1
|
Liu W, Zhang Y, Nie Y, Liu Y, Li Z, Zhang Z, Gong B, Ma M. AGBL2 promotes renal cell carcinoma cells proliferation and migration via α-tubulin detyrosination. Heliyon 2024; 10:e37086. [PMID: 39315218 PMCID: PMC11417249 DOI: 10.1016/j.heliyon.2024.e37086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background AGBL2's role in tumorigenesis and cancer progression has been reported in several cancer studies, and it is closely associated with α-tubulin detyrosination. The roles of AGBL2 and α-tubulin detyrosination in renal cell carcinoma (RCC) pathogenesis remain unclear and require further investigation. Methods In this study, we conducted an analysis of AGBL2 expression differences between renal clear cell carcinoma tissues and normal tissues using data from The Cancer Genome Atlas (TCGA). We performed a comprehensive prognostic analysis of AGBL2 in Kidney Renal Clear Cell Carcinoma (KIRC) using univariate and multivariate Cox regression. Based on the results of the Cox analysis, we constructed a prognostic model to assess its predictive capabilities. Receiver Operating Characteristic (ROC) analysis confirmed the diagnostic value of AGBL2 in renal cancer. We conducted further validation by analyzing cancer tissue samples and renal cancer cell lines, which confirmed the role of AGBL2 in promoting RCC cell proliferation and migration through in vitro experiments. Additionally, we verified the impact of AGBL2's detyrosination on α-tubulin using the tubulin carboxypeptidase (TCP) inhibitor parthenolide. Finally, we performed sequencing analysis on AGBL2 knockdown 786-O cells to investigate the correlation between AGBL2, immune infiltration, and AKT phosphorylation. Moreover, we experimentally demonstrated the enhancing effect of AGBL2 on AKT phosphorylation. Results TCGA analysis revealed a significant increase in AGBL2 expression in RCC patients, which was correlated with poorer overall survival (OS), disease-specific survival (DSS), and progression-free intervals (PFI). According to the analysis results, we constructed column-line plots to predict the 1-, 3-, and 5-year survival outcomes in RCC patients. Additionally, the calibration plots assessing the model's performance exhibited favorable agreement with the predicted outcomes. And the ROC curves showed that AGBL2 showed good diagnostic performance in KIRC (AUC = 0.836)). Cell phenotyping assays revealed that AGBL2 knockdown in RCC cells significantly inhibited cell proliferation and migration. Conversely, overexpression of AGBL2 resulted in increased cell proliferation and migration in RCC cells. We observed that AGBL2 is predominantly located in the nucleus and can elevate the detyrosination level of α-tubulin in RCC cells. Moreover, the enhancement of RCC cell proliferation and migration by AGBL2 was partially inhibited after treatment with the TCP inhibitor parthenolide. Analysis of the sequencing data revealed that AGBL2 is associated with a diverse array of biological processes, encompassing signal transduction and immune infiltration. Interestingly, AGBL2 expression exhibited a negative correlation with the majority of immune cell infiltrations. Additionally, AGBL2 was found to enhance the phosphorylation of AKT in RCC cells. Conclusion Our study suggests that AGBL2 fosters RCC cell proliferation and migration by enhancing α-tubulin detyrosination. Moreover, elevated AGBL2 expression increases phosphorylation of AKT in RCC cells.
Collapse
Affiliation(s)
- Wei Liu
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yifei Zhang
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yechen Nie
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yifu Liu
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhongqi Li
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhicheng Zhang
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Binbin Gong
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ming Ma
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| |
Collapse
|
2
|
Ragavi R, Muthukumaran P, Nandagopal S, Ahirwar DK, Tomo S, Misra S, Guerriero G, Shukla KK. Epigenetics regulation of prostate cancer: Biomarker and therapeutic potential. Urol Oncol 2023:S1078-1439(23)00090-X. [PMID: 37032230 DOI: 10.1016/j.urolonc.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023]
Abstract
Prostate cancer (CaP) is the second leading cause of cancer death and displays a broad range of clinical behavior from relatively indolent to aggressive metastatic disease. The etiology of most cases of CaP is not understood completely, which makes it imperative to search for the molecular basis of CaP and markers for early diagnosis. Epigenetic modifications, including changes in DNA methylation patterns, histone modifications, miRNAs, and lncRNAs are key drivers of prostate tumorigenesis. These epigenetic defects might be due to deregulated expression of the epigenetic machinery, affecting the expression of several important genes like GSTP1, RASSF1, CDKN2, RARRES1, IGFBP3, RARB, TMPRSS2-ERG, ITGB4, AOX1, HHEX, WT1, HSPE, PLAU, FOXA1, ASC, GPX3, EZH2, LSD1, etc. In this review, we highlighted the most important epigenetic gene alterations and their variations as a diagnostic marker and target for therapeutic intervention of CaP in the future. Characterization of epigenetic changes involved in CaP is obscure and adequate validation studies are still required to corroborate the present results that would be the impending future of transforming basic research settings into clinical practice.
Collapse
Affiliation(s)
- Ravindran Ragavi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | | | - Srividhya Nandagopal
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Dinesh Kumar Ahirwar
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Karwar, Jodhpur, Rajasthan, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sanjeev Misra
- Atal Bihari Vajpayee Medical University, Lucknow Uttar Pradesh, India
| | - Giulia Guerriero
- Comparative Endocrinology Lab, Department of Biology, University of Naples Federico II, Naples, Italy
| | - Kamla Kant Shukla
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India.
| |
Collapse
|
3
|
Contreras-Rodríguez JA, Puente-Rivera J, Córdova-Esparza DM, Nuñez-Olvera SI, Silva-Cázares MB. Bioinformatic miRNA-mRNAs Analysis Revels to miR-934 as a Potential Regulator of the Epithelial-Mesenchymal Transition in Triple-Negative Breast Cancer. Cells 2023; 12:cells12060834. [PMID: 36980175 PMCID: PMC10047237 DOI: 10.3390/cells12060834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/10/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer and has the worst prognosis. In patients with TNBC tumors, the tumor cells have been reported to have mesenchymal features, which help them migrate and invade. Various studies on cancer have revealed the importance of microRNAs (miRNAs) in different biological processes of the cell in that aberrations, in their expression, lead to alterations and deregulations in said processes, giving rise to tumor progression and aggression. In the present work, we determined the miRNAs that are deregulated in the epithelial-mesenchymal transition process in breast cancer. We discovered that 25 miRNAs that regulate mesenchymal genes are overexpressed in patients with TNBC. We found that miRNA targets modulate different processes and pathways, such as apoptosis, FoxO signaling pathways, and Hippo. We also found that the expression level of miR-934 is specific to the molecular subtype of the triple-negative breast cancer and modulates a set of related epithelial-mesenchymal genes. We determined that miR-934 inhibition in TNBC cell lines inhibits the migratory abilities of tumor cells.
Collapse
Affiliation(s)
| | | | | | - Stephanie I Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | | |
Collapse
|
4
|
Mechanistic insights into HuR inhibitor MS-444 arresting embryonic development revealed by low-input RNA-seq and STORM. Cell Biol Toxicol 2022; 38:1175-1197. [PMID: 36085230 DOI: 10.1007/s10565-022-09757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023]
Abstract
With improvements in the survival rate of patients with cancer, fertility maintenance has become a major concern in terms of cancer treatment for women of reproductive age. Thus, it is important to examine the impact on fertility of anticancer drugs that are used clinically or are undergoing trials. The HuR small-molecule inhibitor MS-444 has been used in many cancer treatment studies, but its reproductive toxicity in females is unknown. Here, we reported that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization, resulting in the developmental arrest of 2-cell stage embryos in mouse. Combining analysis of low-input RNA-seq for MS-444-treated 2-cell embryos and mapping binding sites of RNA-binding protein, Agbl2 was predicted to be the target gene of MS-444. For further confirmation, RNAi experiment in wild-type zygotes showed that Agbl2 knockdown reduced the proportion of embryos successfully developed to the blastocyst stage: from 71% in controls to 23%. Furthermore, RNA-FISH and luciferase reporter analyses showed that MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA and reduced its stability by inhibiting HuR dimerization. In addition, optimized stochastic optical reconstruction microscopy (STORM) imaging showed that MS-444 significantly reduced the HuR dimerization, and HuR mainly existed in cluster form in 2-cell stage embryos. In conclusion, this study provides clinical guidance for maintaining fertility during the treatment of cancer with MS-444 in women of reproductive age. And also, our research provides guidance for the application of STORM in nanometer scale studies of embryonic cells. HuR inhibitor MS-444 arrested embryonic development at 2-cell stage. Low-input RNA-seq revealed that Agbl2 was the target gene of MS-444. MS-444 blocked the nucleocytoplasmic transport of Agbl2 mRNA by inhibiting HuR dimerization and reduced the stability of Agbl2 mRNA. STORM with our optimized protocol showed that HuR tended to form elliptical and dense clusters in 2-cell stage embryos.
Collapse
|
5
|
Zhang D, Lu W, Cui S, Mei H, Wu X, Zhuo Z. Establishment of an ovarian cancer omentum metastasis-related prognostic model by integrated analysis of scRNA-seq and bulk RNA-seq. J Ovarian Res 2022; 15:123. [PMID: 36424614 PMCID: PMC9686070 DOI: 10.1186/s13048-022-01059-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Ovarian cancer has the highest mortality rate among gynecological malignant tumors, and it preferentially metastasizes to omental tissue, leading to intestinal obstruction and death. scRNA-seq is a powerful technique to reveal tumor heterogeneity. Analyzing omentum metastasis of ovarian cancer at the single-cell level may be more conducive to exploring and understanding omentum metastasis and prognosis of ovarian cancer at the cellular function and genetic levels. METHODS The omentum metastasis site scRNA-seq data of GSE147082 were acquired from the GEO (Gene Expression Omnibus) database, and single cells were clustered by the Seruat package and annotated by the SingleR package. Cell differentiation trajectories were reconstructed through the monocle package. The ovarian cancer microarray data of GSE132342 were downloaded from GEO and were clustered by using the ConsensusClusterPlus package into omentum metastasis-associated clusters according to the marker genes gained from single-cell differentiation trajectory analysis. The tumor microenvironment (TME) and immune infiltration differences between clusters were analyzed by the estimate and CIBERSORT packages. The expression matrix of genes used to cluster GSE132342 patients was extracted from bulk RNA-seq data of TCGA-OV (The Cancer Genome Atlas ovarian cancer), and least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression were performed to establish an omentum metastasis-associated gene (OMAG) signature. The signature was then tested by GSE132342 data. Finally, the clinicopathological characteristics of TCGA-OV were screened by univariate and multivariate Cox regression analysis to draw the nomogram. RESULTS A total of 9885 cells from 6 patients were clustered into 18 cell clusters and annotated into 14 cell types. Reconstruction of differentiation trajectories divided the cells into 5 branches, and a total of 781 cell trajectory-related characteristic genes were obtained. A total of 3769 patients in GSE132342 were subtyped into 3 clusters by 74 cell trajectory-related characteristic genes. Kaplan-Meier (K-M) survival analysis showed that the prognosis of cluster 2 was the worst, P < 0.001. The TME analysis showed that the ESTIMATE score and stromal score in cluster 2 were significantly higher than those in the other two clusters, P < 0.001. The immune infiltration analysis showed differences in the fraction of 8 immune cells among the 3 clusters, P < 0.05. The expression data of 74 genes used for GEO clustering were extracted from 379 patients in TCGA-OV, and combined with survival information, 10 candidates for OMAGs were filtered by LASSO. By using multivariate Cox regression, the 6-OMAGs signature was established as RiskScore = 0.307*TIMP3 + 3.516*FBN1-0.109*IGKC + 0.209*RPL21 + 0.870*UCHL1 + 0.365*RARRES1. Taking TCGA-OV as the training set and GSE132342 as the test set, receiver operating characteristic (ROC) curves were drawn to verify the prognostic value of 6-OMAGs. Screened by univariate and multivariate Cox regression analysis, 3 (age, cancer status, primary therapy outcome) of 5 clinicopathological characteristics were used to construct the nomogram combined with risk score. CONCLUSION We constructed an ovarian cancer prognostic model related to omentum metastasis composed of 6-OMAGs and 3 clinicopathological features and analyzed the potential mechanism of these 6-OMAGs in ovarian cancer omental metastasis.
