151
|
Targeting PTEN in Colorectal Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1110:55-73. [DOI: 10.1007/978-3-030-02771-1_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
152
|
Huang XF, Li JJ, Tao YG, Wang XQ, Zhang RL, Zhang JL, Su ZQ, Huang QH, Deng YH. Geniposide attenuates Aβ25–35-induced neurotoxicity via the TLR4/NF-κB pathway in HT22 cells. RSC Adv 2018; 8:18926-18937. [PMID: 35539637 PMCID: PMC9080630 DOI: 10.1039/c8ra01038b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/16/2018] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is marked by the accumulation of amyloid-β (Aβ) and neuroinflammation which promote the development of AD. Geniposide, the main ingredient isolated from Chinese herbal medicine Gardenia jasminoides Ellis, has a variety of pharmacological functions such as anti-apoptosis and anti-inflammatory activity. Hence, we estimated the inflammatory cytotoxicity caused by Aβ25–35 and the neuroprotective effects of geniposide in HT22 cells. In this research, following incubation with Aβ25–35 (40 μM, 24 h) in HT22 cells, the methylthiazolyl tetrazolium (MTT) and lactate dehydrogenase (LDH) release assays showed that the cell survival rate was significantly decreased. In contrast, the reactive oxygen species (ROS) assay indicated that Aβ25–35 enhanced ROS accumulation and apoptosis showed in both hoechst 33342 staining and annexin V-FITC/PI double staining. And then, immunofluorescence test revealed that Aβ25–35 promoted p65 to transfer into the nucleus indicating p65 was activated by Aβ25–35. Moreover, western blot analysis proved that Aβ25–35 increased the expression of nitric oxide species (iNOS), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2) and interleukin-1β (IL-1β). Simultaneously, Aβ25–35 also promoted the expression of toll-like receptor 4 (TLR4), p-p65 and p-IκB-α accompanied with the increase in the level of beta-secretase 1 (BACE1) and caspase-3 which further supported Aβ25–35 induced apoptosis and inflammation. Fortunately, this up-regulation was reversed by geniposide. In conclusion, our data suggest that geniposide can alleviate Aβ25–35-induced inflammatory response to protect neurons, which is possibly involved with the inhibition of the TLR4/NF-κB pathway in HT22 cells. Geniposide may be the latent treatment for AD induced by neuroinflammation and apoptosis. Alzheimer's disease (AD), a neurodegenerative disorder, is marked by the accumulation of amyloid-β (Aβ) and neuroinflammation which promote the development of AD.![]()
Collapse
Affiliation(s)
- Xiu-Fang Huang
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University
- Guangzhou
- China
| | - Jian-Jun Li
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University
- Guangzhou
- China
| | - Yan-Gu Tao
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University
- Guangzhou
- China
| | - Xie-Qi Wang
- Dermatology Hospital of Southern Medical University
- Dermatology Hospital of Guangdong Province
- Guangzhou
- China
| | - Ru-Lan Zhang
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University
- Guangzhou
- China
| | - Jia-Lin Zhang
- Dermatology Hospital of Southern Medical University
- Dermatology Hospital of Guangdong Province
- Guangzhou
- China
| | - Zu-Qing Su
- Guangdong Provincial Hospital of Traditional Chinese Medicine
- Guangzhou
- China
| | - Qi-Hui Huang
- Sun Yat-Sen Memorial Hospital of Sun Yat-sen University
- Guangzhou
- China
| | - Yuan-Hui Deng
- Guangdong Provincial Hospital of Traditional Chinese Medicine
- Guangzhou
- China
| |
Collapse
|
153
|
Riquelme SA, Hopkins BD, Wolfe AL, DiMango E, Kitur K, Parsons R, Prince A. Cystic Fibrosis Transmembrane Conductance Regulator Attaches Tumor Suppressor PTEN to the Membrane and Promotes Anti Pseudomonas aeruginosa Immunity. Immunity 2017; 47:1169-1181.e7. [PMID: 29246444 PMCID: PMC5738266 DOI: 10.1016/j.immuni.2017.11.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/11/2017] [Accepted: 11/06/2017] [Indexed: 12/14/2022]
Abstract
The tumor suppressor PTEN controls cell proliferation by regulating phosphatidylinositol-3-kinase (PI3K) activity, but the participation of PTEN in host defense against bacterial infection is less well understood. Anti-inflammatory PI3K-Akt signaling is suppressed in patients with cystic fibrosis (CF), a disease characterized by hyper-inflammatory responses to airway infection. We found that Ptenl-/- mice, which lack the NH2-amino terminal splice variant of PTEN, were unable to eradicate Pseudomonas aeruginosa from the airways and could not generate sufficient anti-inflammatory PI3K activity, similar to what is observed in CF. PTEN and the CF transmembrane conductance regulator (CFTR) interacted directly and this interaction was necessary to position PTEN at the membrane. CF patients under corrector-potentiator therapy, which enhances CFTR transport to the membrane, have increased PTEN amounts. These findings suggest that improved CFTR trafficking could enhance P. aeruginosa clearance from the CF airway by activating PTEN-mediated anti-bacterial responses and might represent a therapeutic strategy.
Collapse
Affiliation(s)
| | | | - Andrew L Wolfe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emily DiMango
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Kipyegon Kitur
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Ramon Parsons
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alice Prince
- Department of Pediatrics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
154
|
Bai J, Gao Z, Li X, Dong L, Han W, Nie J. Regulation of PD-1/PD-L1 pathway and resistance to PD-1/PD-L1 blockade. Oncotarget 2017; 8:110693-110707. [PMID: 29299180 PMCID: PMC5746415 DOI: 10.18632/oncotarget.22690] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/08/2017] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint blockades, such as inhibitors against programmed death 1 (PD-1) and its ligand (PD-L1), have received extensive attention in the past decade because of their dramatic clinical outcomes in advanced malignancies. However, both primary and acquired resistance becomes one of the major obstacles, which greatly limits the long-lasting effects and wide application of PD-1/PD-L1 blockade therapy. PD-1/PD-L1 both regulates and is regulated by cellular signaling pathways and epigenetic modification, thus inhibiting the proliferation and effector function of T and B cells. The lack of tumor antigens and effective antigen presentation, aberrant activation of oncogenic pathways, mutations in IFN-γ signaling, immunosuppressive tumor microenvironment such as regulatory T cells, myeloid-derived suppressor cells, M2 macrophages, and immunoinhibitory cytokines can lead to resistance to PD-1/PD-L1 blockade. In this review, we describe PD-1 related signaling pathways, essential factors contributing to the resistance of PD-1 blockade, and discuss strategies to increase the efficacy of immunotherapy. Furthermore, we discuss the possibility of combined epigenetic therapy with PD-1 blockade as a potential promising approach for cancer treatment.
