1
|
Vicari HP, Gomes RDC, Lima K, Rossini NDO, Rodrigues Junior MT, de Miranda LBL, Dias MVB, Costa-Lotufo LV, Coelho F, Machado-Neto JA. Cyclopenta[b]indoles as novel antimicrotubule agents with antileukemia activity. Toxicol In Vitro 2024; 99:105856. [PMID: 38821378 DOI: 10.1016/j.tiv.2024.105856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Acute leukemias present therapeutic challenges despite advances in treatments. Microtubule inhibitors have played a pivotal role in cancer therapy, inspiring exploration into novel compounds like C2E1 from the cyclopenta[b]indole class. In the present study, we investigated C2E1's potential as a therapeutic agent for acute leukemia at molecular, cellular, and genetic levels. C2E1 demonstrated tubulin depolarization activity, significantly reducing leukemia cell viability. Its impact involved multifaceted mechanisms: inducing apoptosis, arrest of cell cycle progression, and inhibition of clonogenicity and migration in leukemia cells. At a molecular level, C2E1 triggered DNA damage, antiproliferative, and apoptosis markers and altered gene expression related to cytoskeletal regulation, disrupting essential cellular processes crucial for leukemia cell survival and proliferation. These findings highlight C2E1's promise as a potential candidate for novel anti-cancer therapies. Notably, its distinct mode of action from conventional microtubule-targeting drugs suggests the potential to bypass common resistance mechanisms encountered with existing treatments. In summary, C2E1 emerges as a compelling compound with diverse effects on leukemia cells, showcasing promising antineoplastic properties. Its ability to disrupt critical cellular functions selective to leukemia cells positions it as a candidate for future therapeutic development.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ralph da Costa Gomes
- Department of Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas, São Paulo, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernando Coelho
- Department of Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas, São Paulo, Brazil.
| | | |
Collapse
|
2
|
Zhang Y, Wei S, Zhang Q, Zhang Y, Sun C. Paris saponin VII inhibits triple-negative breast cancer by targeting the MEK/ERK/STMN1 signaling axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155746. [PMID: 38763012 DOI: 10.1016/j.phymed.2024.155746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a category of breast cancer characterized with high molecular heterogeneity. Owing to the lack of effective therapeutic strategies, patients with TNBC have a poor prognosis. Paris saponin VII (PSⅦ), a steroidal saponin extracted from the rhizome of Trichillium tschonoskii Maxim, exhibits excellent anti-cancer activity in a variety of solid tumors. However, the role and potential mechanism of PSⅦ against TNBC remain unexplored. PURPOSE This study aimed to elucidate the therapeutic effects of PSⅦ against TNBC and explore the potential mechanism of action. METHODS We combined the analysis of public single-cell sequencing data with weighted gene co-expression network analysis (WGCNA) to identity differentially expressed genes (DEGs) that distinguished malignant and normal epithelial cells in TNBC. Subsequently, the biological features of DEGs in TNBC were evaluated. Gene set enrichment analysis (GSEA) was used to define potential pathways associated with the DEGs. The pharmacological activity of PSⅦ for TNBC was evidenced via in vitro and in vivo experiments, and molecular docking, molecular dynamics (MD), surface plasmon resonance (SPR) assay and western blotting were employed to confirm the relative mechanisms. RESULTS Single-cell sequencing and WGCNA revealed STMN1 as a pivotal biomarker of TNBC. STMN1 overexpression in TNBC was associated with poor patient prognosis. GSEA revealed a significant accumulation of STMN1 within the MAPK signaling pathway. Furthermore, In vitro experiments showed that PSⅦ showed significantly suppressive actions on the proliferation, migration and invasion abilities for TNBC cells, while inducing apoptosis. Molecular docking, MD analysis and SPR assay indicated a robust interaction between PSⅦ and the MEK protein. Western blotting revealed that PSⅦ may inhibit tumor progression by suppressing the phosphorylation of MEK1/2 and the downstream phosphorylation of ERK1/2 and STMN1. Intraperitoneal injection of PSⅦ (10 mg/kg) notably reduced tumor growth by 71.26 % in a 4T1 xenograft model. CONCLUSION In our study, the systems biology method was used to identify potential therapeutic targets for TNBC. In vitro and in vivo experiments demonstrated PSⅦ suppresses cancer progression by targeting the MEK/ERK/STMN1 signaling axis. For the first time, the inhibition of STMN1 phosphorylation has been indicated as a possible mechanism for the anticancer effects of PSⅦ. These results emphasize the potential value of PSⅦ as a promising anti-cancer drug candidate for further development in the field of TNBC therapeutics.
Collapse
Affiliation(s)
- Yubao Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Shijie Wei
- Department of Oncology, The Affiliated Qingdao Hiser hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao 266071, China
| | - Qinxiang Zhang
- Institute of Integrated Medicine, Qingdao University, Qingdao 266071, China
| | - Yue Zhang
- Institute of Integrated Medicine, Qingdao University, Qingdao 266071, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, No. 7166, Baotong West Street, Weicheng District, Weifang, Shandong Province 261000, China.
| |
Collapse
|
3
|
Aksoy A, Varoglu A, Onalan EE, Tektemur A, Artas G, Koc M, Cakmak M, Aydin S, Kilic M, Ulas M. The knockdown of stathmin with si-RNA inhibits invasion of mesothelioma. Tissue Cell 2024; 87:102303. [PMID: 38244401 DOI: 10.1016/j.tice.2024.102303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND To investigate the mechanism of action of stathmin1 (STMN1) in mesothelioma (MSM) and whether it has any role in its treatment. METHODS STMN1 expression was examined using immunohistochemistry in biopsy tissues taken from MSM patients. The relationships between the levels of STMN1 expression in the pathology preparations of MSM patients, and the clinicopathological characteristics of these patients, and their survival times were investigated. Transfection of STMN1-specific siRNA into SPC212 cells was compared to negative control siRNAs. The mRNA levels of genes that may play a role in invasion, apoptosis, and autophagy were evaluated by RT-PCR. RESULTS The expression of STMN1 was shown to be high in MSM tissues (p < 0.05). It was found that the only independent predictor factor affecting the survival time of MSM patients was the disease stage (p < 0.05). STMN1 was significantly reduced after siRNA intervention (81.5%). STMN1 with specific siRNA has been shown to suppress invasion by reducing the mRNA levels of cadherin-6 (CDH6), fibroblast growth factor-8 (FGF8), hypoxia-inducible factor 1 (HIF1A), matrix metallopeptidase 1-2 (gelatinase A) (MMP1-2), and TIMP metallopeptidase inhibitor 2 (TIMP2), which are important markers for invasion. Although the expression of apoptosis and autophagy-related genes, caspase-2 (Casp2) and LC-3, was reduced by silencing STMN1 with specific siRNA in western blot analysis, this effect was not observed in PCR results. CONCLUSIONS Immunohistochemical analysis of STMN1 may contribute to the differential diagnosis of MSM, and STMN1 may also be considered as a potential therapeutic target in the early invasive stage of MSM therapy.