Collapse
Affiliation(s)
- Dongni Zhang
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Wenping Lu
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Shasha Cui
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Heting Mei
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xiaoqing Wu
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Zhili Zhuo
- grid.410318.f0000 0004 0632 3409Oncology Department, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
6
|
Oliveira CR, Knapp D, Elewa A, Gerber T, Gonzalez Malagon SG, Gates PB, Walters HE, Petzold A, Arce H, Cordoba RC, Subramanian E, Chara O, Tanaka EM, Simon A, Yun MH. Tig1 regulates proximo-distal identity during salamander limb regeneration. Nat Commun 2022; 13:1141. [PMID: 35241664 PMCID: PMC8894484 DOI: 10.1038/s41467-022-28755-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/10/2022] [Indexed: 12/22/2022] Open
Abstract
Salamander limb regeneration is an accurate process which gives rise exclusively to the missing structures, irrespective of the amputation level. This suggests that cells in the stump have an awareness of their spatial location, a property termed positional identity. Little is known about how positional identity is encoded, in salamanders or other biological systems. Through single-cell RNAseq analysis, we identified Tig1/Rarres1 as a potential determinant of proximal identity. Tig1 encodes a conserved cell surface molecule, is regulated by retinoic acid and exhibits a graded expression along the proximo-distal axis of the limb. Its overexpression leads to regeneration defects in the distal elements and elicits proximal displacement of blastema cells, while its neutralisation blocks proximo-distal cell surface interactions. Critically, Tig1 reprogrammes distal cells to a proximal identity, upregulating Prod1 and inhibiting Hoxa13 and distal transcriptional networks. Thus, Tig1 is a central cell surface determinant of proximal identity in the salamander limb. The mechanisms by which cells determine their position within the 3D space are poorly understood. Research in salamanders offers fresh insights into this question, uncovering Tig1 as a central determinant of proximo-distal identity in regeneration.
Collapse
Affiliation(s)
- Catarina R Oliveira
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Dunja Knapp
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany.
| | - Ahmed Elewa
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Tobias Gerber
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Sandra G Gonzalez Malagon
- Institute of Structural and Molecular Biology, University College London, London, UK.,Biomedical Research Institute, Foundation for Research and Technology, University of Ioannina Campus, 45115, Ioannina, Greece
| | - Phillip B Gates
- Institute of Structural and Molecular Biology, University College London, London, UK
| | - Hannah E Walters
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Andreas Petzold
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Hernan Arce
- Systems Biology Group, Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council (CONICET) and University of La Plata, La Plata, Argentina.,Instituto de Tecnología, Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina
| | - Rodrigo C Cordoba
- Systems Biology Group, Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council (CONICET) and University of La Plata, La Plata, Argentina
| | | | - Osvaldo Chara
- Systems Biology Group, Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council (CONICET) and University of La Plata, La Plata, Argentina.,Instituto de Tecnología, Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina.,Technische Universität Dresden, Center for Information Services and High Performance Computing ZIH, Dresden, Germany
| | - Elly M Tanaka
- Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maximina H Yun
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany. .,Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
7
|
Wang CH, Lu TJ, Wang LK, Wu CC, Chen ML, Kuo CY, Shyu RY, Tsai FM. Tazarotene-induced gene 1 interacts with Polo-like kinase 2 and inhibits cell proliferation in HCT116 colorectal cancer cells. Cell Biol Int 2021; 45:2347-2356. [PMID: 34314079 DOI: 10.1002/cbin.11681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 07/24/2021] [Indexed: 01/05/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) is considered to be a tumor suppressor gene that is highly expressed in normal or well-differentiated colon tissues, while downregulation of TIG1 expression occurs in poorly differentiated colorectal cancer (CRC) tissues. However, it is still unclear how TIG1 regulates the tumorigenesis of CRC. Polo-like kinases (Plks) are believed to play an important role in regulating the cell cycle. The performance of PLK2 in CRC is negatively correlated with the differentiation status of CRC tissues. Here, we found that PLK2 can induce the growth of CRC cells and that TIG1 can prevent PLK2 from promoting the proliferation of CRC cells. We also found that the expression of PLK2 in CRC cells was associated with low levels of Fbxw7 protein and increased expression of cyclin E1. When TIG1 was coexpressed with PLK2, the changes in Fbxw7/cyclin E1 levels induced by PLK2 were reversed. In contrast, silencing TIG1 promoted the proliferation of CRC, and when PLK2 was also silenced, the proliferation of CRC cells induced by TIG1 silencing was significantly inhibited. The above research results suggest that TIG1 can regulate the tumorigenesis of CRC by regulating the activity of PLK2.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Dermatology, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tzung-Ju Lu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
8
|
Sharad S, Allemang TC, Li H, Nousome D, Ku AT, Whitlock NC, Sowalsky AG, Cullen J, Sesterhenn IA, McLeod DG, Srivastava S, Dobi A. Age and Tumor Differentiation-Associated Gene Expression Based Analysis of Non-Familial Prostate Cancers. Front Oncol 2021; 10:584280. [PMID: 33575208 PMCID: PMC7870995 DOI: 10.3389/fonc.2020.584280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/03/2020] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer incidence in young men has increased. Patients diagnosed at an earlier age are likely to have aggressive prostate cancer and treatment decisions are continuing to be weighted by patient age and life expectancy. Identification of age-associated gene-expression signatures hold great potential to augment current and future treatment modalities. To investigate age-specific tumor associated gene signatures and their potential biomarkers for disease aggressiveness, this study was designed and stratified into well and poorly differentiated tumor types of young (42–58 years) and old (66–73 years) prostate cancer patients. The differentially expressed genes related to tumor-normal differences between non-familial prostate cancer patients were identified and several genes uniquely associated with the age and tumor differentiation are markedly polarized. Overexpressed genes known to be associated with somatic genomic alterations was predominantly found in young men, such as TMPRESS2-ERG and c-MYC. On the other hand, old men have mostly down-regulated gene expressions indicating the loss of protective genes and reduced cell mediated immunity indicated by decreased HLA-A and HLA-B expression. The normalization for the benign signatures between the age groups indicates a significant age and tumor dependent heterogeneity exists among the patients with a great potential for age-specific and tumor differentiation-based therapeutic stratification of prostate cancer.
Collapse
Affiliation(s)
- Shashwat Sharad
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Travis C Allemang
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Hua Li
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Darryl Nousome
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Anson Tai Ku
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Nichelle C Whitlock
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Jennifer Cullen
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States
| | | | - David G McLeod
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Shiv Srivastava
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Albert Dobi
- Center for Prostate Disease Research, John P. Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, United States.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| |
Collapse
|
9
|
Wang CH, Wang LK, Wu CC, Chen ML, Kuo CY, Shyu RY, Tsai FM. Cathepsin V Mediates the Tazarotene-induced Gene 1-induced Reduction in Invasion in Colorectal Cancer Cells. Cell Biochem Biophys 2020; 78:483-494. [PMID: 32918681 DOI: 10.1007/s12013-020-00940-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) is a retinoid acid receptor-responsive gene involved in cell differentiation and tumorigenesis. Aberrant methylation of CpG islands in the TIG1 promoter is found in multiple cancers. Currently, the exact mechanism underlying the anticancer effect of TIG1 is unknown. Here, we show that TIG1 interacts with cathepsin V (CTSV), which reduces CTSV stability and subsequently affects the production of activated urokinase-type plasminogen activator (uPA), an epithelial-mesenchymal transition-associated protein. Ectopic expression of CTSV increased the expression of activated uPA and the number of migrated and invaded cells, whereas ectopic TIG1 expression reversed the effects of CTSV on the uPA signaling pathway. Similar patterns in the production of activated uPA and number of migrated and invaded cells were also observed in TIG1-expressing and CTSV-knockdown cells. The results suggest that CTSV may participate in TIG1-regulated uPA activity and the associated downstream signaling pathway.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Dermatology, Taipei Tzuchi Hospital, Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou, Taoyuan, 333, Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, 202, Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| |
Collapse
|
10
|
Chowdhury A, Loaiza S, Yebra-Fernandez E, Nadal-Melsio E, Apperley JF, Khorashad JS. An ex vivo investigation of interactions between primary acute myeloid leukaemia and mesenchymal stromal cells yields novel therapeutic targets. Br J Haematol 2020; 190:e236-e239. [PMID: 32519342 DOI: 10.1111/bjh.16810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/11/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Avirup Chowdhury
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Sandra Loaiza
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Eva Yebra-Fernandez
- SIHMDS North West London Pathology, Imperial College Healthcare NHS Trust, London, UK
| | - Elisabet Nadal-Melsio
- SIHMDS North West London Pathology, Imperial College Healthcare NHS Trust, London, UK
| | - Jane F Apperley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Jamshid S Khorashad
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| |
Collapse
|
11
|
Expression of RARRES1 and AGBL2 and progression of conventional renal cell carcinoma. Br J Cancer 2020; 122:1818-1824. [PMID: 32307444 PMCID: PMC7283229 DOI: 10.1038/s41416-020-0798-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Approximately 15% of clinically localised conventional renal cell carcinoma (RCC) will develop metastasis within 5 years of follow-up. The aim of this study was to identify biomarkers predicting the postoperative tumour relapse. METHODS Tissue microarrays of conventional RCC from a cohort of 691 patients without metastasis at the time of operation were analysed by immunohistochemistry for the expression of carboxypeptase inhibitor RARRES1 and its substrate carboxypeptidase AGBL2. Univariate and multivariate Cox regression models were addressed to postoperative tumour relapse and the metastasis-free survival time was estimated by Kaplan-Meier analysis. RESULTS In multivariate analysis, the lack of staining or cytoplasmic staining of RARRES1 was a significant risk factor indicating five times higher risk of cancer relapse. Combining its co-expression with AGBL2, we found that RARRES1 cytoplasmic/negative and AGBL2-positive/negative staining is a significant risk factor for tumour progression indicating 11-15 times higher risk of cancer relapse, whereas the membranous RARRES1 expression, especially its co-expression with AGBL2, associated with excellent disease outcome. CONCLUSIONS RARRES1 and AGBL2 expression defines groups of patients at low and high risk of tumour progression and may direct an active surveillance to detect metastasis as early as possible and to apply adjuvant therapy.
Collapse
|
12
|
Mocker A, Schmidt M, Huebner H, Wachtveitl R, Cordasic N, Menendez-Castro C, Hartner A, Fahlbusch FB. Expression of Retinoid Acid Receptor-Responsive Genes in Rodent Models of Placental Pathology. Int J Mol Sci 2019; 21:ijms21010242. [PMID: 31905805 PMCID: PMC6981780 DOI: 10.3390/ijms21010242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 01/13/2023] Open
Abstract
In humans, retinoic acid receptor responders (RARRES) have been shown to be altered in third trimester placentas complicated by the pathologies preeclampsia (PE) and PE with intrauterine growth restriction (IUGR). Currently, little is known about the role of placental Rarres in rodents. Therefore, we examined the localization and expression of Rarres1 and 2 in placentas obtained from a Wistar rat model of isocaloric maternal protein restriction (E18.5, IUGR-like features) and from an eNOS-knockout mouse model (E15 and E18.5, PE-like features). In both rodent models, Rarres1 and 2 were mainly localized in the placental spongiotrophoblast and giant cells. Their placental expression, as well as the expression of the Rarres2 receptor chemokine-like receptor 1 (CmklR1), was largely unaltered at the examined gestational ages in both animal models. Our results have shown that RARRES1 and 2 may have different expression and roles in human and rodent placentas, thereby underlining immanent limitations of comparative interspecies placentology. Further functional studies are required to elucidate the potential involvement of these proteins in early placentogenesis.
Collapse
Affiliation(s)
- Alexander Mocker
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.M.); (M.S.); (C.M.-C.); (A.H.)
| | - Marius Schmidt
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.M.); (M.S.); (C.M.-C.); (A.H.)
| | - Hanna Huebner
- Department of Gynaecology and Obstetrics/Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Rainer Wachtveitl
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (R.W.); (N.C.)
| | - Nada Cordasic
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (R.W.); (N.C.)
| | - Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.M.); (M.S.); (C.M.-C.); (A.H.)
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.M.); (M.S.); (C.M.-C.); (A.H.)
| | - Fabian B. Fahlbusch
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany; (A.M.); (M.S.); (C.M.-C.); (A.H.)
- Correspondence: ; Tel.: +49-9131-853-3118; Fax: +49-9131-853-3714
| |
Collapse
|
13
|
Ruiz-Romero C, Lam MPY, Nilsson P, Önnerfjord P, Utz PJ, Van Eyk JE, Venkatraman V, Fert-Bober J, Watt FE, Blanco FJ. Mining the Proteome Associated with Rheumatic and Autoimmune Diseases. J Proteome Res 2019; 18:4231-4239. [PMID: 31599600 DOI: 10.1021/acs.jproteome.9b00360] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A steady increase in the incidence of osteoarthritis and other rheumatic diseases has been observed in recent decades, including autoimmune conditions such as rheumatoid arthritis, spondyloarthropathies, systemic lupus erythematosus, systemic sclerosis, and Sjögren's syndrome. Rheumatic and autoimmune diseases (RADs) are characterized by the inflammation of joints, muscles, or other connective tissues. In addition to often experiencing debilitating mobility and pain, RAD patients are also at a higher risk of suffering comorbidities such as cardiovascular or infectious events. Given the socioeconomic impact of RADs, broad research efforts have been dedicated to these diseases worldwide. In the present work, we applied literature mining platforms to identify "popular" proteins closely related to RADs. The platform is based on publicly available literature. The results not only will enable the systematic prioritization of candidates to perform targeted proteomics studies but also may lead to a greater insight into the key pathogenic processes of these disorders.