Collapse
Affiliation(s)
- Jie Bai
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhitao Gao
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiang Li
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Liang Dong
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Han
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Nie
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
155
|
Yang Z, Cao X, Xu W, Xie C, Chen J, Zhu Y, Lu N. Phosphorylation of phosphatase and tensin homolog induced by Helicobacter pylori promotes cell invasion by activation of focal adhesion kinase. Oncol Lett 2017; 15:1051-1057. [PMID: 29399165 PMCID: PMC5772772 DOI: 10.3892/ol.2017.7430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 10/18/2017] [Indexed: 02/06/2023] Open
Abstract
Phosphorylation of the phosphatase and tensin homolog (PTEN) tumor suppressor at Ser380/Thr382/Thr383 residues is a novel mechanism underlying PTEN inactivation in gastric carcinogenesis, which may be triggered by Helicobacter pylori infection. To investigate this further, the effect of H. pylori infection on PTEN phosphorylation and the subsequent activation of focal adhesion kinase (FAK), were evaluated in gastric tissue samples from Mongolian gerbils and in the human gastric epithelial mucosa cell line GES-1 using immunohistochemistry, western blotting and Transwell assays. The in vivo and in vitro results of the present study demonstrated that H. pylori infection induced the phosphorylation and inactivation of PTEN at Ser380/Thr382/383, and the subsequent phosphorylation and activation of FAK at Tyr397. Gastric epithelial cell invasion was also increased. Furthermore, stable expression of a dominant-negative PTEN mutant inhibited the enhanced FAK activation and cell invasion induced by H. pylori infection. These results suggest that the mechanism underlying H. pylori-induced carcinogenesis may involve promoting cell invasion through the phosphorylation of PTEN and the activation of FAK.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ximei Cao
- Department of Gastroenterology, First Peoples' Hospital of Jiujiang, Jiujiang, Jiangxi 332000, P.R. China
| | - Wenting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
156
|
Makker A, Goel MM, Nigam D, Mahdi AA, Das V, Agarwal A, Pandey A, Gautam A. Aberrant Akt Activation During Implantation Window in Infertile Women With Intramural Uterine Fibroids. Reprod Sci 2017; 25:1243-1253. [PMID: 29113583 DOI: 10.1177/1933719117737844] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The objective of the study was to examine the expression and cellular distribution of key signaling components of the phosphatidylinositol-3-kinase (PI3K)/Phosphatase and Tensin Homolog Deleted on Chromosome Ten (PTEN)/Protein Kinase B (PKB/Akt) pathway during the window of implantation in infertile women with noncavity-distorting intramural uterine fibroids (n = 21) as compared to fertile controls (n = 15). Relative gene expression analysis of PIK3CA, PTEN, Akt1, and Akt2 genes in midluteal endometrial biopsies was performed by real-time polymerase chain reaction. Immunohistochemistry was used to evaluate the expression of PIK3CA, PTEN, phospho-PTEN, Akt1, Akt2, phospho-Akt1 (serine 473), phospho-Akt1 (threonine 308), and Ki67 proteins. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling assay was performed for apoptosis detection. In comparison to fertile controls, significant upregulation of Akt1 messenger RNA levels (2.16-fold; P < .05); cell-specific upregulation of the proteins phospho-PTEN ( P < .05), Akt1 ( P < .05), Akt2 ( P < .05), and p-Akt (S473; P < .001); and downregulation of PTEN ( P < .01) were observed in endometrium of infertile women with intramural fibroids. The ratio of p-PTEN/PTEN and p-Akt1 (S473)/Akt1 was also significantly higher in infertile women. Increased Ki67 labeling index in the glandular epithelium and significantly lower apoptotic index in glandular epithelium and stroma were seen in infertile women during the window of implantation. Aberrant Akt activation and the associated imbalance in endometrial proliferation and apoptosis observed in infertile women with intramural fibroids during the midsecretory phase might contribute to impaired endometrial receptivity leading to infertility in these patients.
Collapse
Affiliation(s)
- Annu Makker
- 1 Department of Pathology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Madhu Mati Goel
- 1 Department of Pathology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Dipti Nigam
- 1 Department of Pathology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Abbas Ali Mahdi
- 2 Department of Biochemistry, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Vinita Das
- 3 Department of Obstetrics and Gynecology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Anjoo Agarwal
- 3 Department of Obstetrics and Gynecology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Amita Pandey
- 3 Department of Obstetrics and Gynecology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Abnish Gautam
- 4 Government Degree College, Mahona, Lucknow, Uttar Pradesh, India
| |
Collapse
|
157
|
Kim DH, Suh J, Surh YJ, Na HK. Regulation of the tumor suppressor PTEN by natural anticancer compounds. Ann N Y Acad Sci 2017; 1401:136-149. [PMID: 28891094 DOI: 10.1111/nyas.13422] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/31/2017] [Accepted: 06/05/2017] [Indexed: 12/20/2022]
Abstract
The tumor suppressor phosphatase and tensin homologue (PTEN) has phosphatase activity, with phosphatidylinositol (3,4,5)-trisphosphate (PIP3), a product of phosphatidylinositol 3-kinase (PI3K), as one of the principal substrates. PTEN is a negative regulator of the Akt pathway, which plays a fundamental role in controlling cell growth, survival, and proliferation. Loss of PTEN function has been observed in many different types of cancer. Functional inactivation of PTEN as a consequence of germ-line mutations or promoter hypermethylation predisposes individuals to malignancies. PTEN undergoes posttranslational modifications, such as oxidation, acetylation, phosphorylation, SUMOylation, and ubiquitination, which influence its catalytic activity, interactions with other proteins, and subcellular localization. Cellular redox status is crucial for posttranslational modification of PTEN and its functional consequences. Oxidative stress and inflammation are major causes of loss of PTEN function. Pharmacologic or nutritional restoration of PTEN function is considered a reliable strategy in the management of PTEN-defective cancer. In this review, we highlight natural compounds, such as curcumin, indol-3 carbinol, and omega-3 fatty acids, that have the potential to restore or potentiate PTEN expression/activity, thereby suppressing cancer cell proliferation, survival, and resistance to chemotherapeutic agents.