Collapse
Affiliation(s)
- Asude Aksoy
- Department of Medical Oncology, University of Health Sciences, Fethi Sekin City Hospital SUAM, Elazig, Turkey.
| | - Asuman Varoglu
- Department of Neurology, Medical Faculty, Medeniyet University, Istanbul, Turkey
| | - Ebru Etem Onalan
- Department of Medical Biology and Genetics, Firat University, Elazig, Turkey
| | - Ahmet Tektemur
- Department of Medical Biology and Genetics, Firat University, Elazig, Turkey
| | - Gokhan Artas
- Department of Pathology, Medical Faculty, Firat University, Elazig, Turkey
| | - Mustafa Koc
- Department of Radiology, Medical Faculty, Firat University, Elazig, Turkey
| | - Muharrem Cakmak
- Department of Thoracic Surgery, Medical Faculty, Firat University, Elazig, Turkey
| | - Siyami Aydin
- Department of Thoracic Surgery, Medical Faculty, Firat University, Elazig, Turkey
| | - Murat Kilic
- Department of Thoracic Surgery, Inonu University, Malatya, Turkey
| | - Mustafa Ulas
- Department of Physiology, Medical Faculty, Firat University, Elazig, Turkey
| |
Collapse
|
4
|
Liu R, Liang X, Guo H, Li S, Yao W, Dong C, Wu J, Lu Y, Tang J, Zhang H. STNM1 in human cancers: role, function and potential therapy sensitizer. Cell Signal 2023:110775. [PMID: 37331415 DOI: 10.1016/j.cellsig.2023.110775] [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: 04/19/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
STMN1 belongs to the stathmin gene family, it encodes a cytoplasmic phosphorylated protein, stathmin1, which is commonly observed in vertebrate cells. STMN1 is a structural microtubule-associated protein (MAP) that binds to microtubule protein dimers rather than microtubules, with each STMN1 binding two microtubule protein dimers and preventing their aggregation, leading to microtubule instability. STMN1 expression is elevated in a number of malignancies, and inhibition of its expression can interfere with tumor cell division. Its expression can change the division of tumor cells, thereby arresting cell growth in the G2/M phase. Moreover, STMN1 expression affects tumor cell sensitivity to anti-microtubule drug analogs, including vincristine and paclitaxel. The research on MAPs is limited, and new insights on the mechanism of STMN1 in different cancers are emerging. The effective application of STMN1 in cancer prognosis and treatment requires further understanding of this protein. Here, we summarize the general characteristics of STMN1 and outline how STMN1 plays a role in cancer development, targeting multiple signaling networks and acting as a downstream target for multiple microRNAs, circRNAs, and lincRNAs. We also summarize recent findings on the function role of STMN1 in tumor resistance and as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Ruiqi Liu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China; Graduate Department, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaodong Liang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China; Graduate Department, Bengbu Medical College, Bengbu, Anhui, China
| | - Haiwei Guo
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Shuang Li
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weiping Yao
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China; Graduate Department, Bengbu Medical College, Bengbu, Anhui, China
| | - Chenfang Dong
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajun Wu
- Graduate Department, Bengbu Medical College, Bengbu, Anhui, China; Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Yanwei Lu
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jianming Tang
- Department of Radiation Oncology, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Haibo Zhang
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Vicari HP, Lima K, Costa-Lotufo LV, Machado-Neto JA. Cellular and Molecular Effects of Eribulin in Preclinical Models of Hematologic Neoplasms. Cancers (Basel) 2022; 14:cancers14246080. [PMID: 36551566 PMCID: PMC9776580 DOI: 10.3390/cancers14246080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Despite the advances in understanding the biology of hematologic neoplasms which has resulted in the approval of new drugs, the therapeutic options are still scarce for relapsed/refractory patients. Eribulin is a unique microtubule inhibitor that is currently being used in the therapy for metastatic breast cancer and soft tissue tumors. Here, we uncover eribulin's cellular and molecular effects in a molecularly heterogeneous panel of hematologic neoplasms. Eribulin reduced cell viability and clonogenicity and promoted apoptosis and cell cycle arrest. The minimal effects of eribulin observed in the normal leukocytes suggested selectivity for malignant blood cells. In the molecular scenario, eribulin induces DNA damage and apoptosis markers. The ABCB1, ABCC1, p-AKT, p-NFκB, and NFκB levels were associated with responsiveness to eribulin in blood cancer cells, and a resistance eribulin-related target score was constructed. Combining eribulin with elacridar (a P-glycoprotein inhibitor), but not with PDTC (an NFkB inhibitor), increases eribulin-induced apoptosis in leukemia cells. In conclusion, our data indicate that eribulin leads to mitotic catastrophe and cell death in blood cancer cells. The expression and activation of MDR1, PI3K/AKT, and the NFκB-related targets may be biomarkers of the eribulin response, and the combined treatment of eribulin and elacridar may overcome drug resistance in these diseases.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo 01246-903, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulos 05508-000, Brazil
- Correspondence: ; Tel.: +55-11-3091-7467
| |
Collapse
|
6
|
Ren D, Sun Y, Li D, Wu H, Jin X. USP22-mediated deubiquitination of PTEN inhibits pancreatic cancer progression by inducing p21 expression. Mol Oncol 2022; 16:1200-1217. [PMID: 34743406 PMCID: PMC8895442 DOI: 10.1002/1878-0261.13137] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/25/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome ten (PTEN) is a dual lipid and protein phosphatase. Multiple mechanisms contributing to the regulation of PTEN levels have been identified thus far, including post-translational modifications, epigenetic mechanisms, and transcriptional mechanisms. In the present study, we identified ubiquitin-specific peptidase 22 (USP22) as a novel deubiquitination-modifying enzyme of PTEN. Furthermore, by inducing deubiquitination and inhibiting the degradation of PTEN, USP22 could induce cyclin-dependent kinase inhibitor 1A (CDKN1A, also symboled as p21) expression in pancreatic cancer. Besides, MDM2 proto-oncogene (MDM2) inhibitor enhanced the antipancreatic cancer effects of USP22 overexpression. In addition to its regulation of MDM2-tumor protein p53 (p53) signaling, we found that PTEN could induce p21 expression by interacting with ankyrin repeat and KH domain containing 1 (ANKHD1) and inhibiting ANKHD1 binding to the p21 promoter. Taken together, our results indicate that ANKHD1 and MDM2 might be novel therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Dianyun Ren
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic CancerUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yan Sun