Collapse
Affiliation(s)
- Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica, INIBIC - Complejo Hospitalario Universitario de A Coruña, SERGAS , Universidad de A Coruña , A Coruña 15006 , Spain
| | - Maggie P Y Lam
- Department of Medicine, Division of Cardiology, Consortium for Fibrosis Research and Translation, Anschutz Medical Campus , University of Colorado Denver , Aurora , Colorado 80045 , United States
| | - Peter Nilsson
- Division of Affinity Proteomics, SciLifeLab, Department of Protein Science , KTH Royal Institute of Technology , Stockholm 17121 , Sweden
| | - Patrik Önnerfjord
- Department of Clinical Sciences, Section for Rheumatology and Molecular Skeletal Biology , Lund University , Lund 22184 , Sweden
| | - Paul J Utz
- Division of Immunology and Rheumatology , Stanford University School of Medicine ; Palo Alto , California 94304 , United States
| | - Jennifer E Van Eyk
- Department of Medicine and The Heart Institute , Cedars-Sinai Medical Center , Los Angeles , California 90048 , United States
| | - Vidya Venkatraman
- Department of Medicine and The Heart Institute , Cedars-Sinai Medical Center , Los Angeles , California 90048 , United States
| | - Justyna Fert-Bober
- Department of Medicine and The Heart Institute , Cedars-Sinai Medical Center , Los Angeles , California 90048 , United States
| | - Fiona E Watt
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology , University of Oxford , Oxford OX3 7FY , United Kingdom
| | - Francisco J Blanco
- Grupo de Investigación de Reumatología, INIBIC-Complejo Hospitalario Universitario de A Coruña, SERGAS , Departamento de Medicina Universidad de A Coruña , A Coruña 15006 , Spain
| |
Collapse
|
14
|
Tazarotene-Induced Gene 1 (TIG1) Interacts with Serine Protease Inhibitor Kazal-Type 2 (SPINK2) to Inhibit Cellular Invasion of Testicular Carcinoma Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6171065. [PMID: 31886233 PMCID: PMC6899300 DOI: 10.1155/2019/6171065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/23/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) encodes a protein that is a retinoid-regulated tumor suppressor. TIG1 is expressed in most normal tissues, and downregulation of TIG1 expression in multiple cancers is caused by promoter hypermethylation. Kazal-type serine protease inhibitor-2 (SPINK2) is a serine protease inhibitor, and the SPINK protein family has been shown to inhibit the expression of urokinase-type plasminogen activator (uPA). In addition, increased levels of uPA and the uPA receptor were observed in testicular cancer tissues. This study demonstrated that TIG1 interacts with SPINK2 in NT2/D1 testicular carcinoma cells. TIG1 and SPINK2 were highly expressed in normal testis tissues, while low expression levels of TIG1 and SPINK2 were found in testicular cancer tissues. TIG1 inhibited cell invasion, migration, and epithelial-mesenchymal transition (EMT) of NT2/D1 cells. SPINK2 enhanced TIG1-regulated uPA activity and EMT suppression, while silencing SPINK2 alleviated TIG1-mediated EMT regulation, cell migration, and invasion. Therefore, the results suggest that the interaction between TIG1 and SPINK2 plays an important role in the inhibition of testicular cancer cell EMT, and suppression is mediated through downregulation of the uPA/uPAR signaling pathway.
Collapse
|
15
|
He WP, Wang LL. High expression of AGBL2 is a novel prognostic factor of adverse outcome in patients with ovarian carcinoma. Oncol Lett 2019; 18:4900-4906. [PMID: 31612000 PMCID: PMC6781648 DOI: 10.3892/ol.2019.10829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/16/2019] [Indexed: 11/19/2022] Open
Abstract
The putative oncogenic role of ATP/GTP binding protein like 2 (AGBL2) in catalyzing α-tubulin detyrosination has recently been characterized in cancer. However, the status of AGBL2 expression in ovarian cancer and its potential clinical and prognostic significance remain unclear. In the present study, immunohistochemistry staining investigated the protein expression level of AGBL2 in paraffin-embedded pathological specimens from 30 normal ovaries, 35 ovarian cystadenomas, 38 borderline ovarian tumors and 165 invasive ovarian carcinomas. The association between AGBL2 expression and clinicopathological characteristics of patients was evaluated using the χ2 test or Fisher's exact test. The survival status of patients was assessed by receiver-operator curve analysis. The results demonstrated that high expression of AGBL2 was observed in 9% of cystadenomas cases, 21% of borderline tumors cases and 38% of ovarian carcinomas cases; however AGBL2 expression was not high in normal ovarian tissues (P<0.01). Furthermore, the results demonstrated that high expression of AGBL2 was associated with tumor histological grade, advanced pT/pN/pM and cancer stage according to the International Federation of Gynecology and Obstetrics (P<0.05). Following univariate survival analysis of the ovarian carcinoma groups, high expression of AGBL2 was significantly associated with shorter patient survival (P<0.001). In addition, multivariate analysis revealed that AGBL2 could be identified as a potential independent prognostic factor for overall survival in patients with ovarian carcinoma (P=0.004). Furthermore, the results demonstrated that AGBL2 expression was significantly associated with the expression of immunity related GTPase M (IRGM) (P=0.013) and LC3A/B (P=0.004). IRGM expression level was also significantly associated with LC3A/B expression level (P=0.023). These findings demonstrated that AGBL2 expression was high in ovarian carcinomas, which suggested that AGBL2 may participate in the acquisition of an aggressive phenotype and may therefore serve as an independent prognostic molecular marker.
Collapse
Affiliation(s)
- Wei-Peng He
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Li-Li Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
16
|
Maimouni S, Lee MH, Sung YM, Hall M, Roy A, Ouaari C, Hwang YS, Spivak J, Glasgow E, Swift M, Patel J, Cheema A, Kumar D, Byers S. Tumor suppressor RARRES1 links tubulin deglutamylation to mitochondrial metabolism and cell survival. Oncotarget 2019; 10:1606-1624. [PMID: 30899431 PMCID: PMC6422194 DOI: 10.18632/oncotarget.26600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
RARRES1, a retinoic acid regulated carboxypeptidase inhibitor associated with fatty acid metabolism, stem cell differentiation and tumorigenesis is among the most commonly methylated loci in multiple cancers but has no known mechanism of action. Here we show that RARRES1 interaction with cytoplasmic carboxypeptidase 2 (CCP2) inhibits tubulin deglutamylation, which in turn regulates the mitochondrial voltage dependent anion channel (VDAC1), mitochondrial membrane potential, AMPK activation, energy balance and metabolically reprograms cells and zebrafish to a more energetic and anabolic phenotype. Depletion of RARRES1 also increases expression of stem cell markers, promotes anoikis, anchorage independent growth and insensitivity to multiple apoptotic stimuli. As depletion of CCP2 or inhibition of VDAC1 reverses the effects of RARRES1 depletion on energy balance and cell survival we conclude that RARRES1 modulation of CCP2-modulated tubulin-mitochondrial VDAC1 interactions is a fundamental regulator of cancer and stem cell metabolism and survival.
Collapse
Affiliation(s)
- Sara Maimouni
- Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| | - Mi-Hye Lee
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - You-Me Sung
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Michael Hall
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Arpita Roy
- University of the District of Columbia, Washington, DC, USA
| | - Chokri Ouaari
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.,University of the District of Columbia, Washington, DC, USA
| | - Yoo-Seok Hwang
- Cancer & Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, MD, USA
| | - Justin Spivak
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eric Glasgow
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Matthew Swift
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Jay Patel
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Amrita Cheema
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Deepak Kumar
- University of the District of Columbia, Washington, DC, USA
| | - Stephen Byers
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.,Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, DC, USA
| |
Collapse
|
17
|
Maimouni S, Issa N, Cheng S, Ouaari C, Cheema A, Kumar D, Byers S. Tumor suppressor RARRES1- A novel regulator of fatty acid metabolism in epithelial cells. PLoS One 2018; 13:e0208756. [PMID: 30557378 PMCID: PMC6296515 DOI: 10.1371/journal.pone.0208756] [Citation(s) in RCA: 12] [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: 07/07/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Retinoic acid receptor responder 1 (RARRES1) is silenced in many cancers and is differentially expressed in metabolism associated diseases, such as hepatic steatosis, hyperinsulinemia and obesity. Here we report a novel function of RARRES1 in metabolic reprogramming of epithelial cells. Using non-targeted LC-MS, we discovered that RARRES1 depletion in epithelial cells caused a global increase in lipid synthesis. RARRES1-depleted cells rewire glucose metabolism by switching from aerobic glycolysis to glucose-dependent de novo lipogenesis (DNL). Treatment with fatty acid synthase (FASN) inhibitor, C75, reversed the effects of RARRES1 depletion. The increased DNL in RARRES1-depleted normal breast and prostate epithelial cells proved advantageous to the cells during starvation, as the increase in fatty acid availability lead to more oxidized fatty acids (FAO), which were used for mitochondrial respiration. Expression of RARRES1 in several common solid tumors is also contextually correlated with expression of fatty acid metabolism genes and fatty acid-regulated transcription factors. Pathway enrichment analysis led us to determine that RARRES1 is regulated by peroxisome proliferating activated receptor (PPAR) signaling. These findings open up a new avenue for metabolic reprogramming and identify RARRES1 as a potential target for cancers and other diseases with impaired fatty acid metabolism.
Collapse
Affiliation(s)
- Sara Maimouni
- Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States of America
| | - Naiem Issa
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia, United States of America
| | - Selina Cheng
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia, United States of America
| | - Chokri Ouaari
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia, United States of America
- University of the District of Columbia, Washington, District of Columbia, United States of America
| | - Amrita Cheema
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia, United States of America
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
| | - Stephen Byers
- Department of Biochemical, Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States of America
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia, United States of America
| |
Collapse
|
18
|
Breast cancer subtype dictates DNA methylation and ALDH1A3-mediated expression of tumor suppressor RARRES1. Oncotarget 2018; 7:44096-44112. [PMID: 27286452 PMCID: PMC5190082 DOI: 10.18632/oncotarget.9858] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/09/2016] [Indexed: 11/25/2022] Open
Abstract
Breast cancer subtyping, based on the expression of hormone receptors and other genes, can determine patient prognosis and potential options for targeted therapy. Among breast cancer subtypes, tumors of basal-like and claudin-low subtypes are typically associated with worse patient outcomes, are primarily classified as triple-negative breast cancers (TNBC), and cannot be treated with existing hormone-receptor-targeted therapies. Understanding the molecular basis of these subtypes will lead to the development of more effective treatment options for TNBC. In this study, we focus on retinoic acid receptor responder 1 (RARRES1) as a paradigm to determine if breast cancer subtype dictates protein function and gene expression regulation. Patient tumor dataset analysis and gene expression studies of a 26 cell-line panel, representing the five breast cancer subtypes, demonstrate that RARRES1 expression is greatest in basal-like TNBCs. Cell proliferation and tumor growth assays reveal that RARRES1 is a tumor suppressor in TNBC. Furthermore, gene expression studies, Illumina HumanMethylation450 arrays, and chromatin immunoprecipitation demonstrate that expression of RARRES1 is retained in basal-like breast cancers due to hypomethylation of the promoter. Additionally, expression of the cancer stem cell marker, aldehyde dehydrogenase 1A3, which provides the required ligand (retinoic acid) for RARRES1 transcription, is also specific to the basal-like subtype. We functionally demonstrate that the combination of promoter methylation and retinoic acid signaling dictates expression of tumor suppressor RARRES1 in a subtype-specific manner. These findings provide a precedent for a therapeutically-inducible tumor suppressor and suggest novel avenues of therapeutic intervention for patients with basal-like breast cancer.