Collapse
Affiliation(s)
- Do-Hee Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jinyoung Suh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul, South Korea
| |
Collapse
|
158
|
Liu T, Chen X, Li T, Li X, Lyu Y, Fan X, Zhang P, Zeng W. Histone methyltransferase SETDB1 maintains survival of mouse spermatogonial stem/progenitor cells via PTEN/AKT/FOXO1 pathway. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:1094-1102. [DOI: 10.1016/j.bbagrm.2017.08.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/28/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
|
159
|
Hauser DN, Mamais A, Conti MM, Primiani CT, Kumaran R, Dillman AA, Langston RG, Beilina A, Garcia JH, Diaz-Ruiz A, Bernier M, Fiesel FC, Hou X, Springer W, Li Y, de Cabo R, Cookson MR. Hexokinases link DJ-1 to the PINK1/parkin pathway. Mol Neurodegener 2017; 12:70. [PMID: 28962651 PMCID: PMC5622528 DOI: 10.1186/s13024-017-0212-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 09/19/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Early onset Parkinson's disease is caused by variants in PINK1, parkin, and DJ-1. PINK1 and parkin operate in pathways that preserve mitochondrial integrity, but the function of DJ-1 and how it relates to PINK1 and parkin is poorly understood. METHODS A series of unbiased high-content screens were used to analyze changes at the protein, RNA, and metabolite level in rodent brains lacking DJ-1. Results were validated using targeted approaches, and cellular assays were performed to probe the mechanisms involved. RESULTS We find that in both rat and mouse brains, DJ-1 knockout results in an age-dependent accumulation of hexokinase 1 in the cytosol, away from its usual location at the mitochondria, with subsequent activation of the polyol pathway of glucose metabolism in vivo. Both in the brain and in cultured cells, DJ-1 deficiency is associated with accumulation of the phosphatase PTEN that antagonizes the kinase AKT. In cells, addition of an inhibitor of AKT (MK2206) or addition of a peptide to dissociate association of hexokinases from mitochondria both inhibit the PINK1/parkin pathway, which works to maintain mitochondrial integrity. CONCLUSION Hexokinases are an important link between three major genetic causes of early onset Parkinson's disease. Because aging is associated with deregulated nutrient sensing, these results help explain why DJ-1 is associated with age-dependent disease.
Collapse
Affiliation(s)
- David N. Hauser
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Adamantios Mamais
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Melissa M. Conti
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Christopher T. Primiani
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Ravindran Kumaran
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Allissa A. Dillman
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Rebekah G. Langston
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Alexandra Beilina
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| | - Joseph H. Garcia
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Alberto Diaz-Ruiz
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Fabienne C. Fiesel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL USA
| | - Xu Hou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL USA
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Building 35, Room 1A116, 35 Convent Drive, MSC 3707, Bethesda, MD 20892-3707 USA
| |
Collapse
|
160
|
PTEN, a negative regulator of PI3K/Akt signaling, sustains brain stem cardiovascular regulation during mevinphos intoxication. Neuropharmacology 2017; 123:175-185. [DOI: 10.1016/j.neuropharm.2017.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/03/2017] [Accepted: 06/06/2017] [Indexed: 01/06/2023]
|
161
|
Khan I, Ansari IA, Singh P, Dass J FP. Prediction of functionally significant single nucleotide polymorphisms in PTEN tumor suppressor gene: An in silico approach. Biotechnol Appl Biochem 2017; 64:657-666. [PMID: 26800850 DOI: 10.1002/bab.1483] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 01/16/2016] [Indexed: 11/06/2022]
Abstract
The phosphatase and tensin homolog (PTEN) gene plays a crucial role in signal transduction by negatively regulating the PI3K signaling pathway. It is the most frequent mutated gene in many human-related cancers. Considering its critical role, a functional analysis of missense mutations of PTEN gene was undertaken in this study. Thirty five nonsynonymous single nucleotide polymorphisms (nsSNPs) within the coding region of the PTEN gene were selected for our in silico investigation, and five nsSNPs (G129E, C124R, D252G, H61D, and R130G) were found to be deleterious based on combinatorial predictions of different computational tools. Moreover, molecular dynamics (MD) simulation was performed to investigate the conformational variation between native and all the five mutant PTEN proteins having predicted deleterious nsSNPs. The results of MD simulation of all mutant models illustrated variation in structural attributes such as root-mean-square deviation, root-mean-square fluctuation, radius of gyration, and total energy; which depicts the structural stability of PTEN protein. Furthermore, mutant PTEN protein structures also showed a significant variation in the solvent accessible surface area and hydrogen bond frequencies from the native PTEN structure. In conclusion, results of this study have established the deleterious effect of the all the five predicted nsSNPs on the PTEN protein structure. Thus, results of the current study can pave a new platform to sort out nsSNPs that can be undertaken for the confirmation of their phenotype and their correlation with diseased status in case of control studies.
Collapse
Affiliation(s)
- Imran Khan
- Department of Biosciences, Integral University, Lucknow, India
| | - Irfan A Ansari
- Department of Biosciences, Integral University, Lucknow, India
| | - Pratichi Singh
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Febin Prabhu Dass J
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| |
Collapse
|
162
|
Malaney P, Uversky VN, Davé V. PTEN proteoforms in biology and disease. Cell Mol Life Sci 2017; 74:2783-2794. [PMID: 28289760 PMCID: PMC11107534 DOI: 10.1007/s00018-017-2500-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 01/30/2023]
Abstract
Proteoforms are specific molecular forms of protein products arising from a single gene that possess different structures and different functions. Therefore, a single gene can produce a large repertoire of proteoforms by means of allelic variations (mutations, indels, SNPs), alternative splicing and other pre-translational mechanisms, post-translational modifications (PTMs), conformational dynamics, and functioning. Resulting proteoforms that have different sizes, alternative splicing patterns, sets of post-translational modifications, protein-protein interactions, and protein-ligand interactions, might dramatically increase the functionality of the encoded protein. Herein, we have interrogated the tumor suppressor PTEN for its proteoforms and find that this protein exists in multiple forms with distinct functions and sub-cellular localizations. Furthermore, the levels of each PTEN proteoform in a given cell may affect its biological function. Indeed, the paradigm of the continuum model of tumor suppression by PTEN can be better explained by the presence of a continuum of PTEN proteoforms, diversity, and levels of which are associated with pathological outcomes than simply by the different roles of mutations in the PTEN gene. Consequently, understanding the mechanisms underlying the dysregulation of PTEN proteoforms by several genomic and non-genomic mechanisms in cancer and other diseases is imperative. We have identified different PTEN proteoforms, which control various aspects of cellular function and grouped them into three categories of intrinsic, function-induced, and inducible proteoforms. A special emphasis is given to the inducible PTEN proteoforms that are produced due to alternative translational initiation. The novel finding that PTEN forms dimers with biological implications supports the notion that PTEN proteoform-proteoform interactions may play hitherto unknown roles in cellular homeostasis and in pathogenic settings, including cancer. These PTEN proteoforms with unique properties and functionalities offer potential novel therapeutic opportunities in the treatment of various cancers and other diseases.