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic CancerUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan Li
- Cardiovascular Medicine DepartmentUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Heshui Wu
- Department of Pancreatic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic CancerUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xin Jin
- Department of UrologyThe Second Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
7
|
Vicari HP, Coelho-Silva JL, Pereira-Martins DA, Lucena-Araujo AR, Lima K, Lipreri da Silva JC, Scheucher PS, Koury LC, de Melo RA, Bittencourt R, Pagnano K, Nunes E, Fagundes EM, Kerbauy F, de Figueiredo-Pontes LL, Costa-Lotufo LV, Rego EM, Traina F, Machado-Neto JA. STMN1 is highly expressed and contributes to clonogenicity in acute promyelocytic leukemia cells. Invest New Drugs 2021; 40:438-452. [PMID: 34837603 DOI: 10.1007/s10637-021-01197-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022]
Abstract
Stathmin 1 (STMN1) is a microtubule-destabilizing protein highly expressed in hematological malignancies and involved in proliferation and differentiation. Although a previous study found that the PML-RARα fusion protein, which contributes to the pathophysiology of acute promyelocytic leukemia (APL), positively regulates STMN1 at the transcription and protein activity levels, little is known about the role of STMN1 in APL. In this study, we aimed to investigate the STMN1 expression levels and their associations with laboratory, clinical, and genomic data in APL patients. We also assessed the dynamics of STMN1 expression during myeloid cell differentiation and cell cycle progression, and the cellular effects of STMN1 silencing and pharmacological effects of microtubule-stabilizing drugs on APL cells. We found that STMN1 transcripts were significantly increased in samples from APL patients compared with those of healthy donors (all p < 0.05). However, this had no effect on clinical outcomes. STMN1 expression was associated with proliferation- and metabolism-related gene signatures in APL. Our data confirmed that STMN1 was highly expressed in early hematopoietic progenitors and reduced during cell differentiation, including the ATRA-induced granulocytic differentiation model. STMN1 phosphorylation was predominant in a pool of mitosis-enriched APL cells. In NB4 and NB4-R2 cells, STMN1 knockdown decreased autonomous cell growth (all p < 0.05) but did not impact ATRA-induced apoptosis and differentiation. Finally, treatment with paclitaxel (as a single agent or combined with ATRA) induced microtubule stabilization, resulting in mitotic catastrophe with repercussions for cell viability, even in ATRA-resistant APL cells. This study provides new insights into the STMN1 functions and microtubule dynamics in APL.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Juan Luiz Coelho-Silva
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Diego A Pereira-Martins
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Keli Lima
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Priscila Santos Scheucher
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Luisa C Koury
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Raul A de Melo
- Department of Internal Medicine, University of Pernambuco and Fundação HEMOPE, Recife, Brazil
| | - Rosane Bittencourt
- Hematology Division, University Hospital of the Federal University of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Katia Pagnano
- University of Campinas/Hemocentro-Unicamp, Campinas, Brazil
| | - Elenaide Nunes
- Hematology Division, Federal University of Paraná, Curitiba, Brazil
| | - Evandro M Fagundes
- Hematology Division, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabio Kerbauy
- Hematology Division, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Lorena Lobo de Figueiredo-Pontes
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Hematology, Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Fabiola Traina
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Brazil.
| | | |
Collapse
|
8
|
Liao TT, Cheng WC, Yang CY, Chen YQ, Su SH, Yeh TY, Lan HY, Lee CC, Lin HH, Lin CC, Lu RH, Chiou AET, Jiang JK, Hwang WL. The microRNA-210-Stathmin1 Axis Decreases Cell Stiffness to Facilitate the Invasiveness of Colorectal Cancer Stem Cells. Cancers (Basel) 2021; 13:cancers13081833. [PMID: 33921319 PMCID: PMC8069838 DOI: 10.3390/cancers13081833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Metastasis of tumor cells is the leading cause of death in cancer patients. Concurrent therapy with surgical removal of primary and metastatic lesions is the main approach for cancer therapy. Currently, therapeutic resistant properties of cancer stem cells (CSCs) are known to drive malignant cancer progression, including metastasis. Our study aimed to identify molecular tools dedicated to the detection and treatment of CSCs. We confirmed that microRNA-210-3p (miR-210) was upregulated in colorectal stem-like cancer cells, which targeted stathmin1 (STMN1), to decrease cell elasticity for increasing mobility. We envision that strategies for softening cellular elasticity will reduce the onset of CSC-orientated metastasis. Abstract Cell migration is critical for regional dissemination and distal metastasis of cancer cells, which remain the major causes of poor prognosis and death in patients with colorectal cancer (CRC). Although cytoskeletal dynamics and cellular deformability contribute to the migration of cancer cells and metastasis, the mechanisms governing the migratory ability of cancer stem cells (CSCs), a nongenetic source of tumor heterogeneity, are unclear. Here, we expanded colorectal CSCs (CRCSCs) as colonospheres and showed that CRCSCs exhibited higher cell motility in transwell migration assays and 3D invasion assays and greater deformability in particle tracking microrheology than did their parental CRC cells. Mechanistically, in CRCSCs, microRNA-210-3p (miR-210) targeted stathmin1 (STMN1), which is known for inducing microtubule destabilization, to decrease cell elasticity in order to facilitate cell motility without affecting the epithelial–mesenchymal transition (EMT) status. Clinically, the miR-210-STMN1 axis was activated in CRC patients with liver metastasis and correlated with a worse clinical outcome. This study elucidates a miRNA-oriented mechanism regulating the deformability of CRCSCs beyond the EMT process.
Collapse
Affiliation(s)
- Tsai-Tsen Liao
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.-T.L.); (H.-Y.L.)
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
| | - Wei-Chung Cheng
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, China Medical University, Taichung 406, Taiwan;
- Research Center for Cancer Biology, China Medical University, Taichung 406, Taiwan
| | - Chih-Yung Yang
- Department of Education and Research, Taipei City Hospital, Taipei 106, Taiwan;
- General Education Center, University of Taipei, Taipei 100, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Shu-Han Su
- Institution of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan; (S.-H.S.); (T.-Y.Y.)
| | - Tzu-Yu Yeh
- Institution of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan; (S.-H.S.); (T.-Y.Y.)
| | - Hsin-Yi Lan
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (T.-T.L.); (H.-Y.L.)