Collapse
|
19
|
Huebner H, Strick R, Wachter DL, Kehl S, Strissel PL, Schneider-Stock R, Hartner A, Rascher W, Horn LC, Beckmann MW, Ruebner M, Fahlbusch FB. Hypermethylation and loss of retinoic acid receptor responder 1 expression in human choriocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:165. [PMID: 29169400 PMCID: PMC5701501 DOI: 10.1186/s13046-017-0634-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/13/2017] [Indexed: 01/06/2023]
Abstract
Background Human placental development resembles tumorigenesis, due to the invasive and fusogenic potential of trophoblasts. However, these features are tightly controlled in trophoblasts. Disturbance of this spatial and temporal regulation is thought to contribute to the rare formation of choriocarcinomas. Promoter hypermethylation and loss of the tumor suppressor Retinoic acid receptor responder 1 (RARRES1) were shown to contribute to cancer progression. Our study investigated the epigenetic and transcriptional regulation of RARRES1 in healthy human placenta in comparison to choriocarcinoma cell lines and cases. Methods Three choriocarcinoma cell lines (Jeg-3, JAR and BeWo) were treated with three different retinoic acid derivates (Am580, Tazarotene and all-trans retinoic acid) and 5-aza-2′-deoxycytidine. We analyzed RARRES1 promoter methylation by pyrosequencing and performed realtime-PCR quantification to determine RARRES1 expression in placental tissue and trophoblastic cell lines. Additionally, RARRES1 was stained in healthy placentas and in biopsies of choriocarcinoma cases (n = 10) as well as the first trimester trophoblast cell line Swan71 by immunofluorescence and immunohistochemistry. Results In the choriocarcinoma cell lines, RARRES1 expression could not be induced by sole retinoic acid treatment. Stimulation with 5-aza-2′-deoxycytidine significantly induced RARRES1 expression, which then could be further increased with Am580, Tazarotene and all-trans retinoic acid. In comparison to healthy placenta, choriocarcinoma cell lines showed a hypermethylation of the RARRES1 promoter, which correlated with a reduced RARRES1 expression. In concordance, RARRES1 protein expression was lost in choriocarcinoma tissue. Additionally, in the trophoblastic cell line Swan71, we found a significant induction of RARRES1 expression with increased cell density, during mitosis and in syncytial knots. Conclusions Our findings showed that RARRES1 expression is absent in choriocarcinoma due to promoter methylation. Based on our analysis, we hypothesize that RARRES1 might exert tumor suppressive functions in multiple cellular processes (e.g. cell cycle regulation, adhesion, invasion and apoptosis). Electronic supplementary material The online version of this article (10.1186/s13046-017-0634-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- H Huebner
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - R Strick
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - D L Wachter
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - S Kehl
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - P L Strissel
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - R Schneider-Stock
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - A Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestraße 15, 91054 Erlangen, Erlangen, Germany
| | - W Rascher
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestraße 15, 91054 Erlangen, Erlangen, Germany
| | - L C Horn
- Division Molecular Pathology, Institute of Pathology, University of Leipzig, Leipzig, Germany
| | - M W Beckmann
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - M Ruebner
- Department of Gynaecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - F B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Loschgestraße 15, 91054 Erlangen, Erlangen, Germany.
| |
Collapse
|
20
|
AGBL2 promotes cancer cell growth through IRGM-regulated autophagy and enhanced Aurora A activity in hepatocellular carcinoma. Cancer Lett 2017; 414:71-80. [PMID: 29126912 DOI: 10.1016/j.canlet.2017.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023]
Abstract
AGBL2 has been reported to catalyze α-tubulin detyrosination, by which it promotes tumorigenesis and cancer progression. However, its potential role in the pathogenesis of hepatocellular carcinoma (HCC) has not been revealed yet. In the present study, AGBL2 was frequently found being overexpressed in HCC tissues and cell lines. In a large cohort of clinical HCC tissues, high expression of AGBL2 was positively associated with tumor size, tumor multiplicity and advanced clinical stage (p < 0.05), and it was an independent prognostic factor for HCC patients. In HCC cell lines, ectopic overexpression of AGBL2 substantially enhanced HCC cells survival and proliferation in vitro and promoted tumor growth in vivo. In addition, we demonstrated that overexpression of AGBL2 in HCC cells notably inhibited apoptosis by enhancing IRGM-regulated autophagy. Meanwhile, AGBL2 could up-regulate the expression of TPX2 and Aurora A activity to promote cell proliferation in HCC cells. In summary, our findings suggest that up-regulation of AGBL2 plays a critical oncogenic role in the pathogenesis of HCC through modulation on autophagy and Aurora A activity, and it could be a candidate for prognostic marker and therapeutic target in HCC.
Collapse
|
21
|
Roy A, Ramalinga M, Kim OJ, Chijioke J, Lynch S, Byers S, Kumar D. Multiple roles of RARRES1 in prostate cancer: Autophagy induction and angiogenesis inhibition. PLoS One 2017; 12:e0180344. [PMID: 28678839 PMCID: PMC5498036 DOI: 10.1371/journal.pone.0180344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/14/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) poses a major health concern in men worldwide. Retinoic Acid Receptor Responder (RARRES1)/ Tazarotene-induced gene-1 (TIG-1) is a putative tumor suppressor gene that exerts its tumor suppressor function via unknown mechanisms. Epigenetic silencing of RARRES1 leads to its loss in several types of cancer, including PCa. Determining the molecular mechanisms that mediate the tumor suppressor role of RARRES1 in PCa is the focus of our study. FINDINGS Our data indicates that RARRES1 over expression in PCa cell lines represses mitogen-activated protein kinase (MAPK) activation. RARRES1 expression induces the levels of autophagy-related genes, beclin, ATG3 and increases LC3B-II conversion. A significant induction of SIRT1 along with mTOR inhibition is noted on RARRES1 expression. Furthermore, RARRES1 over expression elevates the levels of the antioxidant enzyme, catalase. Our results also indicate that RARRES1 expression inhibits angiogenesis in endothelial cells. CONCLUSIONS In summary, the data presented here indicate that forced expression of RARRES1 in PCa cells (a) induces ER stress and autophagic response; (b) increases SIRT1 levels; and (c) higher levels of anti-oxidant enzymes. Our study also implicates the role of RARRES1 as a novel anti-angiogenic molecule. Overall this study reports the molecular players that RARRES1 modulates to serve as a tumor suppressor molecule. Future studies will help determine the in vivo mechanisms by which RARRES1 may serve as a target for therapeutic intervention both in cancer and in angiogenesis-related disorders.
Collapse
Affiliation(s)
- Arpita Roy
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
| | - Malathi Ramalinga
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
| | - Okjin J. Kim
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
| | - Juliet Chijioke
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
| | - Solomon Lynch
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
| | - Stephen Byers
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
| | - Deepak Kumar
- Cancer Research Laboratory, University of the District of Columbia, Washington, DC, United States of America
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States of America
- JLC-BBRI Nutrition Research lab, North Carolina Central University, Kannapolis, North Carolina, United States of America
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
22
|
Zhang M, Osisami M, Dai J, Keller JM, Escara-Wilke J, Mizokami A, Keller ET. Bone Microenvironment Changes in Latexin Expression Promote Chemoresistance. Mol Cancer Res 2017; 15:457-466. [PMID: 28087740 DOI: 10.1158/1541-7786.mcr-16-0392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
Although docetaxel is the standard of care for advanced prostate cancer, most patients develop resistance to docetaxel. Therefore, elucidating the mechanism that underlies resistance to docetaxel is critical to enhance therapeutic intervention. Mining cDNA microarray from the PC-3 prostate cancer cell line and its docetaxel-resistant derivative (PC3-TxR) revealed decreased latexin (LXN) expression in the resistant cells. LXN expression was inversely correlated with taxane resistance in a panel of prostate cancer cell lines. LXN knockdown conferred docetaxel resistance to prostate cancer cells in vitro and in vivo, whereas LXN overexpression reduced docetaxel resistance in several prostate cancer cell lines. A mouse model of prostate cancer demonstrated that prostate cancer cells developed resistance to docetaxel in the bone microenvironment, but not the soft tissue microenvironment. This was associated with decreased LXN expression in prostate cancer cells in the bone microenvironment compared with the soft tissue microenvironment. It was identified that bone stromal cells decreased LXN expression through methylation and induced chemoresistance in prostate cancer in vitro These findings reveal that a subset of prostate cancer develops docetaxel resistance through loss of LXN expression associated with methylation and that the bone microenvironment promotes this drug resistance phenotype.Implications: This study suggests that the LXN pathway should be further explored as a viable target for preventing or reversing taxane resistance in prostate cancer. Mol Cancer Res; 15(4); 457-66. ©2017 AACR.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Urology, University of Michigan, Ann Arbor, Michigan.,Clinical Medicine Program, Xiangya Hospital, Central South University, Changsha, China
| | - Mary Osisami
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Jinlu Dai
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Jill M Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan.,Unit for Laboratory Animal Medicine, University of Michigan; Ann Arbor, Michigan
| | | | | | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan. .,Biointerfaces Institute, University of Michigan; Ann Arbor, Michigan
| |
Collapse
|
23
|
Zinc and zinc-containing biomolecules in childhood brain tumors. J Mol Med (Berl) 2016; 94:1199-1215. [PMID: 27638340 DOI: 10.1007/s00109-016-1454-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Zinc ions are essential cofactors of a wide range of enzymes, transcription factors, and other regulatory proteins. Moreover, zinc is also involved in cellular signaling and enzymes inhibition. Zinc dysregulation, deficiency, over-supply, and imbalance in zinc ion transporters regulation are connected with various diseases including cancer. A zinc ion pool is maintained by two types of proteins: (i) zinc-binding proteins, which act as a buffer and intracellular donors of zinc and (ii) zinc transporters responsible for zinc fluxes into/from cells and organelles. The decreased serum zinc ion levels have been identified in patients suffering from various cancer diseases, including head and neck tumors and breast, prostate, liver, and lung cancer. On the contrary, increased zinc ion levels have been found in breast cancer and other malignant tissues. Zinc metalloproteomes of a majority of tumors including brain ones are still not yet fully understood. Current knowledge show that zinc ion levels and detection of certain zinc-containing proteins may be utilized for diagnostic and prognostic purposes. In addition, these proteins can also be promising therapeutic targets. The aim of the present work is an overview of the importance of zinc ions, zinc transporters, and zinc-containing proteins in brain tumors, which are, after leukemia, the second most common type of childhood cancer and the second leading cause of death in children after accidents.
Collapse
|
24
|
Genes that Affect Brain Structure and Function Identified by Rare Variant Analyses of Mendelian Neurologic Disease. Neuron 2016; 88:499-513. [PMID: 26539891 DOI: 10.1016/j.neuron.2015.09.048] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 09/12/2015] [Accepted: 09/25/2015] [Indexed: 11/23/2022]
Abstract
Development of the human nervous system involves complex interactions among fundamental cellular processes and requires a multitude of genes, many of which remain to be associated with human disease. We applied whole exome sequencing to 128 mostly consanguineous families with neurogenetic disorders that often included brain malformations. Rare variant analyses for both single nucleotide variant (SNV) and copy number variant (CNV) alleles allowed for identification of 45 novel variants in 43 known disease genes, 41 candidate genes, and CNVs in 10 families, with an overall potential molecular cause identified in >85% of families studied. Among the candidate genes identified, we found PRUNE, VARS, and DHX37 in multiple families and homozygous loss-of-function variants in AGBL2, SLC18A2, SMARCA1, UBQLN1, and CPLX1. Neuroimaging and in silico analysis of functional and expression proximity between candidate and known disease genes allowed for further understanding of genetic networks underlying specific types of brain malformations.
Collapse
|
25
|
Chakrabarti KR, Hessler L, Bhandary L, Martin SS. Molecular Pathways: New Signaling Considerations When Targeting Cytoskeletal Balance to Reduce Tumor Growth. Clin Cancer Res 2015; 21:5209-5214. [PMID: 26463706 DOI: 10.1158/1078-0432.ccr-15-0328] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/28/2015] [Indexed: 12/20/2022]
Abstract
The dynamic balance between microtubule extension and actin contraction regulates mammalian cell shape, division, and motility, which has made the cytoskeleton an attractive and very successful target for cancer drugs. Numerous compounds in clinical use to reduce tumor growth cause microtubule breakdown (vinca alkaloids, colchicine-site, and halichondrins) or hyperstabilization of microtubules (taxanes and epothilones). However, both of these strategies indiscriminately alter the assembly and dynamics of all microtubules, which causes significant dose-limiting toxicities on normal tissues. Emerging data are revealing that posttranslational modifications of tubulin (detyrosination, acetylation) or microtubule-associated proteins (Tau, Aurora kinase) may allow for more specific targeting of microtubule subsets, thereby avoiding the broad disruption of all microtubule polymerization. Developing approaches to reduce tumor cell migration and invasion focus on disrupting actin regulation by the kinases SRC and ROCK. Because the dynamic balance between microtubule extension and actin contraction also regulates cell fate decisions and stem cell characteristics, disrupting this cytoskeletal balance could yield unexpected effects beyond tumor growth. This review will examine recent data demonstrating that cytoskeletal cancer drugs affect wound-healing responses, microtentacle-dependent reattachment efficiency, and stem cell characteristics in ways that could affect the metastatic potential of tumor cells, both beneficially and detrimentally.