Collapse
Affiliation(s)
- Prerna Malaney
- Department of Pathology and Cell Biology, Morsani College of Medicine, MDC 64, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, 33612, USA
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave., Saint Petersburg, Russia, 194064
| | - Vrushank Davé
- Department of Pathology and Cell Biology, Morsani College of Medicine, MDC 64, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
- Department of Oncological Sciences, Morsani College of Medicine, University of South Florida, Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
163
|
Sun X, Kellner M, Desai AA, Wang T, Lu Q, Kangath A, Qu N, Klinger C, Fratz S, Yuan JXJ, Jacobson JR, Garcia JGN, Rafikov R, Fineman JR, Black SM. Asymmetric Dimethylarginine Stimulates Akt1 Phosphorylation via Heat Shock Protein 70-Facilitated Carboxyl-Terminal Modulator Protein Degradation in Pulmonary Arterial Endothelial Cells. Am J Respir Cell Mol Biol 2017; 55:275-87. [PMID: 26959555 DOI: 10.1165/rcmb.2015-0185oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) induces the mitochondrial translocation of endothelial nitric oxide synthase (eNOS) through the nitration-mediated activation of Akt1. However, it is recognized that the activation of Akt1 requires phosphorylation events at threonine (T) 308 and serine (S) 473. Thus, the current study was performed to elucidate the potential effect of ADMA on Akt1 phosphorylation and the mechanisms that are involved. Exposure of pulmonary arterial endothelial cells to ADMA enhanced Akt1 phosphorylation at both threonine 308 and Ser473 without altering Akt1 protein levels, phosphatase and tensin homolog activity, or membrane Akt1 levels. Heat shock protein (Hsp) 90 plays a pivotal role in maintaining Akt1 activity, and our results demonstrate that ADMA decreased Hsp90-Akt1 interactions, but, surprisingly, overexpression of a dominant-negative Hsp90 mutant increased Akt1 phosphorylation. ADMA exposure or overexpression of dominant-negative Hsp90 increased Hsp70 levels, and depletion of Hsp70 abolished ADMA-induced Akt1 phosphorylation. ADMA decreased the interaction of Akt1 with its endogenous inhibitor, carboxyl-terminal modulator protein (CTMP). This was mediated by the proteasomal-dependent degradation of CTMP. The overexpression of CTMP attenuated ADMA-induced Akt1 phosphorylation at Ser473, eNOS phosphorylation at Ser617, and eNOS mitochondrial translocation. Finally, we found that the mitochondrial translocation of eNOS in our lamb model of pulmonary hypertension is associated with increased Akt1 and eNOS phosphorylation and reduced Akt1-CTMP protein interactions. In conclusion, our data suggest that CTMP is directly involved in ADMA-induced Akt1 phosphorylation in vitro and in vivo, and that increasing CTMP levels may be an avenue to treat pulmonary hypertension.
Collapse
Affiliation(s)
- Xutong Sun
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Manuela Kellner
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ankit A Desai
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ting Wang
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Qing Lu
- 2 Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia
| | - Archana Kangath
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ning Qu
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Christina Klinger
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Sohrab Fratz
- 3 Pediatric Cardiology and Congenital Heart Disease, German Heart Center at the Technical University of Munich, Munich, Germany
| | - Jason X-J Yuan
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Jeffrey R Jacobson
- 4 Department of Medicine, University of Illinois Chicago, Chicago, Illinois; and
| | - Joe G N Garcia
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Ruslan Rafikov
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| | - Jeffrey R Fineman
- 5 Department of Pediatrics and.,6 Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Stephen M Black
- 1 Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
164
|
Gowda C, Soliman M, Kapadia M, Ding Y, Payne K, Dovat S. Casein Kinase II (CK2), Glycogen Synthase Kinase-3 (GSK-3) and Ikaros mediated regulation of leukemia. Adv Biol Regul 2017. [PMID: 28623166 DOI: 10.1016/j.jbior.2017.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Signaling networks that regulate cellular proliferation often involve complex interactions between several signaling pathways. In this manuscript we review the crosstalk between the Casein Kinase II (CK2) and Glycogen Synthase Kinase-3 (GSK-3) pathways that plays a critical role in the regulation of cellular proliferation in leukemia. Both CK2 and GSK-3 are potential targets for anti-leukemia treatment. Previously published data suggest that CK2 and GSK-3 act synergistically to promote the phosphatidylinositol-3 kinase (PI3K) pathway via phosphorylation of PTEN. More recent data demonstrate another mechanism through which CK2 promotes the PI3K pathway - via transcriptional regulation of PI3K pathway genes by the newly-discovered CK2-Ikaros axis. Together, these data suggest that the CK2 and GSK-3 pathways regulate AKT/PI3K signaling in leukemia via two complementary mechanisms: a) direct phosphorylation of PTEN and b) transcriptional regulation of PI3K-promoting genes. Functional interactions between CK2, Ikaros and GSK3 define a novel signaling network that regulates proliferation of leukemia cells. This regulatory network involves both direct posttranslational modifications (by CK and GSK-3) and transcriptional regulation (via CK2-mediated phosphorylation of Ikaros). This information provides a basis for the development of targeted therapy for leukemia.
Collapse
Affiliation(s)
- Chandrika Gowda
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Mario Soliman
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Malika Kapadia
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Yali Ding
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Kimberly Payne
- Department of Anatomy, Loma Linda University, Loma Linda, CA, USA.
