1
|
Cui Y, Yun T, Zhao GG. Bioinformatics analysis of clinical significance of STMN1 gene in prognosis and immune infiltration in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2022; 30:411-419. [DOI: 10.11569/wcjd.v30.i9.411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND STMN1 (stathmin1), a member of the microtubule destabilizing protein family, is transcriptionally repressed by the functional tumor suppressor protein p53. STMN1 was first identified as a cellular phosphorylated protein overexpressed in leukemia in 1983. STMN1 was found to be upregulated in a variety of cancers, such as non-small cell lung cancer, breast cancer, and gastric cancer, and it can induce cell differentiation, proliferation, and migration in solid tumors and is associated with a poor clinical prognosis. However, the role and mechanism of STMN1 in hepatocellular carcinoma remain unclear.
AIM To assess the expression of STMN1 gene in hepatocellular carcinoma and its relationship with the clinical characteristics and prognosis of patients, and to explore the molecular mechanism of STMN1 gene in this malignancy.
METHODS Clinical information and high-throughput RNA-sequencing data of hepatocellular carcinoma patients were downloaded from The Cancer Genome Atlas (https://portal.gdc.cancer.gov, TCGA) database. R (v3.6.2) software was used to analyze the expression of STMN1 gene in hepatocellular carcinoma tissues, and its relationship with clinical characteristics of patients was analyzed. Univariate and multivariate Cox regression models were used to analyze the prognostic value of STMN1 in hepatocellular carcinoma. Gene enrichment analysis was used to analyze STMN1 expression-related pathway mechanisms. ssGSEA method was used to analyze the infiltration of 24 immune cell types in tumors, and to explore the relationship between STMN1 and immune cells.
RESULTS STMN1 was highly expressed in tumor tissues (P < 0.001). The diagnostic value of STMN1 was assessed by ROC curve analysis, and the area under the ROC curve (AUC) was 0.971. The predictive ability of STMN1 had high accuracy. Chi-square test or Fisher exact test revealed that STMN1 expression was significantly associated with overall survival rate (P = 0.013), T stage (P = 0.002), TNM stage (P = 0.008), age (P = 0.006), and tumor grade (P < 0.001). The overall survival rate, progression-free survival rate, and disease-specific survival rate of the high STMN1 gene expression group were lower than those of the low expression group (P < 0.01). Cox regression analysis showed that the high expression of STMN1 was an independent risk factor for the prognosis of patients with hepatocellular carcinoma (hazard ratio = 1.808, 95% confidence interval: 1.288-2.234, P = 0.014). GSEA analysis showed that STMN1 gene in hepatocellular carcinoma is mainly involved in cell cycle, oocyte meiosis, T cell receptor signaling pathway, natural killer cell-mediated cytotoxicity, and spliceosome. Immune infiltration analysis showed that STMN1 expression was positively correlated with helper T cells 2, follicular helper T cells, and helper T cells, and was negatively correlated with neutrophils, helper T cells 17, and dendritic cells.
CONCLUSION The expression of STMN1 gene is up-regulated in hepato-cellular carcinoma, which is associated with a poor prognosis of patients. It is an independent prognostic factor for hepatocellular carcinoma patients and is expected to become a potential molecular marker of this malignancy.
Collapse
Affiliation(s)
- Yu Cui
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Tao Yun
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Guo-Gang Zhao
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| |
Collapse
|
2
|
PTEN loss promotes oncogenic function of STMN1 via PI3K/AKT pathway in lung cancer. Sci Rep 2021; 11:14318. [PMID: 34253824 PMCID: PMC8275769 DOI: 10.1038/s41598-021-93815-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 06/30/2021] [Indexed: 12/25/2022] Open
Abstract
Among all cancer types, lung cancer has already become the leading cause of cancer-related death around the world. The molecular mechanism understanding this development is still needed to be improved to treat lung cancer. Stathmin (STMN1) was initially identified as a cytoplasmic protein phosphorylated responding to cell signal and controlled cell physiological processes. The dysregulation of STMN1 is found in various kinds of tumors. However, the molecular mechanism of STMN1 regulating lung cancer is still unclear. Here, we found that STMN1 was overexpressed in lung cancer tissues and associated with worse survival rates of lung cancer patients. Inhibition of STMN1 suppressed lung cancer cell growth, migration and invasion, and promoted drug sensitivity. Moreover, PTEN loss promoted STMN1 expression via PI3K/AKT pathway. PTEN loss ameliorated the inhibition of cell growth, migration and invasion, and drug sensitivity induced by STMN1 knockdown in lung cancer. The high expression of STMN1 was negatively correlated with the low expression of PTEN in lung cancer specimens. Overall, our work demonstrated that PTEN regulated the oncogenic function of STMN1 in lung cancer.