Collapse
Affiliation(s)
- Kristi R Chakrabarti
- Marlene and Stewart Greenebaum NCI Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA.,Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Lindsay Hessler
- Marlene and Stewart Greenebaum NCI Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA.,General Surgery Residency Program, University of Maryland Medical Center, 22 S. Greene Street, Baltimore, MD 21201, USA
| | - Lekhana Bhandary
- Marlene and Stewart Greenebaum NCI Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA.,Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Stuart S Martin
- Marlene and Stewart Greenebaum NCI Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, USA.,Program in Molecular Medicine, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA.,Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Namba T, Funahashi Y, Nakamuta S, Xu C, Takano T, Kaibuchi K. Extracellular and Intracellular Signaling for Neuronal Polarity. Physiol Rev 2015; 95:995-1024. [PMID: 26133936 DOI: 10.1152/physrev.00025.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurons are one of the highly polarized cells in the body. One of the fundamental issues in neuroscience is how neurons establish their polarity; therefore, this issue fascinates many scientists. Cultured neurons are useful tools for analyzing the mechanisms of neuronal polarization, and indeed, most of the molecules important in their polarization were identified using culture systems. However, we now know that the process of neuronal polarization in vivo differs in some respects from that in cultured neurons. One of the major differences is their surrounding microenvironment; neurons in vivo can be influenced by extrinsic factors from the microenvironment. Therefore, a major question remains: How are neurons polarized in vivo? Here, we begin by reviewing the process of neuronal polarization in culture conditions and in vivo. We also survey the molecular mechanisms underlying neuronal polarization. Finally, we introduce the theoretical basis of neuronal polarization and the possible involvement of neuronal polarity in disease and traumatic brain injury.
Collapse
Affiliation(s)
- Takashi Namba
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Funahashi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chundi Xu
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Takano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
27
|
Gardeux V, Chelouah R, Wanderley MFB, Siarry P, Braga AP, Reyal F, Rouzier R, Pusztai L, Natowicz R. Computing molecular signatures as optima of a bi-objective function: method and application to prediction in oncogenomics. Cancer Inform 2015; 14:33-45. [PMID: 25983540 PMCID: PMC4426938 DOI: 10.4137/cin.s21111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/14/2014] [Accepted: 12/17/2014] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Filter feature selection methods compute molecular signatures by selecting subsets of genes in the ranking of a valuation function. The motivations of the valuation functions choice are almost always clearly stated, but those for selecting the genes according to their ranking are hardly ever explicit. METHOD We addressed the computation of molecular signatures by searching the optima of a bi-objective function whose solution space was the set of all possible molecular signatures, ie, the set of subsets of genes. The two objectives were the size of the signature–to be minimized–and the interclass distance induced by the signature–to be maximized–. RESULTS We showed that: 1) the convex combination of the two objectives had exactly n optimal non empty signatures where n was the number of genes, 2) the n optimal signatures were nested, and 3) the optimal signature of size k was the subset of k top ranked genes that contributed the most to the interclass distance. We applied our feature selection method on five public datasets in oncology, and assessed the prediction performances of the optimal signatures as input to the diagonal linear discriminant analysis (DLDA) classifier. They were at the same level or better than the best-reported ones. The predictions were robust, and the signatures were almost always significantly smaller. We studied in more details the performances of our predictive modeling on two breast cancer datasets to predict the response to a preoperative chemotherapy: the performances were higher than the previously reported ones, the signatures were three times smaller (11 versus 30 gene signatures), and the genes member of the signature were known to be involved in the response to chemotherapy. CONCLUSIONS Defining molecular signatures as the optima of a bi-objective function that combined the signature size and the interclass distance was well founded and efficient for prediction in oncogenomics. The complexity of the computation was very low because the optimal signatures were the sets of genes in the ranking of their valuation. Software can be freely downloaded from http://gardeux-vincent.eu/DeltaRanking.php
Collapse
Affiliation(s)
- Vincent Gardeux
- EISTI engineering school, Department of Computer Science, Cergy, France. ; LISSI laboratory, University of Paris-Est, Créteil, France
| | - Rachid Chelouah
- EISTI engineering school, Department of Computer Science, Cergy, France
| | - Maria F Barbosa Wanderley
- Federal University of Minas Gerais, Laboratório de Inteligência Computacional, Belo Horizonte, Brazil
| | - Patrick Siarry
- LISSI laboratory, University of Paris-Est, Créteil, France
| | - Antônio P Braga
- Federal University of Minas Gerais, Laboratório de Inteligência Computacional, Belo Horizonte, Brazil
| | - Fabien Reyal
- Curie Institute, Department of Translational Research, Paris, France
| | - Roman Rouzier
- Curie Institute, Department of Surgery, Paris, France
| | - Lajos Pusztai
- Breast Medical Oncology, Yale School of Medicine, New Haven, CT, USA
| | - René Natowicz
- ESIEE-Paris, University of Paris-Est, Noisy-le-Grand, France
| |
Collapse
|
28
|
Tanco S, Tort O, Demol H, Aviles FX, Gevaert K, Van Damme P, Lorenzo J. C-terminomics screen for natural substrates of cytosolic carboxypeptidase 1 reveals processing of acidic protein C termini. Mol Cell Proteomics 2014; 14:177-90. [PMID: 25381060 DOI: 10.1074/mcp.m114.040360] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytosolic carboxypeptidases (CCPs) constitute a new subfamily of M14 metallocarboxypeptidases associated to axonal regeneration and neuronal degeneration, among others. CCPs are deglutamylating enzymes, able to catalyze the shortening of polyglutamate side-chains and the gene-encoded C termini of tubulin, telokin, and myosin light chain kinase. The functions of these enzymes are not entirely understood, in part because of the lack of information about C-terminal protein processing in the cell and its functional implications. By means of C-terminal COFRADIC, a positional proteomics approach, we searched for cellular substrates targets of CCP1, the most relevant member of this family. We here identified seven new putative CCP1 protein substrates, including ribosomal proteins, translation factors, and high mobility group proteins. Furthermore, we showed for the first time that CCP1 processes both glutamates as well as C-terminal aspartates. The implication of these C termini in molecular interactions furthermore suggests that CCP1-mediated shortening of acidic protein tails might regulate protein-protein and protein-DNA interactions.
Collapse
Affiliation(s)
- Sebastian Tanco
- From the ‡Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; §Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium; ¶Institute for Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Olivia Tort
- ¶Institute for Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Hans Demol
- From the ‡Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; §Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Francesc Xavier Aviles
- ¶Institute for Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Kris Gevaert
- From the ‡Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; §Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Petra Van Damme
- From the ‡Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; §Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium;
| | - Julia Lorenzo
- ¶Institute for Biotechnology and Biomedicine and Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
29
|
Whipple RA, Vitolo MI, Boggs AE, Charpentier MS, Thompson K, Martin SS. Parthenolide and costunolide reduce microtentacles and tumor cell attachment by selectively targeting detyrosinated tubulin independent from NF-κB inhibition. Breast Cancer Res 2014; 15:R83. [PMID: 24028602 PMCID: PMC3979133 DOI: 10.1186/bcr3477] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 07/22/2013] [Indexed: 12/13/2022] Open
Abstract
Introduction Detyrosinated tubulin, a post-translational modification of α-tubulin and a hallmark of stable microtubules, has gained recent attention given its association with tumor progression, invasiveness, and chemoresistance. We also recently reported that epithelial-to-mesenchymal transition (EMT) promotes tubulin detyrosination through tubulin tyrosine ligase (TTL) suppression. Furthermore, detyrosinated tubulin-enriched membrane protrusions, termed microtentacles (McTN), facilitate tumor cell reattachment to endothelial layers. Given the induction of EMT associated with inflammation and cancer progression, we tested anti-inflammatory nuclear factor-kappaB (NF-κB) inhibitors on a panel of human breast carcinoma cells to examine their effects on detyrosinated tubulin to identify more specific tubulin-directed anti-cancer treatments. Methods Using metastatic human breast carcinoma cells MDA-MB-157, MDA-MB-436, and Bt-549, we measured the impact of NF-κB inhibitors parthenolide, costunolide, and resveratrol on detyrosinated tubulin using protein expression analysis and immunofluorescence. A luciferase reporter assay and a viability screen were performed to determine if the effects were associated with their NF-κB inhibitory properties or were a result of apoptosis. Real-time monitoring of cell-substratum attachment was measured utilizing electrical impedance across microelectronic sensor arrays. We compared the selectivity of the NF-κB inhibitors to specifically target detyrosinated tubulin with traditional tubulin-targeted therapeutics, paclitaxel and colchicine, throughout the study. Results Sesquiterpene lactones, parthenolide and costunolide, selectively decrease detyrosinated tubulin independent of their inhibition of NF-κB. Live-cell scoring of suspended cells treated with parthenolide and costunolide show reduction in the frequency of microtentacles and inhibition of reattachment. Structural analysis shows that parthenolide and costunolide can decrease detyrosinated microtubules without significantly disrupting the overall microtubule network or cell viability. Paclitaxel and colchicine display indiscriminate disruption of the microtubule network. Conclusions Our data demonstrate that selective targeting of detyrosinated tubulin with parthenolide and costunolide can reduce McTN frequency and inhibit tumor cell reattachment. These actions are independent of their effects on NF-κB inhibition presenting a novel anti-cancer property and therapeutic opportunity to selectively target a stable subset of microtubules in circulating tumor cells to reduce metastatic potential with less toxicity in breast cancer patients.
Collapse
|
30
|
Tort O, Tanco S, Rocha C, Bièche I, Seixas C, Bosc C, Andrieux A, Moutin MJ, Avilés FX, Lorenzo J, Janke C. The cytosolic carboxypeptidases CCP2 and CCP3 catalyze posttranslational removal of acidic amino acids. Mol Biol Cell 2014; 25:3017-27. [PMID: 25103237 PMCID: PMC4230590 DOI: 10.1091/mbc.e14-06-1072] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The posttranslational modification of tubulin plays an important role in regulating microtubule function. Enzymes responsible for deglutamylating tubulin are members of a family of cytosolic carboxypeptidases. By completing the functional characterization of this protein family in mammals, it is demonstrated that CCP2 and CCP3 are deglutamylases. The posttranslational modification of carboxy-terminal tails of tubulin plays an important role in the regulation of the microtubule cytoskeleton. Enzymes responsible for deglutamylating tubulin have been discovered within a novel family of mammalian cytosolic carboxypeptidases. The discovery of these enzymes also revealed the existence of a range of other substrates that are enzymatically deglutamylated. Only four of six mammalian cytosolic carboxypeptidases had been enzymatically characterized. Here we complete the functional characterization of this protein family by demonstrating that CCP2 and CCP3 are deglutamylases, with CCP3 being able to hydrolyze aspartic acids with similar efficiency. Deaspartylation is a novel posttranslational modification that could, in conjunction with deglutamylation, broaden the range of potential substrates that undergo carboxy-terminal processing. In addition, we show that CCP2 and CCP3 are highly regulated proteins confined to ciliated tissues. The characterization of two novel enzymes for carboxy-terminal protein modification provides novel insights into the broadness of this barely studied process.