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
165
|
Ohkawa K, Asakura T, Tsukada Y, Matsuura T. Antibody to human α-fetoprotein inhibits cell growth of human hepatocellular carcinoma cells by resuscitating the PTEN molecule: in vitro experiments. Int J Oncol 2017; 50:2180-2190. [PMID: 28498467 DOI: 10.3892/ijo.2017.3982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
It has been proposed that α-fetoprotein (AFP) is a new member of the intracellular signaling molecule family of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway via interaction with the phosphatase and tensin homolog (PTEN). In this study, the effects of anti-human AFP antibody on the functions of PTEN were examined using an AFP-producing human hepatoma cell line. The antibody caused significant inhibition of cell growth, compared to a normal IgG control, with the accumulation of intracellular immune complexes followed by significant reduction of cytosolic functional AFP. Decrease in the amount of AKT phosphorylated on serine (S) 473 indicated that PI3K/AKT signaling was suppressed in the cells. S380-phosphorylated PTEN increased markedly by the second day after antibody treatment, with slight but significant increase in the PTEN protein level. Since phosphorylation at S380 is critical for PTEN stability, the increase in S380-phosphorylated PTEN indicated maintenance of the number of PTEN molecules and the related potential to control PI3K/AKT signaling. p53 protein (P53) significantly, but slightly increased during antibody treatment, because PTEN expression increased the stability and function of P53 via both molecular interactions. P53 phosphorylated at S20 or at S392 dramatically increased, suggesting an increase in the stability, accumulation and activation of P53. Glucose transporter 1 (GLUT1) increased immediately after antibody treatment, pointing to a deficiency of glucose in the cells. Immunofluorescence cytology revealed that antibody-treatment re-distributed GLUT1 molecules throughout the cytoplasm with a reduction of their patchy localization on the cell surface. This suggested that translocation of GLUT1 depends on the PI3K/AKT pathway, in particular on PTEN expression. Antibody therapy targeted at AFP-producing tumor cells showed an inhibitory effect on the PI3K/AKT pathway via the liberation, restoration and functional stabilization of PTEN. PTEN simultaneously induced both P53 activation and intracellular translocation of GLUT1, since these are closely associated with PTEN.
Collapse
Affiliation(s)
- Kiyoshi Ohkawa
- Stable Isotope Medical Applications Laboratory, Research Center for Medical Science, Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Tadashi Asakura
- Radioisotope Research Facilities, Research Center for Medical Science, Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| | - Yutaka Tsukada
- Hachioji Laboratory, SRL Inc., Komiya-cho, Hachioji, Tokyo 192-8535, Japan
| | - Tomokazu Matsuura
- Department of Laboratory Medicine, Jikei University School of Medicine, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
166
|
Sullenberger C, Piqué D, Ogata Y, Mensa-Wilmot K. AEE788 Inhibits Basal Body Assembly and Blocks DNA Replication in the African Trypanosome. Mol Pharmacol 2017; 91:482-498. [PMID: 28246189 PMCID: PMC5399642 DOI: 10.1124/mol.116.106906] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022] Open
Abstract
Trypanosoma brucei causes human African trypanosomiasis (HAT). The pyrrolopyrimidine AEE788 (a hit for anti-HAT drug discovery) associates with three trypanosome protein kinases. Herein we delineate the effects of AEE788 on T. brucei using chemical biology strategies. AEE788 treatment inhibits DNA replication in the kinetoplast (mitochondrial nucleoid) and nucleus. In addition, AEE788 blocks duplication of the basal body and the bilobe without affecting mitosis. Thus, AEE788 prevents entry into the S-phase of the cell division cycle. To study the kinetics of early events in trypanosome division, we employed an "AEE788 block and release" protocol to stage entry into the S-phase. A time-course of DNA synthesis (nuclear and kinetoplast DNA), duplication of organelles (basal body, bilobe, kinetoplast, nucleus), and cytokinesis was obtained. Unexpected findings include the following: 1) basal body and bilobe duplication are concurrent; 2) maturation of probasal bodies, marked by TbRP2 recruitment, is coupled with nascent basal body assembly, monitored by localization of TbSAS6 at newly forming basal bodies; and 3) kinetoplast division is observed in G2 after completion of nuclear DNA synthesis. Prolonged exposure of trypanosomes to AEE788 inhibited transferrin endocytosis, altered cell morphology, and decreased cell viability. To discover putative effectors for the pleiotropic effects of AEE788, proteome-wide changes in protein phosphorylation induced by the drug were determined. Putative effectors include an SR protein kinase, bilobe proteins, TbSAS4, TbRP2, and BILBO-1. Loss of function of one or more of these effectors can, from published literature, explain the polypharmacology of AEE788 on trypanosome biology.
Collapse
Affiliation(s)
- Catherine Sullenberger
- Department of Cellular Biology, and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia (C.S., D.P., K.M.-W.); and the Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington (Y.O.)
| | - Daniel Piqué
- Department of Cellular Biology, and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia (C.S., D.P., K.M.-W.); and the Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington (Y.O.)
| | - Yuko Ogata
- Department of Cellular Biology, and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia (C.S., D.P., K.M.-W.); and the Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington (Y.O.)
| | - Kojo Mensa-Wilmot
- Department of Cellular Biology, and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia (C.S., D.P., K.M.-W.); and the Proteomics Facility, Fred Hutchinson Cancer Research Center, Seattle, Washington (Y.O.)
| |
Collapse
|
167
|
Du Z, Dong C, Ren J. A study of the dynamics of PTEN proteins in living cells using in vivo fluorescence correlation spectroscopy. Methods Appl Fluoresc 2017; 5:024008. [PMID: 28373603 DOI: 10.1088/2050-6120/aa6b07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PTEN (phosphatase and tensin homolog on chromosome 10) is one of the most important tumor-suppressor proteins, which plays a key role in negative regulation of the PI3K/AKT pathway, and governs many cellular processes including growth, proliferation, survival and migration. The dynamics of PTEN proteins in single living cells is as yet unclear owing to a shortage of suitable in vivo approaches. Here, we report a single-molecule method for in vivo study of the dynamics of PTEN proteins in living cells using fluorescence correlation spectroscopy (FCS). First, we established a monoclonal H1299 stable cell line expressing enhanced green fluorescent protein (EGFP) and PTEN (EGFP-PTEN) fusion proteins; we then developed an in vivo FCS method to study the dynamics of EGFP-PTEN both in the nucleus and the cytoplasm. We investigated the diffusion behaviors of EGFP and EGFP-PTEN in solution, nucleus and cytosol, and observed that the motion of PTEN in living cells was restricted compared with EGFP. Finally, we investigated the protein dynamics in living cells under oxidative stress stimulation and a cellular ATP depletion treatment. Under oxidative stress stimulation, the EGFP-PTEN concentration increased in the nucleus, but slightly decreased in the cytoplasm. The diffusion coefficient and alpha value of EGFP-PTEN reduced significantly both in the nucleus and cytoplasm; the significantly decreased alpha parameter indicates a more restricted Brownian diffusion behavior. Under the cellular ATP depletion treatment, the concentration of EGFP-PTEN remained unchanged in the nucleus and decreased significantly in cytosol. The diffusion coefficient of EGFP-PTEN decreased significantly in cytosol, but showed no significant change in the nucleus; the alpha value decreased significantly in both the nucleus and cytoplasm. These results suggest that the concentration and mobility of PTEN in the nucleus and cytoplasm can be regulated by stimulation methods. Our approach provides a unique method for real-time monitoring of protein dynamics in different subcellular compartments under different stimulation treatments.