Collapse
|
3
|
Pollock KE, Talton OO, Schulz LC. Morphology and gene expression in mouse placentas lacking leptin receptors. Biochem Biophys Res Commun 2020; 528:336-342. [PMID: 32248977 DOI: 10.1016/j.bbrc.2020.03.104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 11/24/2022]
Abstract
In the pregnant mouse, the hormone leptin is primarily produced by adipose tissue and does not significantly cross the placenta into fetal circulation. Nonetheless, leptin treatment during gestation affects offspring phenotypes. Leptin treatment also affects placental trophoblast cells in vitro, by altering proliferation, invasion and nutrient transport. The goal of the present study was to determine whether the absence of placental leptin receptors alters placental development and gene expression. Leprdb-3j+ mice possessing only one functional copy of the leptin receptor were mated to obtain wildtype, Leprdb-3j+ and Leprdb-3j/db-3j conceptuses, which were then transferred to wildtype recipient dams. Placentas were collected at gestational d18.5 to examine placental morphology and gene expression. Placentas lacking functional leptin receptor had reduced weights, but were otherwise morphologically indistinguishable from control placentas. Relative mRNA levels, however, were altered in Leprdb-3j/db-3j placentas, particularly transcripts related to amino acid and lipid metabolism and transport. Consistent with a previous in vitro study, leptin was found to promote expression of stathmin, a positive regulator of trophoblast invasion, and of serotonin receptors, potential mediators of offspring neurological development. Overall placental leptin receptor was found not to play a significant role in morphological development of the placenta, but to regulate placental gene expression, including in metabolic pathways that affect fetal growth.
Collapse
Affiliation(s)
- Kelly E Pollock
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65212, USA
| | | | - Laura C Schulz
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
4
|
Shan W, Han F, Xu Y, Shi Y. Stathmin Regulates Spatiotemporal Variation in the Memory Loop in Single-Prolonged Stress Rats. J Mol Neurosci 2020; 70:576-589. [PMID: 31933182 DOI: 10.1007/s12031-019-01459-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/29/2019] [Indexed: 12/29/2022]
Abstract
Posttraumatic stress disorder (PTSD) is closely related to brain structures of the memory loop such as the hippocampus, amygdala, and medial prefrontal cortex (mPFC). The fear gene stathmin plays an important role in regulating fear memory. However, whether the fear gene stathmin is related to fear memory loop anomalies caused by PTSD is unclear. A single-prolonged stress (SPS) rat model of PTSD was constructed. Wistar rats were randomly divided into 5 groups: the control group, SPS 1-day group, SPS 4-day group, SPS 7-day group, and SPS 14-day group. Then, we measured the protein and mRNA expression of stathmin, p-stathmin (Ser16, Ser25, Ser38, and Ser63), β-tubulin, and MAP-1B in the hippocampus, amygdala, and mPFC in the 5 groups by immunohistochemistry, Western blotting, and qRT-PCR. The expression of the stathmin protein in the hippocampus, mPFC, and amygdala of the rat memory loop decreased gradually in the SPS 1-day group, the SPS 4-day group, and the SPS 7-day group, in which it was the lowest, and then increased. The trend of the expression of stathmin mRNA in the three areas of the memory loop was consistent with the trend of the expression of the stathmin protein. The trend of the protein expression of p-stathmin (Ser25 and Ser38) was opposite of that of stathmin; it reached a peak on the 7th day, and then decreased in the hippocampus. The protein expression of p-stathmin (Ser63) showed the same trend in the mPFC. The protein and mRNA expression of β-tubulin and MAP-1B was consistent with that of p-stathmin; it reached a peak on the 7th day, and then decreased in the rat hippocampus, mPFC, and amygdala. Stathmin in the memory loop, especially in the hippocampus, regulates microtubule structure through its phosphorylation at Ser25 and Ser38 and thereby participates in the mediation of fear memory abnormalities in PTSD.