Collapse
Affiliation(s)
- Olivia Tort
- Institut de Biotecnologia i de Biomedicina, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain Institut Curie, 91405 Orsay, France
| | - Sebastián Tanco
- Institut de Biotecnologia i de Biomedicina, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain Department of Medical Protein Research, VIB, 9000 Ghent, Belgium Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Cecilia Rocha
- Institut Curie, 91405 Orsay, France PSL Research University, 75005 Paris, France Centre National de la Recherche Scientifique, UMR3306, 91405 Orsay, France Institut National de la Santé et de la Recherche Médicale, U1005, 91405 Orsay, France
| | - Ivan Bièche
- PSL Research University, 75005 Paris, France Department of Genetics, Institut Curie, 75248 Paris, France
| | - Cecilia Seixas
- Centro de Estudos de Doenças Crónicas, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Christophe Bosc
- Institut des Neurosciences de Grenoble, Institut National de la Santé et de la Recherche Médicale, U836, CEA, Université Joseph Fourier, 38042 Grenoble, France Université Grenoble Alpes, 38000 Grenoble, France CEA, Institut de Recherches en Technologies et Sciences pour le Vivant, 38000 Grenoble, France
| | - Annie Andrieux
- Institut des Neurosciences de Grenoble, Institut National de la Santé et de la Recherche Médicale, U836, CEA, Université Joseph Fourier, 38042 Grenoble, France Université Grenoble Alpes, 38000 Grenoble, France CEA, Institut de Recherches en Technologies et Sciences pour le Vivant, 38000 Grenoble, France
| | - Marie-Jo Moutin
- Institut des Neurosciences de Grenoble, Institut National de la Santé et de la Recherche Médicale, U836, CEA, Université Joseph Fourier, 38042 Grenoble, France Université Grenoble Alpes, 38000 Grenoble, France CEA, Institut de Recherches en Technologies et Sciences pour le Vivant, 38000 Grenoble, France
| | - Francesc Xavier Avilés
- Institut de Biotecnologia i de Biomedicina, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain
| | - Julia Lorenzo
- Institut de Biotecnologia i de Biomedicina, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain
| | - Carsten Janke
- Institut Curie, 91405 Orsay, France PSL Research University, 75005 Paris, France Centre National de la Recherche Scientifique, UMR3306, 91405 Orsay, France Institut National de la Santé et de la Recherche Médicale, U1005, 91405 Orsay, France
| |
Collapse
|
31
|
Meyer MA. Highly expressed genes in human high grade gliomas: immunohistochemical analysis of data from the human protein atlas. Neurol Int 2014; 6:5348. [PMID: 24987500 PMCID: PMC4077206 DOI: 10.4081/ni.2014.5348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/07/2014] [Indexed: 11/22/2022] Open
Abstract
Gene expression within human glioblastomas were analyzed from data on 20,083 genes entered into the on-line Human Protein Atlas. In selecting genes that are strongly expressed within normal human brain tissue, 58 genes were identified from a search of the 20,083 entries that were rated as showing 90% or greater intensity of expression within normal brain tissues. Of these 58, a subset of 48 genes was identified that not only had expression data for human glioblastomas but also for the human glioblastoma cell line U-251. Four of these 48 selected genes were found to be strongly expressed within the cytoplasm when assessed by both histologic sampling of high grade glioma patient cases as well as U-251 glioblastoma cell line immunofluoresence analysis. These four human genes are: AGBL2 (ATP/GTP binding protein-like 2), BLOC1S6 (biogenesis of lysosomal organelles complex-1, subunit 6), MAP1A (microtubule-associated protein 1A) and ZSWIM5 (zinc finger, SWIM-type containing 5, also known as KIAA1511). Further research is advocated to investigate the role of ZSWIM5 and AGBL2 in glioma cell biology.
Collapse
Affiliation(s)
- Michael A Meyer
- Department of Neurology, Sisters Hospital , Buffalo, NY, USA
| |
Collapse
|
32
|
Zhang H, Ren Y, Pang D, Liu C. Clinical implications of AGBL2 expression and its inhibitor latexin in breast cancer. World J Surg Oncol 2014; 12:142. [PMID: 24884516 PMCID: PMC4069086 DOI: 10.1186/1477-7819-12-142] [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: 08/31/2013] [Accepted: 04/20/2014] [Indexed: 11/29/2022] Open
Abstract
Background We investigated the expression status of AGBL2 and its inhibitor latexin in breast cancer stem cells and its clinical implications in order to lay a foundation for managing breast cancer. Methods CD44+/CD24- tumor cells (CSC) from clinical specimens were sorted using flow cytometry. AGBL2 expression status was detected in CSC and 126 breast cancer specimens by western blot and immunohistochemistry staining. The relationship between the AGBL2 protein and clinicopathological parameters and prognosis was subsequently determined. Result As a result, CSC are more likely to generate new tumors in mice and cell microspheres that are deficient in non-obese diabetic/severe combined immunodeficiency mice (NOD/SCID) compared to the control group. The AGBL2 protein was expressed higher in CSC induced to epithelial to mesenchymal transition (EMT) when compared to the control cells, and was found to be related to CSC chemotherapy resistance. After Spearman regression correlation analysis, AGBL2 was observed to be related to clinical stage, histological stage, and lymph node metastasis. In the Cox regression test, the AGBL2 protein was detected as an independent prognostic factor. Through immunoprecipitation, AGBL2 and latexin could form immune complexes. Conclusions These results demonstrate that AGBL2 is a latexin-interacting protein that regulates the tubulin tyrosination cycle and is a potential target for intervention.
Collapse
Affiliation(s)
| | | | | | - Caigang Liu
- Department of Breast Surgery, Second hospital of Dalian Medical University, Zhongshan Road, Dalian 116023, People's Republic of China.
| |
Collapse
|
33
|
Rimsa V, Eadsforth TC, Joosten RP, Hunter WN. High-resolution structure of the M14-type cytosolic carboxypeptidase from Burkholderia cenocepacia refined exploiting PDB_REDO strategies. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:279-89. [PMID: 24531462 PMCID: PMC3940198 DOI: 10.1107/s1399004713026801] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 09/30/2013] [Indexed: 01/01/2023]
Abstract
A potential cytosolic metallocarboxypeptidase from Burkholderia cenocepacia has been crystallized and a synchrotron-radiation microfocus beamline allowed the acquisition of diffraction data to 1.9 Å resolution. The asymmetric unit comprises a tetramer containing over 1500 amino acids, and the high-throughput automated protocols embedded in PDB_REDO were coupled with model-map inspections in refinement. This approach has highlighted the value of such protocols for efficient analyses. The subunit is constructed from two domains. The N-terminal domain has previously only been observed in cytosolic carboxypeptidase (CCP) proteins. The C-terminal domain, which carries the Zn2+-containing active site, serves to classify this protein as a member of the M14D subfamily of carboxypeptidases. Although eukaryotic CCPs possess deglutamylase activity and are implicated in processing modified tubulin, the function and substrates of the bacterial family members remain unknown. The B. cenocepacia protein did not display deglutamylase activity towards a furylacryloyl glutamate derivative, a potential substrate. Residues previously shown to coordinate the divalent cation and that contribute to peptide-bond cleavage in related enzymes such as bovine carboxypeptidase are conserved. The location of a conserved basic patch in the active site adjacent to the catalytic Zn2+, where an acetate ion is identified, suggests recognition of the carboxy-terminus in a similar fashion to other carboxypeptidases. However, there are significant differences that indicate the recognition of substrates with different properties. Of note is the presence of a lysine in the S1' recognition subsite that suggests specificity towards an acidic substrate.
Collapse
Affiliation(s)
- Vadim Rimsa
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Thomas C. Eadsforth
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Robbie P. Joosten
- Department of Biochemistry, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - William N. Hunter
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| |
Collapse
|
34
|
Lyons PJ, Sapio MR, Fricker LD. Zebrafish cytosolic carboxypeptidases 1 and 5 are essential for embryonic development. J Biol Chem 2013; 288:30454-30462. [PMID: 24022483 DOI: 10.1074/jbc.m113.497933] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cytosolic carboxypeptidases (CCPs) are a subfamily of metalloenzymes within the larger M14 family of carboxypeptidases that have been implicated in the post-translational modification of tubulin. It has been suggested that at least four of the six mammalian CCPs function as tubulin deglutamylases. However, it is not yet clear whether these enzymes play redundant or unique roles within the cell. To address this question, genes encoding CCPs were identified in the zebrafish genome. Analysis by quantitative polymerase chain reaction indicated that CCP1, CCP2, CCP5, and CCP6 mRNAs were detectable between 2 h and 8 days postfertilization with highest levels 5-8 days postfertilization. CCP1, CCP2, and CCP5 mRNAs were predominantly expressed in tissues such as the brain, olfactory placodes, and pronephric ducts. Morpholino oligonucleotide-mediated knockdown of CCP1 and CCP5 mRNA resulted in a common phenotype including ventral body curvature and hydrocephalus. Confocal microscopy of morphant zebrafish revealed olfactory placodes with defective morphology as well as pronephric ducts with increased polyglutamylation. These data suggest that CCP1 and CCP5 play important roles in developmental processes, particularly the development and functioning of cilia. The robust and similar defects upon knockdown suggest that each CCP may have a function in microtubule modification and ciliary function and that other CCPs are not able to compensate for the loss of one.
Collapse
Affiliation(s)
| | - Matthew R Sapio
- Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
35
|
Berezniuk I, Lyons PJ, Sironi JJ, Xiao H, Setou M, Angeletti RH, Ikegami K, Fricker LD. Cytosolic carboxypeptidase 5 removes α- and γ-linked glutamates from tubulin. J Biol Chem 2013; 288:30445-30453. [PMID: 24022482 DOI: 10.1074/jbc.m113.497917] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytosolic carboxypeptidase 5 (CCP5) is a member of a subfamily of enzymes that cleave C-terminal and/or side chain amino acids from tubulin. CCP5 was proposed to selectively cleave the branch point of glutamylated tubulin, based on studies involving overexpression of CCP5 in cell lines and detection of tubulin forms with antisera. In the present study, we examined the activity of purified CCP5 toward synthetic peptides as well as soluble α- and β-tubulin and paclitaxel-stabilized microtubules using a combination of antisera and mass spectrometry to detect the products. Mouse CCP5 removes multiple glutamate residues and the branch point glutamate from the side chains of porcine brain α- and β-tubulin. In addition, CCP5 excised C-terminal glutamates from detyrosinated α-tubulin. The enzyme also removed multiple glutamate residues from side chains and C termini of paclitaxel-stabilized microtubules. CCP5 both shortens and removes side chain glutamates from synthetic peptides corresponding to the C-terminal region of β3-tubulin, whereas cytosolic carboxypeptidase 1 shortens the side chain without cleaving the peptides' γ-linked residues. The rate of cleavage of α linkages by CCP5 is considerably slower than that of removal of a single γ-linked glutamate residue. Collectively, our data show that CCP5 functions as a dual-functional deglutamylase cleaving both α- and γ-linked glutamate from tubulin.
Collapse
Affiliation(s)
- Iryna Berezniuk
- From the Departments of Molecular Pharmacology and; Neuroscience and
| | | | | | - Hui Xiao
- the Laboratory of Macromolecular Analysis and Proteomics, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Mitsutoshi Setou
- the Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
| | - Ruth H Angeletti
- the Laboratory of Macromolecular Analysis and Proteomics, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Koji Ikegami
- the Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
| | - Lloyd D Fricker
- From the Departments of Molecular Pharmacology and; Neuroscience and.
| |
Collapse
|
36
|
Ullah M, Stich S, Häupl T, Eucker J, Sittinger M, Ringe J. Reverse differentiation as a gene filtering tool in genome expression profiling of adipogenesis for fat marker gene selection and their analysis. PLoS One 2013; 8:e69754. [PMID: 23922792 PMCID: PMC3724870 DOI: 10.1371/journal.pone.0069754] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 06/11/2013] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND During mesenchymal stem cell (MSC) conversion into adipocytes, the adipogenic cocktail consisting of insulin, dexamethasone, indomethacin and 3-isobutyl-1-methylxanthine not only induces adipogenic-specific but also genes for non-adipogenic processes. Therefore, not all significantly expressed genes represent adipogenic-specific marker genes. So, our aim was to filter only adipogenic-specific out of all expressed genes. We hypothesize that exclusively adipogenic-specific genes change their expression during adipogenesis, and reverse during dedifferentiation. Thus, MSC were adipogenic differentiated and dedifferentiated. RESULTS Adipogenesis and reverse adipogenesis was verified by Oil Red O staining and expression of PPARG and FABP4. Based on GeneChips, 991 genes were differentially expressed during adipogenesis and grouped in 4 clusters. According to bioinformatic analysis the relevance of genes with adipogenic-linked biological annotations, expression sites, molecular functions, signaling pathways and transcription factor binding sites was high in cluster 1, including all prominent adipogenic genes like ADIPOQ, C/EBPA, LPL, PPARG and FABP4, moderate in clusters 2-3, and negligible in cluster 4. During reversed adipogenesis, only 782 expressed genes (clusters 1-3) were reverted, including 597 genes not reported for adipogenesis before. We identified APCDD1, CHI3L1, RARRES1 and SEMA3G as potential adipogenic-specific genes. CONCLUSION The model system of adipogenesis linked to reverse adipogenesis allowed the filtration of 782 adipogenic-specific genes out of total 991 significantly expressed genes. Database analysis of adipogenic-specific biological annotations, transcription factors and signaling pathways further validated and valued our concept, because most of the filtered 782 genes showed affiliation to adipogenesis. Based on this approach, the selected and filtered genes would be potentially important for characterization of adipogenesis and monitoring of clinical translation for soft-tissue regeneration. Moreover, we report 4 new marker genes.