Collapse
Affiliation(s)
- Zhixue Du
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | | | | |
Collapse
|
168
|
KIF7 attenuates prostate tumor growth through LKB1-mediated AKT inhibition. Oncotarget 2017; 8:54558-54571. [PMID: 28903364 PMCID: PMC5589603 DOI: 10.18632/oncotarget.17421] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/10/2017] [Indexed: 12/26/2022] Open
Abstract
This study investigated kinesin family member 7 (KIF7) expression and function in prostate cancer (PCa). Our results showed that KIF7 was significantly downregulated in PCa, compared with normal, benign prostatic hyperplasia and prostate intraepithelial neoplasia tissues, partially through promoter hypermethylation. We further investigated the effects of KIF7 coiled coil (CC) domain and motor domain (MD) on PCa development in vitro and in vivo. Our results showed that KIF7-CC but not KIF7-MD significantly attenuated proliferation and colony formation, impeded migration and invasion, induced apoptosis and sensitized PCa cells to paclitaxel. Further analysis revealed that KIF7-CC enhanced LKB1 expression and phosphorylation at Ser428, which induced PTEN phosphorylation at Ser380/Thr382/383 and consequently blocked AKT phosphorylation at Ser473. Downregulation of LKB1 significantly attenuated the suppressive effects of KIF7-CC on cell proliferation, colony formation and AKT phosphorylation. Furthermore, our in vivo studies showed that KIF7-CC reduced prostate tumorigenesis in cell-derived xenografts. Downregulation of LKB1 abrogated the anti-tumor effects of KIF7-CC in these xenografts. Taken together, these findings provide the first evidence to support the role of KIF7 as a negative regulator that inhibits PCa development partially through LKB1-mediated AKT inhibition.
Collapse
|
169
|
TGF-β induces phosphorylation of phosphatase and tensin homolog: implications for fibrosis of the trabecular meshwork tissue in glaucoma. Sci Rep 2017; 7:812. [PMID: 28400560 PMCID: PMC5429747 DOI: 10.1038/s41598-017-00845-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/16/2017] [Indexed: 01/06/2023] Open
Abstract
Fundamental cell signaling mechanisms that regulate dynamic remodeling of the extracellular matrix (ECM) in mechanically loaded tissues are not yet clearly understood. Trabecular meshwork (TM) tissue in the eye is under constant mechanical stress and continuous remodeling of ECM is crucial to maintain normal aqueous humor drainage and intraocular pressure (IOP). However, excessive ECM remodeling can cause fibrosis of the TM as in primary open-angle glaucoma (POAG) patients, and is characterized by increased resistance to aqueous humor drainage, elevated IOP, optic nerve degeneration and blindness. Increased levels of active transforming growth factor-β2 (TGF-β2) in the aqueous humor is the main cause of fibrosis of TM in POAG patients. Herein, we report a novel finding that, in TM cells, TGF-β-induced increase in collagen expression is associated with phosphorylation of phosphatase and tensin homolog (PTEN) at residues Ser380/Thr382/383. Exogenous overexpression of a mutated form of PTEN with enhanced phosphatase activity prevented the TGF-β-induced collagen expression by TM cells. We propose that rapid alteration of PTEN activity through changes in its phosphorylation status could uniquely regulate the continuous remodeling of ECM in the normal TM. Modulating PTEN activity may have high therapeutic potential to alleviating the fibrosis of TM in POAG patients.
Collapse
|
170
|
Zhang J, Storey KB. Insect cold hardiness: the role of mitogen-activated protein kinase and Akt signalling in freeze avoiding larvae of the goldenrod gall moth, Epiblema scudderiana. INSECT MOLECULAR BIOLOGY 2017; 26:181-189. [PMID: 27880024 DOI: 10.1111/imb.12283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Larvae of the goldenrod gall moth, Epiblema scudderiana, use the freeze avoidance strategy of cold hardiness to survive the winter. Here we report that protein kinase-dependent signal transduction featuring mitogen-activated protein kinase (MAPK) signalling cascades (extracellular signal regulated kinase, c-jun N-terminal kinase and p38 MAPK pathways) and the Akt (also known as protein kinase B, or PKB) pathway could be integral parts of the development of cold hardiness by E. scudderiana. We used Luminex technology to assess the protein levels and phosphorylation status of key components and downstream targets of those pathways in larvae in response to low temperature acclimation. The data showed that MAPK pathways (both total protein and phosphorylated MAPK targets) were inhibited after 5°C acclimation, but not -15°C exposure, as compared with the 15°C control group. However, total heat shock protein 27 (HSP27) levels increased dramatically by ∼12-fold in the -15°C acclimated insects. Elevated HSP27 may facilitate anti-apoptotic mechanisms in an Akt-dependent fashion. By contrast, both 5 and -15°C acclimation produced signs of Akt pathway activation. In particular, the inhibitor phosphorylated Glycogen Synthase Kinase 3a (p-GSK3) levels remained high in cold-exposed larvae. Additionally, activation of the Akt pathway might also facilitate inhibition of apoptosis independently of GSK3. Overall, the current study indicates that both MAPK and Akt signal transduction may play essential roles in freeze avoidance by E. scudderiana.
Collapse
Affiliation(s)
- J Zhang
- Institute of Biochemistry and Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - K B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
171
|
Brandmaier A, Hou SQ, Demaria S, Formenti SC, Shen WH. PTEN at the interface of immune tolerance and tumor suppression. ACTA ACUST UNITED AC 2017. [PMID: 29527223 DOI: 10.1007/s11515-017-1443-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND PTEN is well known to function as a tumor suppressor that antagonizes oncogenic signaling and maintains genomic stability. The PTEN gene is frequently deleted or mutated in human cancers and the wide cancer spectrum associated with PTEN deficiency has been recapitulated in a variety of mouse models of Pten deletion or mutation. Pten mutations are highly penetrant in causing various types of spontaneous tumors that often exhibit resistance to anticancer therapies including immunotherapy. Recent studies demonstrate that PTEN also regulates immune functionality. OBJECTIVE To understand the multifaceted functions of PTEN as both a tumor suppressor and an immune regulator. METHODS This review will summarize the emerging knowledge of PTEN function in cancer immunoediting. In addition, the mechanisms underlying functional integration of various PTEN pathways in regulating cancer evolution and tumor immunity will be highlighted. RESULTS Recent preclinical and clinical studies revealed the essential role of PTEN in maintaining immune homeostasis, which significantly expands the repertoire of PTEN functions. Mechanistically, aberrant PTEN signaling alters the interplay between the immune system and tumors, leading to immunosuppression and tumor escape. CONCLUSION Rational design of personalized anti-cancer treatment requires mechanistic understanding of diverse PTEN signaling pathways in modulation of the crosstalk between tumor and immune cells.
Collapse
Affiliation(s)
- Andrew Brandmaier
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Sheng-Qi Hou
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Wen H Shen
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
172
|
Tan L, Chen X, Wang W, Zhang J, Li S, Zhao Y, Wang J, Luo A. Pharmacological inhibition of PTEN attenuates cognitive deficits caused by neonatal repeated exposures to isoflurane via inhibition of NR2B-mediated tau phosphorylation in rats. Neuropharmacology 2017; 114:135-145. [PMID: 27836791 DOI: 10.1016/j.neuropharm.2016.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/30/2016] [Accepted: 11/07/2016] [Indexed: 10/20/2022]
Abstract
Evidence has shown that children exposed to repeated anesthesia in early childhood display long-term cognitive disabilities. However, the underlying mechanisms remain largely unclear. Our previous study has indicated the involvement of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in isoflurane-induced decrease of self-renewal capacity in hippocampal neural precursor cells. Additionally, it is demonstrated by others that PTEN inhibition could protect against cognitive impairment via reduction of tau phosphorylation in the alzheimer's disease model. Therefore, in the present in vivo study, we aimed to examine the effects of PTEN inhibition on the cognitive dysfunction and tau hyperphosphorylation caused by neonatal repeated exposures to isoflurane. Our results showed that the neonatal repeated exposures to isoflurane resulted in the activation of PTEN in the hippocampus. The treatment of PTEN inhibitor BPV (pic) restored PSD-95 synthesis, and attenuated tau phosphorylation as well as the cognitive dysfunction caused by the repeated isoflurane exposures. In addition, BPV (pic) treatment reversed the activation of NR2B-containing NMDARs induced by repeated isoflurane exposures, while in turn, the antagonism of NR2B subunit with ifenprodil alleviated tau phosphorylation, indicating a possible role of NR2B as the downstream of PTEN in mediating tau phosphorylation in the neonatal rats repeatedly exposed to isoflurane. In conclusion, our results reveal a novel role of PTEN in mediating tau phosphorylation and cognitive deficits caused by neonatal repeated exposures to isoflurane, implying that targeting on PTEN may be a potential therapeutic approach for the anesthetic-related cognitive decline in the developing brain.
Collapse
Affiliation(s)
- Lei Tan
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Wei Wang
- Department of Anesthesiology, First Hospital of Xian Jiaotong University, Xi'an 710061, China
| | - Jianfang Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jintao Wang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
173
|
Andreani C, Bartolacci C, Wijnant K, Crinelli R, Bianchi M, Magnani M, Hysi A, Iezzi M, Amici A, Marchini C. Resveratrol fuels HER2 and ERα-positive breast cancer behaving as proteasome inhibitor. Aging (Albany NY) 2017; 9:508-523. [PMID: 28238967 PMCID: PMC5361678 DOI: 10.18632/aging.101175] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/11/2017] [Indexed: 11/25/2022]
Abstract
The phytoestrogen resveratrol has been reported to possess cancer chemo-preventive activity on the basis of its effects on tumor cell lines and xenograft or carcinogen-inducible in vivo models. Here we investigated the effects of resveratrol on spontaneous mammary carcinogenesis using Δ16HER2 mice as HER2+/ERα+ breast cancer model. Instead of inhibiting tumor growth, resveratrol treatment (0.0001% in drinking water; daily intake of 4μg/mouse) shortened tumor latency and enhanced tumor multiplicity in Δ16HER2 mice. This in vivo tumor-promoting effect of resveratrol was associated with up-regulation of Δ16HER2 and down-regulation of ERα protein levels and was recapitulated in vitro by murine (CAM6) and human (BT474) tumor cell lines. Our results demonstrate that resveratrol, acting as a proteasome inhibitor, leads to Δ16HER2 accumulation which favors the formation of Δ16HER2/HER3 heterodimers. The consequential activation of downstream mTORC1/p70S6K/4EBP1 pathway triggers cancer growth and proliferation. This study provides evidence that resveratrol mechanism of action (and hence its effects) depends on the intrinsic molecular properties of the cancer model under investigation, exerting a tumor-promoting effect in luminal B breast cancer subtype models.
Collapse
Affiliation(s)
- Cristina Andreani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, 62032, Italy
| | - Caterina Bartolacci
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, 62032, Italy
| | - Kathleen Wijnant
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, 62032, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, Section of Biochemistry and Molecular Biology, University of Urbino “Carlo Bo”, Urbino, 61029, Italy
| | - Marzia Bianchi
- Department of Biomolecular Sciences, Section of Biochemistry and Molecular Biology, University of Urbino “Carlo Bo”, Urbino, 61029, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, Section of Biochemistry and Molecular Biology, University of Urbino “Carlo Bo”, Urbino, 61029, Italy
| | - Albana Hysi
- Aging Research Centre, G. d'Annunzio University, Chieti, 66100, Italy
| | - Manuela Iezzi
- Aging Research Centre, G. d'Annunzio University, Chieti, 66100, Italy
| | - Augusto Amici
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, 62032, Italy
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, 62032, Italy
| |
Collapse
|
174
|
Regulation of PI3K effector signalling in cancer by the phosphoinositide phosphatases. Biosci Rep 2017; 37:BSR20160432. [PMID: 28082369 PMCID: PMC5301276 DOI: 10.1042/bsr20160432] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/24/2022] Open
Abstract
Class I phosphoinositide 3-kinase (PI3K) generates phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) at the plasma membrane in response to growth factors, activating a signalling cascade that regulates many cellular functions including cell growth, proliferation, survival, migration and metabolism. The PI3K pathway is commonly dysregulated in human cancer, and drives tumorigenesis by promoting aberrant cell growth and transformation. PtdIns(3,4,5)P3 facilitates the activation of many pleckstrin homology (PH) domain-containing proteins including the serine/threonine kinase AKT. There are three AKT isoforms that are frequently hyperactivated in cancer through mutation, amplification or dysregulation of upstream regulatory proteins. AKT isoforms have converging and opposing functions in tumorigenesis. PtdIns(3,4,5)P3 signalling is degraded and terminated by phosphoinositide phosphatases such as phosphatase and tensin homologue (PTEN), proline-rich inositol polyphosphate 5-phosphatase (PIPP) (INPP5J) and inositol polyphosphate 4-phosphatase type II (INPP4B). PtdIns(3,4,5)P3 is rapidly hydrolysed by PIPP to generate phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2), which is further hydrolysed by INPP4B to form phosphatidylinositol 3-phosphate (PtdIns3P). PtdIns(3,4)P2 and PtdIns3P are also important signalling molecules; PtdIns(3,4)P2 together with PtdIns(3,4,5)P3 are required for maximal AKT activation and PtdIns3P activates PI3K-dependent serum and glucocorticoid-regulated kinase (SGK3) signalling. Loss of Pten, Pipp or Inpp4b expression or function promotes tumour growth in murine cancer models through enhanced AKT isoform-specific signalling. INPP4B inhibits PtdIns(3,4)P2-mediated AKT activation in breast and prostate cancer; however, INPP4B expression is increased in acute myeloid leukaemia (AML), melanoma and colon cancer where it paradoxically promotes cell proliferation, transformation and/or drug resistance. This review will discuss how PTEN, PIPP and INPP4B distinctly regulate PtdIns(3,4,5)P3 signalling downstream of PI3K and how dysregulation of these phosphatases affects cancer outcomes.
Collapse
|
175
|
Lai X, Friedman A. Exosomal microRNA concentrations in colorectal cancer: A mathematical model. J Theor Biol 2017; 415:70-83. [DOI: 10.1016/j.jtbi.2016.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/06/2016] [Accepted: 12/10/2016] [Indexed: 12/19/2022]
|
176
|
Vaquero J, Nguyen Ho-Bouldoires TH, Clapéron A, Fouassier L. Role of the PDZ-scaffold protein NHERF1/EBP50 in cancer biology: from signaling regulation to clinical relevance. Oncogene 2017; 36:3067-3079. [PMID: 28068322 DOI: 10.1038/onc.2016.462] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The transmission of cellular information requires fine and subtle regulation of proteins that need to interact in a coordinated and specific way to form efficient signaling networks. The spatial and temporal coordination relies on scaffold proteins. Thanks to protein interaction domains such as PDZ domains, scaffold proteins organize multiprotein complexes enabling the proper transmission of cellular information through intracellular networks. NHERF1/EBP50 is a PDZ-scaffold protein that was initially identified as an organizer and regulator of transporters and channels at the apical side of epithelia through actin-binding ezrin-moesin-radixin proteins. Since, NHERF1/EBP50 has emerged as a major regulator of cancer signaling network by assembling cancer-related proteins. The PDZ-scaffold EBP50 carries either anti-tumor or pro-tumor functions, two antinomic functions dictated by EBP50 expression or subcellular localization. The dual function of NHERF1/EBP50 encompasses the regulation of several major signaling pathways engaged in cancer, including the receptor tyrosine kinases PDGFR and EGFR, PI3K/PTEN/AKT and Wnt-β-catenin pathways.
Collapse
Affiliation(s)
- J Vaquero
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - T H Nguyen Ho-Bouldoires
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,FONDATION ARC, Villejuif, France
| | - A Clapéron
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - L Fouassier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
177
|
Lai X, Friedman A. Exosomal miRs in Lung Cancer: A Mathematical Model. PLoS One 2016; 11:e0167706. [PMID: 28002496 PMCID: PMC5176278 DOI: 10.1371/journal.pone.0167706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/18/2016] [Indexed: 01/11/2023] Open
Abstract
Lung cancer, primarily non-small-cell lung cancer (NSCLC), is the leading cause of cancer deaths in the United States and worldwide. While early detection significantly improves five-year survival, there are no reliable diagnostic tools for early detection. Several exosomal microRNAs (miRs) are overexpressed in NSCLC, and have been suggested as potential biomarkers for early detection. The present paper develops a mathematical model for early stage of NSCLC with emphasis on the role of the three highest overexpressed miRs, namely miR-21, miR-205 and miR-155. Simulations of the model provide quantitative relationships between the tumor volume and the total mass of each of the above miRs in the tumor. Because of the positive correlation between these miRs in the tumor tissue and in the blood, the results of the paper may be viewed as a first step toward establishing a combination of miRs 21, 205, 155 and possibly other miRs as serum biomarkers for early detection of NSCLC.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, P. R. China
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
178
|
Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD-L1/2 Pathway from Discovery to Clinical Implementation. Front Immunol 2016; 7:550. [PMID: 28018338 PMCID: PMC5149523 DOI: 10.3389/fimmu.2016.00550] [Citation(s) in RCA: 432] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/16/2016] [Indexed: 12/14/2022] Open
Abstract
The immune system maintains a critically organized network to defend against foreign particles, while evading self-reactivity simultaneously. T lymphocytes function as effectors and play an important regulatory role to orchestrate the immune signals. Although central tolerance mechanism results in the removal of the most of the autoreactive T cells during thymic selection, a fraction of self-reactive lymphocytes escapes to the periphery and pose a threat to cause autoimmunity. The immune system evolved various mechanisms to constrain such autoreactive T cells and maintain peripheral tolerance, including T cell anergy, deletion, and suppression by regulatory T cells (TRegs). These effects are regulated by a complex network of stimulatory and inhibitory receptors expressed on T cells and their ligands, which deliver cell-to-cell signals that dictate the outcome of T cell encountering with cognate antigens. Among the inhibitory immune mediators, the pathway consisting of the programed cell death 1 (PD-1) receptor (CD279) and its ligands PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273) plays an important role in the induction and maintenance of peripheral tolerance and for the maintenance of the stability and the integrity of T cells. However, the PD-1:PD-L1/L2 pathway also mediates potent inhibitory signals to hinder the proliferation and function of T effector cells and have inimical effects on antiviral and antitumor immunity. Therapeutic targeting of this pathway has resulted in successful enhancement of T cell immunity against viral pathogens and tumors. Here, we will provide a brief overview on the properties of the components of the PD-1 pathway, the signaling events regulated by PD-1 engagement, and their consequences on the function of T effector cells.
Collapse
Affiliation(s)
- Kankana Bardhan
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Theodora Anagnostou
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Vassiliki A. Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
179
|
Ola R, Dubrac A, Han J, Zhang F, Fang JS, Larrivée B, