolonged the phosphorylation and nuclear retention of STAT3α, but a STAT3β R609L mutant, with a disrupted SH2 (Src homology 2) domain, was not tyrosine phosphorylated following cytokine stimulation and could not cross-regulate STAT3α. The physiological importance of prolonged phosphorylation and nuclear retention was indicated by transcriptome profiling of STAT3−/− cells expressing either STAT3α or STAT3β, revealing the complexity of genes that are up- and down-regulated by the STAT3 spliceforms, including a distinct set of STAT3β-specific genes regulated under basal conditions and after cytokine stimulation. These results highlight STAT3β as a significant transcriptional regulator in its own right, with additional actions to cross-regulate STAT3α phosphorylation and nuclear retention after cytokine stimulation.
Collapse
|
14
|
Lavi-Itzkovitz A, Tcherpakov M, Levy Z, Itzkovitz S, Muscatelli F, Fainzilber M. Functional consequences of necdin nucleocytoplasmic localization. PLoS One 2012; 7:e33786. [PMID: 22442722 PMCID: PMC3307762 DOI: 10.1371/journal.pone.0033786] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 02/19/2012] [Indexed: 02/02/2023] Open
Abstract
Background Necdin, a MAGE family protein expressed primarily in the nervous system, has been shown to interact with both nuclear and cytoplasmic proteins, but the mechanism of its nucleocytoplasmic transport are unknown. Methodology/Principal Findings We carried out a large-scale interaction screen using necdin as a bait in the yeast RRS system, and found a wide range of potential interactors with different subcellular localizations, including over 60 new candidates for direct binding to necdin. Integration of these interactions into a comprehensive network revealed a number of coherent interaction modules, including a cytoplasmic module connecting to necdin through huntingtin-associated protein 1 (Hap1), dynactin and hip-1 protein interactor (Hippi); a nuclear P53 and Creb-binding-protein (Crebbp) module, connecting through Crebbp and WW domain-containing transcription regulator protein 1 (Wwtr1); and a nucleocytoplasmic transport module, connecting through transportins 1 and 2. We validated the necdin-transportin1 interaction and characterized a sequence motif in necdin that modulates karyopherin interaction. Surprisingly, a D234P necdin mutant showed enhanced binding to both transportin1 and importin β1. Finally, exclusion of necdin from the nucleus triggered extensive cell death. Conclusions/Significance These data suggest that necdin has multiple roles within protein complexes in different subcellular compartments, and indicate that it can utilize multiple karyopherin-dependent pathways to modulate its localization.
Collapse
Affiliation(s)
- Anat Lavi-Itzkovitz
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Marianna Tcherpakov
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Zehava Levy
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Shalev Itzkovitz
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Francoise Muscatelli
- Institut de Neurobiologie de la Méditerranée, INSERM U901, Parc Scientifique de Luminy BP 13, Marseille, France
| | - Mike Fainzilber
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|