Collapse
Affiliation(s)
- Mujib Ullah
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Stefan Stich
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Thomas Häupl
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Jan Eucker
- Department of Hematology and Oncology, Charité-University Medicine Berlin, Berlin, Germany
| | - Michael Sittinger
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | - Jochen Ringe
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
37
|
Rovini A, Gauthier G, Bergès R, Kruczynski A, Braguer D, Honoré S. Anti-migratory effect of vinflunine in endothelial and glioblastoma cells is associated with changes in EB1 C-terminal detyrosinated/tyrosinated status. PLoS One 2013; 8:e65694. [PMID: 23750272 PMCID: PMC3672205 DOI: 10.1371/journal.pone.0065694] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 05/01/2013] [Indexed: 12/26/2022] Open
Abstract
We previously showed that vinflunine, a microtubule-targeting drug of the Vinca-alkaloid family exerted its anti-angiogenic/anti-migratory activities through an increase in microtubule dynamics and an inhibition of microtubule targeting to adhesion sites. Such effect was associated with a reduction of EB1 comet length at microtubule (+) ends. In this work we first showed that the pro-angiogenic vascular endothelial growth factor VEGF suppressed microtubule dynamics in living Human Umbilical Vein Endothelial Cells (HUVECs), increased EB1 comet length by 40%, and induced EB1 to bind all along the microtubules, without modifying its expression level. Such microtubule (+) end stabilization occurred close to the plasma membrane in the vicinity of focal adhesion as shown by TIRF microscopy experiments. Vinflunine completely abolished the effect of VEGF on EB1 comets. Interestingly, we found a correlation between the reduction of EB1 comet length by vinflunine and the inhibition of cell migration. By using 2D gel electrophoresis we demonstrated for the first time that EB1 underwent several post-translational modifications in endothelial and tumor cells. Particularly, the C-terminal EEY sequence was poorly detectable in control and VEGF-treated HUVECs suggesting the existence of a non-tyrosinated form of EB1. By using specific antibodies that specifically recognized and discriminated the native tyrosinated form of EB1 and a putative C-terminal detyrosinated form, we showed that a detyrosinated form of EB1 exists in HUVECs and tumor cells. Interestingly, vinflunine decreased the level of the detyrosinated form and increased the native tyrosinated form of EB1. Using 3-L-Nitrotyrosine incorporation experiments, we concluded that the EB1 C-terminal modifications result from a detyrosination/retyrosination cycle as described for tubulin. Altogether, our results show that vinflunine inhibits endothelial cell migration through an alteration of EB1 comet length and EB1 detyrosination/retyrosination cycle.
Collapse
Affiliation(s)
- Amandine Rovini
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale UMR_S 911, Marseille, France
| | - Géraldine Gauthier
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale UMR_S 911, Marseille, France
- APHM, Hôpital Timone, Marseille, France
| | - Raphaël Bergès
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale UMR_S 911, Marseille, France
| | - Anna Kruczynski
- Centre de Recherche d'Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Diane Braguer
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale UMR_S 911, Marseille, France
- APHM, Hôpital Timone, Marseille, France
| | - Stéphane Honoré
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale UMR_S 911, Marseille, France
- APHM, Hôpital Timone, Marseille, France
- * E-mail:
| |
Collapse
|
38
|
Oldridge EE, Walker HF, Stower MJ, Simms MS, Mann VM, Collins AT, Pellacani D, Maitland NJ. Retinoic acid represses invasion and stem cell phenotype by induction of the metastasis suppressors RARRES1 and LXN. Oncogenesis 2013; 2:e45. [PMID: 23588494 PMCID: PMC3641360 DOI: 10.1038/oncsis.2013.6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mouse haematopoietic stem cell (SC) regulator Latexin (LXN) is the only known homologue of the retinoic acid receptor responder 1 (RARRES1) gene. Both genes lie adjacent on chromosome 3 and differ mostly by the presence of a transmembrane domain in RARRES1. Despite their homology, it is not known whether they possess similar regulatory mechanisms, cellular localization and function. Here, we identified RARRES1 and LXN as highly significantly downregulated genes in human prostate SCs, whose expression was induced by the pro-differentiation agent all-trans retinoic acid (atRA). AtRA induced expression in the most differentiated cells compared with the SC fraction, suggesting that this subpopulation was less responsive to atRA. Small interfering RNA suppression of RARRES1 and LXN enhanced the SC properties of primary prostate cultures, as shown by a significant increase in their colony-forming ability. Expression of both RARRES1 and LXN was co-ordinately repressed by DNA methylation in prostate cancer cell lines and inhibition of RARRES1 and LXN increased the invasive capacity of primary prostate cultures, which also fully rescued an inhibitory effect induced by atRA. Moreover, we showed that RARRES1 and LXN reside within different sub-cellular compartments, providing evidence that RARRES1 is not a plasma membrane protein as previously supposed but is located primarily in the endoplasmic reticulum; whereas LXN was detected in the nucleus of prostate epithelial cells. Thus, LXN and RARRES1 are potential tumour suppressor genes, which are co-ordinately regulated, SC-silenced genes functioning to suppress invasion and colony-forming ability of prostate cancer cells; yet the proteins reside within different sub-cellular compartments.
Collapse
Affiliation(s)
- E E Oldridge
- YCR Cancer Research Unit, Department of Biology, University of York, York, UK
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu WB, Han F, Jiang X, Yang LJ, Li YH, Liu Y, Chen HQ, Ao L, Cui ZH, Cao J, Liu JY. ANKRD18A as a novel epigenetic regulation gene in lung cancer. Biochem Biophys Res Commun 2012; 429:180-5. [PMID: 23131552 DOI: 10.1016/j.bbrc.2012.10.116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 10/27/2012] [Indexed: 01/10/2023]
Abstract
Lung cancer is one of the most common causes of cancer-related mortality worldwide. Effective early diagnosis and targeted therapies for lung cancer to reduce incidence and mortality would benefit from a better understanding of the key molecular changes that occur from normal to malignant tumor cells during lung cancer initiation and development, but these are largely unknown. Previous studies have shown that DNA methylation, an important mechanism for the regulation of gene expression, plays a key role in lung carcinogenesis. In this study, we screened a novel methylation gene, ANKRD18A, encoding ankyrin repeat domain 18A, to determine whether it is regulated by DNA methylation in lung cancer. Methylation-specific PCR and bisulfite sequencing PCR were used to analyze gene methylation status, and real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) examined mRNA levels. Promoter hypermethylation of ANKRD18A was detected in 68.4% (26/38) of lung cancer tissues but not (0/20) in normal lung tissues (P<0.01), whereas ANKRD18A mRNA expression was significantly decreased in lung cancer tissues compared with adjacent normal tissues. In addition, we found that ANKRD18A expression was significantly decreased in 9 of 10 lung cancer cell lines. This was associated with hypermethylation of the ANKRD18A promoter region. Moreover, weak expression of ANKRD18A in methylated lung cancer cell lines increased markedly after treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. These results suggest that ANKRD18A hypermethylation and consequent mRNA alterations might be a vital molecular mechanism in lung cancer.
Collapse
Affiliation(s)
- Wen-Bin Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Key Laboratory of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Chongqing 400038, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rodríguez de la Vega Otazo M, Lorenzo J, Tort O, Avilés FX, Bautista JM. Functional segregation and emerging role of cilia-related cytosolic carboxypeptidases (CCPs). FASEB J 2012; 27:424-31. [PMID: 23085998 DOI: 10.1096/fj.12-209080] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent experimental data indicating axonal regeneration, axogenesis, dendritogenesis, and ciliary axoneme assembly and wellness have linked the role of cytosolic metallocarboxypeptidase 1 (CCP1/AGTPBP1/Nna1) to the microtubule network. In addition, 5 of the 6 mammalian ccp genes have been shown to participate in post-translational modifications of tubulin, which occur in the microtubules of neurons, mitotic spindles, cilia, and basal bodies. Here, we compile evidence for the idea that the occurrence of CCPs strongly correlates with the presence of cilia, suggesting that CCP functions might be primarily related to cilia and basal bodies (CBBs). In agreement with this hypothesis, CCPs were localized in centrioles, basal bodies, and mitotic spindles in HeLa cells by confocal microscopy. By reconstructing the evolutionary history of CCPs, we show their presence in the last eukaryotic common ancestor and relate each group of CCP orthologs to specific roles in CBBs. The clues presented in this study suggest that during the evolution of eukaryotes, mechanisms mediated by CCPs through tubulin post-translational modifications controlling assembly, trafficking, and signaling in the microtubules, were transferred from cilia to cell and axon microtubules.
Collapse
|
41
|
A combined approach identifies three mRNAs that are down-regulated by microRNA-29b and promote invasion ability in the breast cancer cell line MCF-7. J Cancer Res Clin Oncol 2012; 138:2127-36. [PMID: 22864815 DOI: 10.1007/s00432-012-1288-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND AND AIMS MicroRNAs (miRNAs) are short noncoding RNAs that regulate gene expression by targeting mRNAs. Our previous study found that miR-29b strongly regulates the migration and invasion of breast cancer cells. Here, we aimed to identify the mRNAs targeted by miR-29b. METHODS We used microarray experiments in conjunction with computational methods to identify the mRNAs that were most susceptible to miR-29b-mediated repression. We further confirmed the activities of three target genes, C1QTNF6, SPARC, and COL4A2, by luciferase reporter analyses and invasion assays. RESULTS We evaluated the impact of miR-29b on global mRNA expression in MCF-7 human breast cancer cells through microarray analysis and further analyzed four genes that were at least twofold down-regulated and predicted as miR-29b targets by at least two of the four widely used miRNA target prediction algorithms. We also analyzed one mRNA that was down-regulated by 1.8-fold but was predicted to have significant interactions with miR-29b in pathway analysis and was predicted as a miR-29b target by all four algorithms. Luciferase reporter and invasion assays revealed that C1QTNF6, SPARC, and COL4A2 were targeted by miR-29b and that the degradation of any one of these mRNAs could promote invasion in MCF-7 cells. CONCLUSIONS C1QTNF6, SPARC, and COL4A2 are targeted by miR-29b, and the down-regulation of these three mRNAs can contribute to the invasion ability of MCF-7 cells.
Collapse
|
42
|
Chen HL, Seol H, Brown KJ, Gordish-Dressman H, Hill A, Gallo V, Packer R, Hathout Y. Secretome survey of human plexiform neurofibroma derived Schwann Cells reveals a secreted form of the RARRES1 protein. Int J Mol Sci 2012; 13:9380-9399. [PMID: 22942771 PMCID: PMC3430302 DOI: 10.3390/ijms13079380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/12/2012] [Accepted: 07/19/2012] [Indexed: 01/28/2023] Open
Abstract
To bring insights into neurofibroma biochemistry, a comprehensive secretome analysis was performed on cultured human primary Schwann cells isolated from surgically resected plexiform neurofibroma and from normal nerve tissue. Using a combination of SDS-PAGE and high precision LC-MS/MS, 907 proteins were confidently identified in the conditioned media of Schwann cell cultures combined. Label free proteome profiling revealed consistent release of high levels of 22 proteins by the four biological replicates of NF1 Schwann cell cultures relative to the two normal Schwann cell cultures. Inversely, 9 proteins displayed decreased levels in the conditioned media of NF1 relative to normal Schwann cells. The proteins with increased levels included proteins involved in cell growth, angiogenesis and complement pathway while proteins with decreased levels included those involved in cell adhesion, plasminogen pathway and extracellular matrix remodeling. Retinoic acid receptor responder protein-1 (RARRES1), previously described as an integral membrane tumor suppressor, was found exclusively secreted by NF1 Schwann cells but not by normal Schwann cells. All-trans retinoic acid modulated secretion of RARRES1 in a dose dependent manner. This study shows altered secretion of key proteins in NF1 derived Schwann cells. The potential implication of these proteins in neurofibroma biology is discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yetrib Hathout
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-202-476-3136; Fax: +1-202-476-6014
| |
Collapse
|
43
|
Kloth M, Goering W, Ribarska T, Arsov C, Sorensen KD, Schulz WA. The SNP rs6441224 influences transcriptional activity and prognostically relevant hypermethylation of RARRES1 in prostate cancer. Int J Cancer 2012; 131:E897-904. [PMID: 22573467 DOI: 10.1002/ijc.27628] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/23/2012] [Indexed: 02/03/2023]
Abstract
Epigenetic aberrations are frequent in prostate cancer and could be useful for detection and prognostication. However, the underlying mechanisms and the sequence of these changes remain to be fully elucidated. The tumor suppressor gene RARRES1 (TIG1) is frequently hypermethylated in several cancers. Having noted changes in the expression of its paralogous neighbor gene LXN at 3q25.32, we used pyrosequencing to quantify DNA methylation at both genes and determine its relationship with clinicopathological parameters in 86 prostate cancer tissues from radical prostatectomies. Methylation at LXN and RARRES1 was highly correlated. Increasing methylation was associated with worse clinical features, including biochemical recurrence, and decreased expression of both genes. However, expression of three neighboring genes was unaffected. Intriguingly, RARRES1 methylation was influenced by the genotype of the rs6441224 single-nucleotide polymorphism (SNP) in its promoter. We found that this SNP is located within an ETS-family-response element and that the more strongly methylated allele confers lower activity in reporter assays. Concomitant methylation of RARRES1 and LXN in cancerous tissues was also detected in prostate cancer cell lines and was shown to be associated with repressive histone modifications and transcriptional downregulation. In conclusion, we found that genotype-associated hypermethylation of the ETS-family target gene RARRES1 influences methylation at its neighbor gene LXN and could be useful as a prognostic biomarker.