Collapse
Affiliation(s)
- Wei Shan
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shengbei New District, Shenyang, 110001, People's Republic of China.,Department of Human Anatomy, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shengbei New District, Shenyang, 110001, People's Republic of China
| | - Yanhao Xu
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shengbei New District, Shenyang, 110001, People's Republic of China.
| | - Yuxiu Shi
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 77, Puhe Road, Shengbei New District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
5
|
Liu H, Li Y, Li Y, Zhou L, Bie L. STMN1 as a candidate gene associated with atypical meningioma progression. Clin Neurol Neurosurg 2017. [DOI: 10.1016/j.clineuro.2017.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
6
|
Chauvin S, Sobel A. Neuronal stathmins: A family of phosphoproteins cooperating for neuronal development, plasticity and regeneration. Prog Neurobiol 2015; 126:1-18. [DOI: 10.1016/j.pneurobio.2014.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/23/2014] [Accepted: 09/29/2014] [Indexed: 02/06/2023]
|
7
|
Lu Y, Liu C, Xu YF, Cheng H, Shi S, Wu CT, Yu XJ. Stathmin destabilizing microtubule dynamics promotes malignant potential in cancer cells by epithelial-mesenchymal transition. Hepatobiliary Pancreat Dis Int 2014; 13:386-94. [PMID: 25100123 DOI: 10.1016/s1499-3872(14)60038-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Stathmin is a ubiquitous cytosolic regulatory phosphoprotein and is overexpressed in different human malignancies. The main physiological function of stathmin is to interfere with microtubule dynamics by promoting depolymerization of microtubules or by preventing polymerization of tubulin heterodimers. Stathmin plays important roles in regulating many cellular functions as a result of its microtubule-destabilizing activity. Currently, the critical roles of stathmin in cancer cells, as well as in lymphocytes have been valued. This review discusses stathmin and microtubule dynamics in cancer development, and hypothesizes their possible relationship with epithelial-mesenchymal transition (EMT). DATA SOURCES A PubMed search using such terms as "stathmin", "microtubule dynamics", "epithelial-mesenchymal transition", "EMT", "malignant potential" and "cancer" was performed to identify relevant studies published in English. More than 100 related articles were reviewed. RESULTS The literature clearly documented the relationship between stathmin and its microtubule-destabilizing activity of cancer development. However, the particular mechanism is poorly understood. Microtubule disruption is essential for EMT, which is a crucial process during cancer development. As a microtubule-destabilizing protein, stathmin may promote malignant potential in cancer cells by initiating EMT. CONCLUSIONS We propose that there is a stathmin-microtubule dynamics-EMT (S-M-E) axis during cancer development. By this axis, stathmin together with its microtubule-destabilizing activity contributes to EMT, which stimulates the malignant potential in cancer cells.
Collapse
Affiliation(s)
- Yu Lu
- Pancreatic Cancer Institute, Fudan University; Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | | | |
Collapse
|
8
|
Duncan JE, Lytle NK, Zuniga A, Goldstein LSB. The Microtubule Regulatory Protein Stathmin Is Required to Maintain the Integrity of Axonal Microtubules in Drosophila. PLoS One 2013; 8:e68324. [PMID: 23840848 PMCID: PMC3694009 DOI: 10.1371/journal.pone.0068324] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 05/22/2013] [Indexed: 12/25/2022] Open
Abstract
Axonal transport, a form of long-distance, bi-directional intracellular transport that occurs between the cell body and synaptic terminal, is critical in maintaining the function and viability of neurons. We have identified a requirement for the stathmin (stai) gene in the maintenance of axonal microtubules and regulation of axonal transport in Drosophila. The stai gene encodes a cytosolic phosphoprotein that regulates microtubule dynamics by partitioning tubulin dimers between pools of soluble tubulin and polymerized microtubules, and by directly binding to microtubules and promoting depolymerization. Analysis of stai function in Drosophila, which has a single stai gene, circumvents potential complications with studies performed in vertebrate systems in which mutant phenotypes may be compensated by genetic redundancy of other members of the stai gene family. This has allowed us to identify an essential function for stai in the maintenance of the integrity of axonal microtubules. In addition to the severe disruption in the abundance and architecture of microtubules in the axons of stai mutant Drosophila, we also observe additional neurological phenotypes associated with loss of stai function including a posterior paralysis and tail-flip phenotype in third instar larvae, aberrant accumulation of transported membranous organelles in stai deficient axons, a progressive bang-sensitive response to mechanical stimulation reminiscent of the class of Drosophila mutants used to model human epileptic seizures, and a reduced adult lifespan. Reductions in the levels of Kinesin-1, the primary anterograde motor in axonal transport, enhance these phenotypes. Collectively, our results indicate that stai has an important role in neuronal function, likely through the maintenance of microtubule integrity in the axons of nerves of the peripheral nervous system necessary to support and sustain long-distance axonal transport.