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Chih-Chan Lee
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Hung-Hsin Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan;
| | - Chun-Chi Lin
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ruey-Hwa Lu
- Department of Surgery, Zhongxing Branch, Taipei City Hospital, Taipei 106, Taiwan;
| | - Arthur Er-Terg Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Jeng-Kai Jiang
- Division of Colon & Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: (J.-K.J.); (W.-L.H.); Tel.: +886-2-2826-7000 (ext. 65832) (W.-L.H.)
| | - Wei-Lun Hwang
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Correspondence: (J.-K.J.); (W.-L.H.); Tel.: +886-2-2826-7000 (ext. 65832) (W.-L.H.)
| |
Collapse
|
9
|
Carlos JAEG, Lima K, Costa-Lotufo LV, Leitão A, Machado-Neto JA. AD80, a multikinase inhibitor, exhibits antineoplastic effects in acute leukemia cellular models targeting the PI3K/STMN1 axis. Invest New Drugs 2021; 39:1139-1149. [PMID: 33475938 DOI: 10.1007/s10637-021-01066-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
Despite the great advances in the understanding of the molecular basis of acute leukemia, very little of this knowledge has been translated into new therapies. Stathmin 1 (STMN1), a phosphoprotein that regulates microtubules dynamics, is highly expressed in acute leukemia cells and promotes cell cycle progression and proliferation. GDP366 has been described as a STMN1 and survivin inhibitor in solid tumors. This study identified structural GDP366 analogs and the cellular and molecular mechanisms underlying their suppressive effects on acute leukemia cellular models. STMN1 mRNA levels were higher in AML and ALL patients, independent of risk stratification (all p < 0.001). Cheminformatics analysis identified three structural GDP366 analogs, with AD80 more potent and effective than GSK2606414 and GW768505A. In acute leukemia cells, GDP366 and AD80 reduced cell viability and autonomous clonal growth in a dose- and/or time-dependent manner (p < 0.05) and induced apoptosis and cell cycle arrest (p < 0.05). At the molecular level, GDP366 and AD80 reduced Ki-67 (a proliferation marker) expression and S6 ribosomal protein (a PI3K/AKT/mTOR effector) phosphorylation, and induced PARP1 (an apoptosis marker) cleavage and γH2AX (a DNA damage marker) expression. GDP366 induced STMN1 phosphorylation and survivin expression, while AD80 reduced survivin and STMN1 expression. GDP366 and AD80 modulated 18 of the 84 cytoskeleton regulators-related genes. These results indicated that GDP366 and AD80 reduced the PI3K/STMN1 axis and had cytotoxic effects in acute leukemia cellular models. Our findings further highlight STMN1-mediated signaling as a putative anticancer target for acute leukemia.
Collapse
Affiliation(s)
- Jorge Antonio Elias Godoy Carlos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, CEP 05508-900, São Paulo, SP, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, CEP 05508-900, São Paulo, SP, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, CEP 05508-900, São Paulo, SP, Brazil
| | - Andrei Leitão
- Medicinal & Biological Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, São Paulo, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, CEP 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Vicari HP, Lima K, Gomes RDC, Fernandes DC, da Silva JCL, Rodrigues Junior MT, Barroso de Oliveira AS, Dos Santos RN, Andricopulo AD, Coelho F, Costa-Lotufo LV, Machado-Neto JA. Synthetic cyclopenta[b]indoles exhibit antineoplastic activity by targeting microtubule dynamics in acute myeloid leukemia cells. Eur J Pharmacol 2021; 894:173853. [PMID: 33422507 DOI: 10.1016/j.ejphar.2021.173853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Acute promyelocytic leukemia (APL) is associated with PML-RARα oncogene, which is treated using all-trans retinoic acid (ATRA)-based chemotherapy. However, chemoresistance is observed in 20-30% of treated patients and represents a clinical challenge, raising the importance of the development of new therapeutic options. In the present study, the effects of three synthetic cyclopenta[b]indoles on the leukemia phenotype were investigated using NB4 (ATRA-sensitive) and NB4-R2 (ATRA-resistant) cells. Among the tested synthetic cyclopenta[b]indoles, compound 2, which contains a heterocyclic nucleus, was the most active, presenting time-dependent cytotoxic activity in the μM range in APL cells, without cytotoxicity for normal leukocytes, and was selected for further characterization. Compound 2 significantly decreased clonogenicity, increased apoptosis, and caused cell cycle arrest at S and G2/M phases in a drug concentration-dependent manner. Morphological analyses indicated aberrant mitosis and diffuse tubulin staining upon compound 2 exposure, which corroborates cell cycle findings. In the molecular scenario, compound 2 reduced STMN1 expression and activity, and induced PARP1 cleavage and H2AX and CHK2 phosphorylation, and modulated CDKN1A, PMAIP1, GADD45A, and XRCC3 expressions, indicating reduction of cell proliferation, apoptosis, and DNA damage. Moreover, in the in vivo tubulin polymerization assay, NB4 and NB4-R2 cells showed a reduction in the levels of polymerized tubulin upon compound 2 exposure, which indicates tubulin as a target of the drug. Molecular docking supports this hypothesis. Taken together, these data indicated that compound 2 exhibits antileukemic effects through disrupting the microtubule dynamics, identifying a possible novel potential antineoplastic agent for the treatment of ATRA-resistant APL.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Ralph da Costa Gomes
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, Campinas, São Paulo, SP, 13083-970, Brazil
| | - Daniara Cristina Fernandes
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, Campinas, São Paulo, SP, 13083-970, Brazil; Currently at Instituto Federal de Educação Ciência e Tecnologia de São Paulo, Matão, SP, 15991-502, Brazil
| | - Jean Carlos Lipreri da Silva
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | | | | | | | | | - Fernando Coelho
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, Campinas, São Paulo, SP, 13083-970, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
11
|
Ramlogan-Steel CA, Steel JC, Fathallah H, Iancu-Rubin C, Atweh GF. Stathmin 1 deficiency induces erythro-megakaryocytic defects leading to macrocytic anemia and thrombocythemia in Stathmin 1 knock out mice. Blood Cells Mol Dis 2020; 87:102522. [PMID: 33260083 DOI: 10.1016/j.bcmd.2020.102522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 10/22/2022]
Abstract
Stathmin 1 (STMN1) is a cytosolic phosphoprotein that was discovered as a result of its high level of expression in leukemic cells. It plays an important role in the regulation of mitosis by promoting depolymerization of the microtubules that make up the mitotic spindle and, aging has been shown to impair STMN1 levels and change microtubule stability. We have previously demonstrated that a high level of STMN1 expression during early megakaryopoiesis is necessary for proliferation of megakaryocyte progenitors and that down-regulation of STMN1 expression during late megakaryopoiesis is important for megakaryocyte maturation and platelet production. In this report, we examined the effects of STMN1 deficiency on erythroid and megakaryocytic lineages in the mouse. Our studies show that STMN1 deficiency results in mild thrombocytopenia in young animals which converts into profound thrombocythemia as the mice age. STMN1 deficiency also lead to macrocytic changes in both erythrocytes and megakaryocytes that persisted throughout the life of STMN1 knock-out mice. Furthermore, STMN1 knock-out mice displayed a lower number of erythroid and megakaryocytic progenitor cells and had delayed recovery of their blood counts after chemotherapy. These studies show an important role for STMN1 in normal erythro-megakaryopoietic development and suggests potential implications for disorders affecting these hematopoietic lineages.
Collapse
Affiliation(s)
- Charmaine A Ramlogan-Steel
- School of Health, Medical and Applied Science, Central Queensland University, Rockhampton, QLD, Australia.
| | - Jason C Steel
- School of Health, Medical and Applied Science, Central Queensland University, Rockhampton, QLD, Australia
| | - Hassana Fathallah
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Camelia Iancu-Rubin
- Department of Pathology, Molecular and Cell-Based Medicine, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George F Atweh
- Division of Hematology/Oncology, University of New Mexico Cancer Center, Albuquerque, NM, USA.
| |
Collapse
|
12
|
Almeida BOD, Machado-Neto JA. Emerging functions for ANKHD1 in cancer-related signaling pathways and cellular processes. BMB Rep 2020. [PMID: 32635985 PMCID: PMC7473474 DOI: 10.5483/bmbrep.2020.53.8.087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
ANKHD1 (ankyrin repeat and KH domain containing 1) is a large protein characterized by the presence of multiple ankyrin repeats and a K-homology domain. Ankyrin repeat domains consist of widely existing protein motifs in nature, they mediate protein-protein interactions and regulate fundamental biological processes, while the KH domain binds to RNA or ssDNA and is associated with transcriptional and translational regulation. In recent years, studies containing relevant information on ANKHD1 in cancer biology and its clinical relevance, as well as the increasing complexity of signaling networks in which this protein acts, have been reported. Among the signaling pathways of interest in oncology regulated by ANKHD1 are Hippo signaling, JAK/STAT, and STMN1. The scope of the present review is to survey the current knowledge and high-light future perspectives for ANKHD1 in the malignant phenotype of cancer cells, exploring biological, functional, and clinical reports of this protein in cancer.
Collapse
Affiliation(s)
- Bruna Oliveira de Almeida
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-000, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
13
|
de Almeida BO, Machado-Neto JA. Emerging functions for ANKHD1 in cancer-related signaling pathways and cellular processes. BMB Rep 2020; 53:413-418. [PMID: 32635985 PMCID: PMC7473474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/10/2020] [Accepted: 06/22/2020] [Indexed: 08/02/2024] Open
Abstract
ANKHD1 (ankyrin repeat and KH domain containing 1) is a large protein characterized by the presence of multiple ankyrin repeats and a K-homology domain. Ankyrin repeat domains consist of widely existing protein motifs in nature, they mediate protein-protein interactions and regulate fundamental biological processes, while the KH domain binds to RNA or ssDNA and is associated with transcriptional and translational regulation. In recent years, studies containing relevant information on ANKHD1 in cancer biology and its clinical relevance, as well as the increasing complexity of signaling networks in which this protein acts, have been reported. Among the signaling pathways of interest in oncology regulated by ANKHD1 are Hippo signaling, JAK/STAT, and STMN1. The scope of the present review is to survey the current knowledge and highlight future perspectives for ANKHD1 in the malignant phenotype of cancer cells, exploring biological, functional, and clinical reports of this protein in cancer. [BMB Reports 2020; 53(8): 413-418].
Collapse
Affiliation(s)
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
14
|
Xue Y, Ge Y, Kang M, Wu C, Wang Y, Rong L, Fang Y. Selection of three miRNA signatures with prognostic value in non-M3 acute myeloid leukemia. BMC Cancer 2019; 19:109. [PMID: 30700251 PMCID: PMC6483142 DOI: 10.1186/s12885-019-5315-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND MiRNAs that are potential biomarkers for predicting prognosis for acute myeloid leukemia (AML) have been identified. However, comprehensive analyses investigating the association between miRNA expression profiles and AML survival remain relatively deficient. METHOD In the present study, we performed multivariate Cox's analysis and principal component analysis (PCA) using data from The Cancer Genome Atlas (TCGA) to identify potential molecular signatures for predicting non-M3 AML prognosis. RESULT We found that patients who were still living were significantly younger at diagnosis than those who had died (P = 0.001). In addition, there was a marked difference in living status among different risk category groups (P = 0.022). A multivariate Cox model suggested that three miRNAs were potential biomarkers of non-M3 AML prognosis, including miR-181a-2, miR-25 and miR-362. Subsequently, PCA analyses were conducted to comprehensively represent the expression levels of these three miRNAs in each patient with a PCA value. According to the log-rank test, AML outcome for patients with lower PCA values was significantly different from those with higher PCA values (P < 0.001). Further bioinformatic analysis revealed the biological functions of the selected miRNAs. CONCLUSION We conducted a comprehensive analysis of TCGA non-M3 AML data, identifying three miRNAs that are significantly correlated with AML survival. PCA values for the identified miRNAs are valuable for predicting AML prognosis.
Collapse
Affiliation(s)
- Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Yuqiu Ge
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Cong Wu
- Engineering Research Center of Wideband Wireless Communication Technology, Ministry of Education, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Yaping Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Liucheng Rong
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China. .,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Bai H, Sun Y, Liu N, Xue F, Li Y, Xu S, Ye J, Zhang L, Chen Y, Chen J. Single SNP- and pathway-based genome-wide association studies for beak deformity in chickens using high-density 600K SNP arrays. BMC Genomics 2018; 19:501. [PMID: 29954329 PMCID: PMC6022433 DOI: 10.1186/s12864-018-4882-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Background Beak deformity, typically expressed as the crossing of upper and lower mandibles, is found in several indigenous chicken breeds, including the Beijing-You chickens studied here. Beak deformity severely impairs the birds’ growth and welfare. Although previous studies shed some light on the genetic regulation of this complex trait, the genetic basis of this malformation remains incompletely understood. Results In this study, single SNP- and pathway-based genome-wide association studies (GWASs) were performed using ROADTRIPS and SNP ratio test (SRT), respectively. A total of 48 birds with deformed beaks (case) and 48 normal birds (control) were genotyped using Affymetrix 600 K HD genotyping arrays. As a result, 95 individuals and 429,539 SNPs were obtained after quality control. The P-value was corrected by a Bonferroni adjustment based on linkage disequilibrium pruning. The single SNP-based association study identified one associated SNP with 5% genome-wide significance and seven suggestively associated SNPs. Four high-confidence genes, LOC421892, TDRD3, RET, and STMN1, were identified as the most promising candidate genes underlying this complex trait in view of their positions, functions, and overlaps with previous studies. The pathway-based association study highlighted the association of six pathways with beak deformity, including the calcium signaling pathway. Conclusions Potentially useful candidate genes and pathways for beak deformity were identified, which should be the subject of further functional characterization. Electronic supplementary material The online version of this article (10.1186/s12864-018-4882-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Bai
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yanyan Sun
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Nian Liu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fuguang Xue
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yunlei Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Songshan Xu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jianhua Ye
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Lei Zhang
- CapitalBio Corporation, Beijing, 102206, China
| | - Yu Chen
- Beijing General Station of Animal Husbandry Service, Beijing, 102200, China
| | - Jilan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
16
|
Gieryng A, Pszczolkowska D, Bocian K, Dabrowski M, Rajan WD, Kloss M, Mieczkowski J, Kaminska B. Immune microenvironment of experimental rat C6 gliomas resembles human glioblastomas. Sci Rep 2017; 7:17556. [PMID: 29242629 PMCID: PMC5730558 DOI: 10.1038/s41598-017-17752-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/22/2017] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor, with ineffective anti-tumor responses and a poor prognosis despite aggressive treatments. GBM immune microenvironment is heterogenous and activation of specific immune populations in GBM is not fully characterized. Reliable animal models are critical for defining mechanisms of anti-tumor immunity. First we analyzed the immune subpopulations present in rat C6 gliomas. Using flow cytometry we determined kinetics of infiltration of myeloid cells and T lymphocytes into glioma-bearing brains. We found significant increases of the amoeboid, pro-tumorigenic microglia/macrophages, T helper (Th) and T regulatory (Treg) cells in tumor-bearing brains, and rare infiltrating T cytotoxic (Tc) cells. Transcriptomic analyses of glioma-bearing hemispheres revealed overexpression of invasion and immunosuppression-related genes, reflecting the immunosuppressive microenvironment. Microglia, sorted as CD11b+CD45low cells from gliomas, displayed the pro-invasive and immunosuppressive type of activation. Accumulation of Th and Treg cells combined with the reduced presence of Tc lymphocytes in rat gliomas may result in the lack of effective anti–tumor responses. Transcriptional profiles of CD11b+ cells and composition of immune infiltrates in C6 gliomas indicate that rat C6 gliomas employ similar immune system evasion strategies as human GBMs.
Collapse
Affiliation(s)
- Anna Gieryng
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Dominika Pszczolkowska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Katarzyna Bocian
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Wenson David Rajan
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Michal Kloss
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warszawa, Poland.
| |
Collapse
|
17
|
Machado-Neto JA, Rodrigues Alves APN, Fernandes JC, Coelho-Silva JL, Scopim-Ribeiro R, Fenerich BA, da Silva FB, Scheucher PS, Simões BP, Rego EM, Traina F. Paclitaxel induces Stathmin 1 phosphorylation, microtubule stability and apoptosis in acute lymphoblastic leukemia cells. Heliyon 2017; 3:e00405. [PMID: 29034341 PMCID: PMC5629349 DOI: 10.1016/j.heliyon.2017.e00405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/04/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a hematological malignancy characterized by abnormal proliferation and accumulation of lymphoblasts in the hematopoietic system. Stathmin 1 is a proliferation marker for normal lymphocytes, which has been described as highly expressed in ALL patients and functionally important for leukemia phenotype. In the present study, we expand our previous observations and aim to investigate Stathmin 1 expression and its impact on laboratory features and clinical outcomes in an independent cohort of ALL patients, and to verify the effects of paclitaxel treatment on Stathmin 1 phosphorylation and cell viability in ALL cell lines. In ALL patients, Stathmin 1 expression was significantly increased, associated with lower age onset and positively correlated with white blood cell counts, but did not impact on clinical outcomes. Functional assays revealed that paclitaxel induces Stathmin 1 phosphorylation at serine 16 (an inhibitory site), microtubule stability and apoptosis in Jurkat and Namalwa cell lines. Paclitaxel treatment did not modulate cell viability of normal peripheral blood leukocytes. In conclusion, our data confirm increased levels of Stathmin 1 in ALL patients and that therapeutic doses of paclitaxel inhibits Stathmin 1 function and promote microtubule stability and apoptosis in ALL cells.
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Ana Paula Nunes Rodrigues Alves
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Jaqueline Cristina Fernandes
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Juan Luiz Coelho-Silva
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Renata Scopim-Ribeiro
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Bruna Alves Fenerich
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Fernanda Borges da Silva
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Priscila Santos Scheucher
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Belinda Pinto Simões
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Eduardo Magalhães Rego
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Fabiola Traina
- Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
18
|
Vetter NS, Kolb EA, Mills CC, Sampson VB. The Microtubule Network and Cell Death Are Regulated by an miR-34a/Stathmin 1/βIII-Tubulin Axis. Mol Cancer Res 2017; 15:953-964. [PMID: 28275089 PMCID: PMC5500423 DOI: 10.1158/1541-7786.mcr-16-0372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/06/2016] [Accepted: 03/02/2017] [Indexed: 01/05/2023]
Abstract
MicroRNA-34a (miR-34a) is a master regulator of signaling networks that maintains normal physiology and disease and is currently in development as a miRNA-based therapy for cancer. Prior studies have reported low miR-34a expression in osteosarcoma; however, the molecular mechanisms underlying miR-34a activity in osteosarcoma are not well-defined. Therefore, this study evaluated the role of miR-34a in regulating signal transduction pathways that influence cell death in osteosarcoma. Levels of miR-34a were attenuated in human osteosarcoma cells and xenografts of the Pediatric Preclinical Testing Consortium (PPTC). Bioinformatics predictions identified stathmin 1 (STMN1) as a potential miR-34a target. Biotin pull-down assay and luciferase reporter analysis confirmed miR-34a target interactions within the STMN1 mRNA 3'-untranslated region. Overexpression of miR-34a in osteosarcoma cells suppressed STMN1 expression and reduced cell growth in vitro Restoration of miR-34a led to microtubule destabilization and increased βIII-tubulin expression, with corresponding G1-G2 phase cell-cycle arrest and apoptosis. Knockdown of the Sp1 transcription factor, by siRNA silencing, also upregulated βIII-tubulin expression in osteosarcoma cells, suggesting that miR-34a indirectly affects Sp1. Validating the coordinating role of miR-34a in microtubule destabilization, when miR-34a was combined with either microtubule inhibitors or chemotherapy, STMN1 phosphorylation was suppressed and there was greater cytotoxicity in osteosarcoma cells. These results demonstrate that miR-34a directly represses STMN1 gene and protein expression and upregulates βIII-tubulin, leading to disruption of the microtubule network and cell death.Implications: The miR-34a/STMN1/βIII-tubulin axis maintains the microtubule cytoskeleton in osteosarcoma, and combining miR-34a with microtubule inhibitors can be investigated as a novel therapeutic strategy. Mol Cancer Res; 15(7); 953-64. ©2017 AACR.
Collapse
Affiliation(s)
- Nancy S Vetter
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - E A Kolb
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | | | - Valerie B Sampson
- Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware.
| |
Collapse
|
19
|
RNAi targeting STMN alleviates the resistance to taxol and collectively contributes to down regulate the malignancy of NSCLC cells in vitro and in vivo. Cell Biol Toxicol 2017; 34:7-21. [DOI: 10.1007/s10565-017-9398-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
|
20
|
Machado-Neto JA, Pericole FV, Costa FF, Traina F, Olalla Saad ST. Stathmin 1 expression in plasma cell neoplasms. Rev Bras Hematol Hemoter 2017; 39:183-185. [PMID: 28577660 PMCID: PMC5457464 DOI: 10.1016/j.bjhh.2017.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/30/2017] [Accepted: 02/08/2017] [Indexed: 11/29/2022] Open
Affiliation(s)
| | | | | | - Fabiola Traina
- Universidade Estadual de Campinas/Hemocentro (UNICAMP), Campinas, SP, Brazil; Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | | |
Collapse
|
21
|
Fernandes JC, Rodrigues Alves APN, Machado-Neto JA, Scopim-Ribeiro R, Fenerich BA, da Silva FB, Simões BP, Rego EM, Traina F. IRS1/β-Catenin Axis Is Activated and Induces MYC Expression in Acute Lymphoblastic Leukemia Cells. J Cell Biochem 2017; 118:1774-1781. [DOI: 10.1002/jcb.25845] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/14/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Jaqueline Cristina Fernandes
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | | | - João Agostinho Machado-Neto
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Renata Scopim-Ribeiro
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Bruna Alves Fenerich
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Fernanda Borges da Silva
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Belinda Pinto Simões
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Eduardo Magalhães Rego
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| | - Fabiola Traina
- Department of Internal Medicine; University of São Paulo at Ribeirão Preto Medical School; Ribeirão Preto São Paulo Brazil
| |
Collapse
|
22
|
Biaoxue R, Xiguang C, Hua L, Shuanying Y. Stathmin-dependent molecular targeting therapy for malignant tumor: the latest 5 years' discoveries and developments. J Transl Med 2016; 14:279. [PMID: 27670291 PMCID: PMC5037901 DOI: 10.1186/s12967-016-1000-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/03/2016] [Indexed: 12/20/2022] Open
Abstract
Knowledge of the molecular mechanisms on malignant tumors is very critical for the development of new treatment strategies like molecularly targeted therapies. In last 5 years, many investigations suggest that stathmin is over-expressed in a variety of human malignant tumors, and potentially promotes the occurrence and development of tumors. Rather, down-regulation of stathmin can reduce cell proliferation, motility and metastasis and induce apoptosis of malignant tumors. Thus, a stathmin antagonist, such as a specific inhibitor (antibody, small molecule compound, peptide, or siRNA), may be a novel strategy of molecular targeted therapy. This review summarizes the research progress of recent 5 years on the role of stathmin in tumorigenesis, the molecular mechanisms and development of anti-stathmin treatment, which suggest that continued investigations into the function of stathmin in the tumorigenesis could lead to more rationally designed therapeutics targeting stathmin for treating human malignant tumors.
Collapse
Affiliation(s)
- Rong Biaoxue
- Department of Respiratory Medicine, First Affiliated Hospital, Xi'an Medical University, Xi'an, China.
| | - Cai Xiguang
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Liu Hua
- Department of Respiratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yang Shuanying
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Machado-Neto JA, de Melo Campos P, Favaro P, Lazarini M, da Silva Santos Duarte A, Lorand-Metze I, Costa FF, Saad STO, Traina F. Stathmin 1 inhibition amplifies ruxolitinib-induced apoptosis in JAK2V617F cells. Oncotarget 2016; 6:29573-84. [PMID: 26356819 PMCID: PMC4745747 DOI: 10.18632/oncotarget.4998] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/11/2015] [Indexed: 12/21/2022] Open
Abstract
The JAK/STAT pathway is constitutively activated in myeloproliferative neoplasms and can be inhibited by ruxolitinib, a selective JAK1/2 inhibitor. The JAK2(V617F) mutation leads to constitutive STAT3 phosphorylation and potentially leads to inhibition of Stathmin 1 activity via STAT3. In support of this hypothesis, we found that, in HEL JAK2(V617F) cells, ruxolitinib treatment decreased STAT3 and Stathmin 1 association, induced Stathmin 1 activation and microtubule instability. Silencing of Stathmin 1 significantly reduced cell proliferation and clonal growth, and increased apoptosis induced by ruxolitinib. Stathmin 1 silencing also prevented ruxolitinib-induced microtubule instability. To phenocopy the effect of Stathmin 1 inhibition, cells were treated with paclitaxel, a microtubule-stabilizing drug, in association or not with ruxolitinib; combined treatment significantly increased apoptosis, when compared to monotherapy. Notably, Stathmin 1 mRNA levels were highly expressed in CD34(+) cells from primary myelofibrosis patients. We then proposed that an undesired effect of ruxolitinib treatment may constitute Stathmin 1 activation and microtubule instability in JAK2(V617F) cells. Induction of microtubule stability, through Stathmin 1 silencing or paclitaxel treatment, combined with ruxolitinib could be an effective strategy for promoting apoptosis in JAK2(V617F) cells.
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Paula de Melo Campos
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Patricia Favaro
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil.,Current address: Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Mariana Lazarini
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil.,Current address: Department of Biological Sciences, Federal University of São Paulo, Diadema, São Paulo, Brazil
| | - Adriana da Silva Santos Duarte
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Irene Lorand-Metze
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Fabiola Traina
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil.,Current address: Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
24
|
Lazarini M, Machado-Neto JA, Duarte ADSS, Pericole FV, Vieira KP, Niemann FS, Alvarez M, Traina F, Saad STO. BNIP3L in myelodysplastic syndromes and acute myeloid leukemia: impact on disease outcome and cellular response to decitabine. Haematologica 2016; 101:e445-e448. [PMID: 27443286 DOI: 10.3324/haematol.2016.142521] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Mariana Lazarini
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil .,Department of Biological Sciences, Federal University of São Paulo, Diadema, Brazil
| | - João Agostinho Machado-Neto
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Adriana da Silva Santos Duarte
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Fernando Vieira Pericole
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Karla Priscila Vieira
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Fernanda S Niemann
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Marisa Alvarez
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| | - Fabiola Traina
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil.,Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Blood Transfusion Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| |
Collapse
|
25
|
Lin X, Yu T, Zhang L, Chen S, Chen X, Liao Y, Long D, Shen F. Silencing Op18/stathmin by RNA Interference Promotes the Sensitivity of Nasopharyngeal Carcinoma Cells to Taxol and High-Grade Differentiation of Xenografted Tumours in Nude Mice. Basic Clin Pharmacol Toxicol 2016; 119:611-620. [DOI: 10.1111/bcpt.12633] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/01/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Xuechi Lin
- Department of Medical Laboratory; Changsha Medical University; Changsha China
- Department of Anatomy, Histology and Embryology; Institute of Neuroscience; Changsha Medical University; Changsha China
| | - Ting Yu
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Lingxi Zhang
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Sangyan Chen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Xian Chen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Ying Liao
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Dan Long
- Department of Medical Laboratory; Changsha Medical University; Changsha China
| | - Fang Shen
- Department of Medical Laboratory; Changsha Medical University; Changsha China
- Department of Clinical Laboratory; the First Affiliated Hospital of Hunan Normal University; Changsha Hunan China
| |
Collapse
|
26
|
Lin X, Liao Y, Chen X, Long D, Yu T, Shen F. Regulation of Oncoprotein 18/Stathmin Signaling by ERK Concerns the Resistance to Taxol in Nonsmall Cell Lung Cancer Cells. Cancer Biother Radiopharm 2016; 31:37-43. [PMID: 26881937 DOI: 10.1089/cbr.2015.1921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Taxol is a cytotoxic antiepithelioma chemotherapy drug widely used clinically, which results in appearing a broad range of taxol-resistant tumors. Oncoprotein 18 (Op18)/stathmin is a genetically highly conserved small-molecule cytosolic phosphoprotein and highly expressed in tumors. Extracellular signal-regulated kinase (ERK) is a main member of mitogen-activated protein kinases (MAPKs). The study demonstrated that combination of blockage of ERK signal by ERK inhibitor PD98059 and Taxol greatly promoted taxol-induced cellular apoptosis and growth inhibition, decreased the expression of Op18/stathmin and total levels of phosphor-Op18/stathmin, while weakened the cyclin-dependent kinase 2 (cdc2) activity and antiapoptotic protein Bcl-2 expression and inhibited IL-10 autocrine in taxol-resistant NCI-H1299 cells; Taxol-resistant NCI-H1299 cells expressed high levels of ERK and phosphor-ERK in contrast to taxol-sensitive CNE1 cells, and ERK mainly phosphorylated Op18/stathmin at Ser 25 site. These findings suggest that ERK-mediated Op18/stathmin is involved in taxol resistance of tumors; blockage of ERK signal improves the sensitivity of tumor cells to taxol, which provides new clues for treating taxol-resistant carcinomas.
Collapse
Affiliation(s)
- Xuechi Lin
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China .,2 Department of Clinical Laboratory, Hunan Normal University , Changsha, Hunan
| | - Ying Liao
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China .,3 Department of Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University , Changsha, China
| | - Xian Chen
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China
| | - Dan Long
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China
| | - Ting Yu
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China
| | - Fang Shen
- 1 Department of Medical Laboratory, Changsha Medical University , Changsha, China .,2 Department of Clinical Laboratory, Hunan Normal University , Changsha, Hunan
| |
Collapse
|
27
|
High expression of stathmin 1 is a strong prognostic marker in oral squamous cell carcinoma patients treated by docetaxel-containing regimens. Clin Exp Med 2015; 17:45-50. [DOI: 10.1007/s10238-015-0403-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023]
|
28
|
Machado-Neto JA, Saad STO, Traina F. Stathmin 1 in normal and malignant hematopoiesis. BMB Rep 2015; 47:660-5. [PMID: 24667172 PMCID: PMC4345509 DOI: 10.5483/bmbrep.2014.47.12.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Indexed: 12/18/2022] Open
Abstract
Stathmin 1 is a microtubule destabilizer that plays an important role in cell cycle progression, segregation of chromosomes, clonogenicity, cell motility and survival. Stathmin 1 overexpression has been reported in malignant hematopoietic cells and Stathmin 1 inhibition reduces the highly proliferative potential of leukemia cell lines. However, during the differentiation of primary hematopoietic cells, Stathmin 1 expression decreases in parallel to decreases in the proliferative potential of early hematopoietic progenitors. The scope of the present review is to survey the current knowledge and highlight future perspectives for Stathmin 1 in normal and malignant hematopoiesis, with regard to the expression, function and clinical implications of this protein. [BMB Reports 2014; 47(12): 660-665]
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Fabiola Traina
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas; Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
29
|
Li X, Wang L, Li T, You B, Shan Y, Shi S, Qian L, Cao X. STMN1 overexpression correlates with biological behavior in human cutaneous squamous cell carcinoma. Pathol Res Pract 2015; 211:816-23. [PMID: 26235036 DOI: 10.1016/j.prp.2015.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 02/07/2023]
Abstract
Stathmin 1 (STMN1) is an important molecule in regulating cellular microtubule dynamics and promoting microtubule depolymerization in interphase and late mitosis. Evidences showed that STMN1 was up-regulated in many cancers, but there was no report about the roles of STMN1 in human cutaneous squamous cell carcinoma (cSCC). Here, we confirmed significant upregulation of STMN1 in cSCC tissues and cell lines compared with non-tumor counterparts. STMN1 upregulation was associated with the proliferation, migration, invasion and apoptosis of cSCC cells. The results suggested that STMN1 may play an important role in the development and tumor progression of cSCC.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Pathological Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Lulu Wang
- Nantong Municipal Center for Disease Control and Prevention, Nantong, Jiangsu 226001, People's Republic of China
| | - Tiejun Li
- Department of Pathological Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China; Small RNA Technology and Application Institute, Nantong University, Nantong, Jiangsu 226016, People's Republic of China
| | - Bo You
- Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Yin Shan
- Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Si Shi
- Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Li Qian
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| | - Xiaolei Cao
- Department of Pathological Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, People's Republic of China.
| |
Collapse
|
30
|
Machado-Neto JA, Lazarini M, Favaro P, de Melo Campos P, Scopim-Ribeiro R, Franchi Junior GC, Nowill AE, Lima PRM, Costa FF, Benichou S, Olalla Saad ST, Traina F. ANKHD1 silencing inhibits Stathmin 1 activity, cell proliferation and migration of leukemia cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:583-93. [PMID: 25523139 DOI: 10.1016/j.bbamcr.2014.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/29/2014] [Accepted: 12/10/2014] [Indexed: 12/30/2022]
Abstract
ANKHD1 is highly expressed in human acute leukemia cells and potentially regulates multiple cellular functions through its ankyrin-repeat domains. In order to identify interaction partners of the ANKHD1 protein and its role in leukemia cells, we performed a yeast two-hybrid system screen and identified SIVA, a cellular protein known to be involved in proapoptotic signaling pathways. The interaction between ANKHD1 and SIVA was confirmed by co-imunoprecipitation assays. Using human leukemia cell models and lentivirus-mediated shRNA approaches, we showed that ANKHD1 and SIVA proteins have opposing effects. While it is known that SIVA silencing promotes Stathmin 1 activation, increased cell migration and xenograft tumor growth, we showed that ANKHD1 silencing leads to Stathmin 1 inactivation, reduced cell migration and xenograft tumor growth, likely through the inhibition of SIVA/Stathmin 1 association. In addition, we observed that ANKHD1 knockdown decreases cell proliferation, without modulating apoptosis of leukemia cells, while SIVA has a proapoptotic function in U937 cells, but does not modulate proliferation in vitro. Results indicate that ANKHD1 binds to SIVA and has an important role in inducing leukemia cell proliferation and migration via the Stathmin 1 pathway. ANKHD1 may be an oncogene and participate in the leukemia cell phenotype.
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Mariana Lazarini
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Patricia Favaro
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Paula de Melo Campos
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Renata Scopim-Ribeiro
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Gilberto Carlos Franchi Junior
- Integrated Center for Childhood Onco-Hematological Investigation, University of Campinas, Campinas 13083-878, São Paulo, Brazil
| | - Alexandre Eduardo Nowill
- Integrated Center for Childhood Onco-Hematological Investigation, University of Campinas, Campinas 13083-878, São Paulo, Brazil
| | - Paulo Roberto Moura Lima
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | | | - Sara Teresinha Olalla Saad
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil
| | - Fabiola Traina
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas 13083-878, São Paulo, Brazil.
| |
Collapse
|