Collapse
Affiliation(s)
- Michael Kloth
- Department of Urology, Heinrich Heine University, Duesseldorf, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Otero A, Rodríguez de la Vega M, Tanco S, Lorenzo J, Avilés FX, Reverter D. The novel structure of a cytosolic M14 metallocarboxypeptidase (CCP) from Pseudomonas aeruginosa: a model for mammalian CCPs. FASEB J 2012; 26:3754-64. [PMID: 22645247 DOI: 10.1096/fj.12-209601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PaCCP is a metallocarboxypeptidase (MCP) of the M14 family from Pseudomonas aeruginosa, which belongs to a bacterial clade of carboxypeptidases that are homologous to the recently discovered M14D subfamily of human nonsecretory cytosolic carboxypeptidases (CCPs). CCPs are intracellular peptidases involved, among other roles, in the post-translational modifications of tubulin. Here we report the crystal structure of PaCCP at high resolution (1.6 Å). Its 375 residues are folded in a novel β-sandwich N-terminal domain followed by the classical carboxypeptidase α/β-hydrolase domain, this one in a shorter and more compact form. The former is unique in the whole family and does not have sequential or structural homology with other domains that are usually flanking the latter, like the prodomain of the M14A subfamily or the C-terminal transthyretin/prealbumin-like domains of the M14B subfamily. PaCCP does not display activity against small carboxypeptidase substrates, so in this form it might constitute an inactive precursor of the protease. Structural results derived from cocrystallization with well-known inhibitors of MCPs indicate that the enzyme might only possess C-terminal hydrolase activity against cellular substrates of particular specificity and/or when undergoes structural rearrangements. The derived PaCCP structure allows a first structural insight into the more complex and largely unknown mammalian CCP subfamily.
Collapse
Affiliation(s)
- Anabel Otero
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i de Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
45
|
Sahab ZJ, Kirilyuk A, Zhang L, Khamis ZI, Pompach P, Sung Y, Byers SW. Analysis of tubulin alpha-1A/1B C-terminal tail post-translational poly-glutamylation reveals novel modification sites. J Proteome Res 2012; 11:1913-23. [PMID: 22296162 PMCID: PMC3292626 DOI: 10.1021/pr2011044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tubulin-α(1A/1B) C-terminal tail (CTT) has seven glutamic acid residues among the last 11 amino acids of its sequence that are potential sites for glutamylation. Cleavage of C-terminal tyrosine resulting in the detyrosinated form of tubulin-α(1A/1B) is another major modification. These modifications among others bring about highly heterogeneous tubulin samples in brain cells and microtubules, play a major role in directing intracellular trafficking, microtubule dynamics, and mitotic events, and can vary depending on the cell and disease state, such as cancer and neurodegenerative disorders. Identified previously using primary mass spectrometry (MS) ions and partial Edman sequencing, tubulin-α(1A/1B) glutamylation was found exclusively on the E(445) residue. We here describe the analysis of tubulin-α(1A/1B) glutamylation and detyrosination after 2-DE separation, trypsin and proteinase K in-gel digestion, and nanoUPLC-ESI-QqTOF-MS/MS of mouse brain and bovine microtubules. Tyrosinated, detyrosinated, and Δ2-tubulin-α(1A/1B) CTTs were identified on the basis of a comparison of fragmentation patterns and retention times between endogenous and synthetic peptides. Stringent acceptance criteria were adapted for the identification of novel glutamylation sites. In addition to the previously identified site at E(445), glutamylation on mouse and bovine tubulin-α(1A/1B) CTTs was identified on E(441) and E(443) with MASCOT Expect values below 0.01. O-Methylation of glutamates was also observed.
Collapse
Affiliation(s)
- Ziad J Sahab
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University , Washington, DC 20007, United States.
| | | | | | | | | | | | | |
Collapse
|
46
|
Sang QXA, Man YG, Sung YM, Khamis ZI, Zhang L, Lee MH, Byers SW, Sahab ZJ. Non-receptor tyrosine kinase 2 reaches its lowest expression levels in human breast cancer during regional nodal metastasis. Clin Exp Metastasis 2012; 29:143-53. [PMID: 22116632 PMCID: PMC3449303 DOI: 10.1007/s10585-011-9437-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 11/12/2011] [Indexed: 01/19/2023]
Abstract
Almost half of breast Ductal Carcinoma in situ are likely to remain non threatening in situ lesions with no invasion to the surrounding stroma and no metastases. The majority of focal disruptions in myoepithelial (ME) cell layers indicative of invasion onset were found to be overlying epithelial cell clusters with no or substantially reduced estrogen receptor α (ERα) expression. Here we report the down-regulation of tyrosine kinase-2 (TYK2) and up-regulation of strumpellin expression, among other proteins in ERα(-) cells located at disrupted ME layers compared to adjacent ERα(+) cells overlying an intact myoepithelial layer. ERα(+) and ERα(-) cells were microdissected from the same in vivo human breast cancer tissues, proteins were extracted and separated utilizing Differential in-Gel Electrophoresis followed by trypsin digestion, MALDI-TOF analysis, and protein identification. Proteins expressed by ERα(-) cell clusters were found to express higher levels of strumpellin that binds to valosin-containing protein (VCP) to slow-down wound closure and promote growth; and lower levels of TYK2, a jak protein necessary for lineage specific differentiation. TYK2 levels were further analyzed by immunohistochemistry in a cohort composed of 70 patients with broad clinical characteristics. TYK2 levels were minimal in TxN1M0 breast cancers which is the stage where the initial regional lymph node metastasis is observed. Our data highlight the role of TYK2 downregulation in breast cancer cell de-differentitation and initiation of regional metastasis. In addition, the aggressiveness of the ERα(-) cell clusters compared to ERα(+) ones present in the same duct of the same patient was confirmed.
Collapse
Affiliation(s)
- Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4390, USA
| | - Yan-Gao Man
- The Diagnostic and Translational Research Center, the Henry Jackson Foundation for the Advancement of Military Medicine, Walter Reed Army Medical Center, Washington DC 20307
- Jilin University, Changchun, Jilin, China
| | - You Me Sung
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Zahraa I. Khamis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Lihua Zhang
- Proteomics and Metabolomics Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007
| | - Mi-Hye Lee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Stephen W. Byers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Ziad J. Sahab
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
47
|
Tsai FM, Wu CC, Shyu RY, Wang CH, Jiang SY. Tazarotene-induced gene 1 inhibits prostaglandin E2-stimulated HCT116 colon cancer cell growth. J Biomed Sci 2011; 18:88. [PMID: 22126303 PMCID: PMC3247857 DOI: 10.1186/1423-0127-18-88] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 11/30/2011] [Indexed: 01/21/2023] Open
Abstract
Background The tazarotene-induced gene 1 (TIG1) is a putative tumor suppressor gene. We have recently demonstrated both TIG1A and TIG1B isoforms inhibited cell growth and induced the expression of G protein-coupled receptor kinase 5 (GRK5) in colon cancer cells. Because elevated prostaglandin E2 (PGE2) signaling plays a significant role in colorectal carcinogenesis, the objective of this study was to explore the effect of TIG1 on PGE2-induced cellular proliferation and signaling in colon cancer cells. Methods HCT116 cells as well as TIG1A and TIG1B stable cells established from HCT116 colon cancer cells using the GeneSwitch system were used. TIG1 isoform expression was induced by mifepristone treatment in stable cells. Cell growth was determined using the WST-1 cell proliferation assay. Activation of β-catenin/TCF and cyclic adenosine monophosphate (cAMP)/CREB signaling pathways were determined using luciferase reporter assays. Expression and subcellular distribution of β-catenin were analyzed using Western blot and confocal microscope. Levels of cAMP were measured using an enzyme immunoassay. RNA interference was used to examine the effects of TIG1- and GRK5-mediated changes. Results PGE2-stimulated cell growth was reduced in inducible TIG1A- and TIG1B-stable HCT116 cells. GRK5 expression was upregulated by both TIG1A and TIG1B isoforms, and its expression suppressed PGE2-stimulated HCT116 cell growth. GRK5, TIG1A, and TIG1B expression significantly inhibited PGE2-stimulated β-catenin/TCF and cAMP signaling pathway reporters and cAMP. Also, PGE2-stimulated nuclear localization of β-catenin was inhibited by expression of TIG1A and TIG1B, which was ameliorated by both TIG1 and GRK5 siRNAs. Conclusions TIG1 suppressed PGE2-stimulated Wnt and cAMP signaling pathways in colon cancer cells through GRK5.
Collapse
Affiliation(s)
- Fu-Ming Tsai
- Department of Research, Buddhist Tzu Chi General Hospital Taipei Branch, 289 Jianguo Rd, Sindian District, New Taipei City, 231 Taiwan
| | | | | | | | | |
Collapse
|
48
|
Novel stromal biomarkers in human breast cancer tissues provide evidence for the more malignant phenotype of estrogen receptor-negative tumors. J Biomed Biotechnol 2011; 2011:723650. [PMID: 21976967 PMCID: PMC3185288 DOI: 10.1155/2011/723650] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 07/19/2011] [Indexed: 01/25/2023] Open
Abstract
Research efforts were focused on genetic alterations in epithelial cancer cells. Epithelial-stromal interactions play a crucial role in cancer initiation, progression, invasion, angiogenesis, and metastasis; however, the active role of stroma in human breast tumorigenesis in relation to estrogen receptor (ER) status of epithelial cells has not been explored. Using proteomics and biochemical approaches, we identified two stromal proteins in ER-positive and ER-negative human breast cancer tissues that may affect malignant transformation in breast cancer. Two putative biomarkers, T-cell receptor alpha (TCR-α) and zinc finger and BRCA1-interacting protein with a KRAB domain (ZBRK1), were detected in leukocytes of ER-positive and endothelial cells of ER-negative tissues, respectively. Our data suggest an immunosuppressive role of leukocytes in invasive breast tumors, propose a multifunctional nature of ZBRK1 in estrogen receptor regulation and angiogenesis, and demonstrate the aggressiveness of ER-negative human breast carcinomas. This research project may identify new stromal drug targets for the treatment of breast cancer patients.
Collapse
|
49
|
Sahab ZJ, Man YG, Byers SW, Sang QXA. Putative biomarkers and targets of estrogen receptor negative human breast cancer. Int J Mol Sci 2011; 12:4504-21. [PMID: 21845093 PMCID: PMC3155366 DOI: 10.3390/ijms12074504] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/27/2011] [Accepted: 07/04/2011] [Indexed: 11/26/2022] Open
Abstract
Breast cancer is a progressive and potentially fatal disease that affects women of all ages. Like all progressive diseases, early and reliable diagnosis is the key for successful treatment and annihilation. Biomarkers serve as indicators of pathological, physiological, or pharmacological processes. Her2/neu, CA15.3, estrogen receptor (ER), progesterone receptor (PR), and cytokeratins are biomarkers that have been approved by the Food and Drug Administration for disease diagnosis, prognosis, and therapy selection. The structural and functional complexity of protein biomarkers and the heterogeneity of the breast cancer pathology present challenges to the scientific community. Here we review estrogen receptor-related putative breast cancer biomarkers, including those of putative breast cancer stem cells, a minor population of estrogen receptor negative tumor cells that retain the stem cell property of self-renewal. We also review a few promising cytoskeleton targets for ER alpha negative breast cancer.
Collapse
Affiliation(s)
- Ziad J. Sahab
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA; E-Mail:
| | - Yan-Gao Man
- Diagnostic and Translational Research Center, Henry Jackson Foundation for the Advancement of Military Medicine, Gaithersburg, MD 20789, USA; E-Mail:
- Jilin University, Changchun 130012, China
| | - Stephen W. Byers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA; E-Mail:
| | - Qing-Xiang A. Sang
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, 102 Varsity Way, Tallahassee, FL 32306, USA; E-Mail:
| |
Collapse
|