Collapse
Affiliation(s)
- Jason E. Duncan
- Department of Biology, Willamette University, Salem, Oregon, United States of America
- * E-mail:
| | - Nikki K. Lytle
- Department of Biology, Willamette University, Salem, Oregon, United States of America
| | - Alfredo Zuniga
- Department of Biology, Willamette University, Salem, Oregon, United States of America
| | - Lawrence S. B. Goldstein
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
9
|
Decreased stathmin expression ameliorates neuromuscular defects but fails to prolong survival in a mouse model of spinal muscular atrophy. Neurobiol Dis 2013; 52:94-103. [DOI: 10.1016/j.nbd.2012.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 11/08/2012] [Accepted: 11/22/2012] [Indexed: 02/02/2023] Open
|
10
|
Carney BK, Caruso Silva V, Cassimeris L. The microtubule cytoskeleton is required for a G2 cell cycle delay in cancer cells lacking stathmin and p53. Cytoskeleton (Hoboken) 2012; 69:278-89. [PMID: 22407961 DOI: 10.1002/cm.21024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 02/24/2012] [Accepted: 02/29/2012] [Indexed: 12/25/2022]
Abstract
In several cancer cell lines, depleting the microtubule (MT)-destabilizing protein stathmin/oncoprotein18 leads to a G2 cell cycle delay and apoptosis. These phenotypes are observed only in synergy with low levels of p53, but the pathway(s) activated by stathmin depletion to delay the cell cycle are unknown. We found that stathmin depletion caused greater MT stability in synergy with loss of p53, measured by the levels of acetylated α-tubulin and the rate of centrosomal MT nucleation. Nocodazole or vinblastine-induced MT depolymerization abrogated the stathmin-depletion induced G2 delay, measured by the percentage of cells staining positive for several markers (TPX2, CDK1 with inhibitory phosphorylation), indicating that MTs are required to lengthen G2. Live cell imaging showed that stathmin depletion increased time in G2 without an impact on the duration of mitosis, indicating that the longer interphase duration is not simply a consequence of a previous slowed mitosis. In contrast, stabilization of MTs with paclitaxel (8 nM) slowed mitosis without lengthening the duration of interphase, demonstrating that increased MT stability alone is not sufficient to delay cells in G2.
Collapse
Affiliation(s)
- Bruce K Carney
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | | | | |
Collapse
|
11
|
A Study of the Wound Healing Mechanism of a Traditional Chinese Medicine, Angelica sinensis, Using a Proteomic Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:467531. [PMID: 22536285 PMCID: PMC3319019 DOI: 10.1155/2012/467531] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/19/2012] [Indexed: 11/18/2022]
Abstract
Angelica sinensis (AS) is a traditional Chinese herbal medicine that has been formulated clinically to treat various form of skin trauma and to help wound healing. However, the mechanism by which it works remains a mystery. In this study we have established a new platform to evaluate the pharmacological effects of total AS herbal extracts as well as its major active component, ferulic acid (FA), using proteomic and biochemical analysis. Cytotoxic and proliferation-promoting concentrations of AS ethanol extracts (AS extract) and FA were tested, and then the cell extracts were subject to 2D PAGE analysis. We found 51 differentially expressed protein spots, and these were identified by mass spectrometry. Furthermore, biomolecular assays, involving collagen secretion, migration, and ROS measurements, gave results that are consistent with the proteomic analysis. In this work, we have demonstrated a whole range of pharmacological effects associated with Angelica sinensis that might be beneficial when developing a wound healing pharmaceutical formulation for the herbal medicine.
Collapse
|
12
|
Baquero MT, Hanna JA, Neumeister V, Cheng H, Molinaro AM, Harris LN, Rimm DL. Stathmin expression and its relationship to microtubule-associated protein tau and outcome in breast cancer. Cancer 2012; 118:4660-9. [PMID: 22359235 DOI: 10.1002/cncr.27453] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/06/2011] [Indexed: 01/23/2023]
Abstract
BACKGROUND Microtubule-associated proteins (MAPs) endogenously regulate microtubule stability. Here, the prognostic value of stathmin, a destabilizing protein, was assessed in combination with MAP-tau, a stabilizing protein, in order to evaluate microtubule stabilization as a potential biomarker. METHODS Stathmin and MAP-tau expression levels were measured in a breast cancer cohort (n = 651) using the tissue microarray format and quantitative immunofluorescence (AQUA) technology, then correlated with clinical and pathological characteristics and disease-free survival. RESULTS Univariate Cox proportional hazard models indicated that high stathmin expression predicts worse overall survival (hazard ratio [HR] = 1.48; 95% confidence interval [CI] = 1.119-1.966; P = .0061). Survival analysis showed 10-year survival of 53.1% for patients with high stathmin expression versus 67% for low expressers (log-rank, P < .003). Cox multivariate analysis showed high stathmin expression was independent of age, menopausal status, nodal status, nuclear grade, tumor size, and estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 expression (HR = 1.19; 95% CI = 1.03-1.37; P = .01). The ratio of MAP-tau to stathmin expression showed a positive correlation to disease-free survival (HR = 0.679; 95% CI = 0.517-0.891; P = .0053) with a 10-year survival of 65.4% for patients who had a high ratio of MAP-tau to stathmin versus 52.5% 10-year survival rate for those with a low ratio (log-rank, P = .0009). Cox multivariate analysis showed the ratio of MAP-tau to stathmin was an independent predictor of overall survival (HR = 0.609; 95% CI = 0.422-0.879; P = .008). CONCLUSIONS Low stathmin and high MAP-tau are associated with increased microtubule stability and better prognosis in breast cancer.
Collapse
Affiliation(s)
- Maria T Baquero
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Belletti B, Baldassarre G. Stathmin: a protein with many tasks. New biomarker and potential target in cancer. Expert Opin Ther Targets 2011; 15:1249-66. [PMID: 21978024 DOI: 10.1517/14728222.2011.620951] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Stathmin is a microtubule-destabilizing phosphoprotein, firstly identified as the downstream target of many signal transduction pathways. Several studies then indicated that stathmin is overexpressed in many types of human malignancies, thus deserving the name of Oncoprotein 18 (Op18). At molecular level, stathmin depolymerizes microtubules by either sequestering free tubulin dimers or directly inducing microtubule-catastrophe. A crucial role for stathmin in the control of mitosis has been proposed, since both its overexpression and its downregulation induce failure in the correct completion of cell division. Accordingly, stathmin is an important target of the main regulator of M phase, cyclin-dependent kinase 1. AREAS COVERED Recent evidences support a role for stathmin in the regulation of cell growth and motility, both in vitro and in vivo, and indicate its involvement in advanced, invasive and metastatic cancer more than in primary tumors. EXPERT OPINION Many studies suggest that high stathmin expression levels in cancer negatively influence the response to microtubule-targeting drugs. These notions together with the fact that stathmin is expressed at very low levels in most adult tissues strongly support the use of stathmin as marker of prognosis and as target for novel anti-tumoral and anti-metastatic therapies.
Collapse
Affiliation(s)
- Barbara Belletti
- National Cancer Institute, Centro di Riferimento Oncologico, Division of Experimental Oncology 2, Via Franco Gallini, 2, 33081 Aviano, Italy
| | | |
Collapse
|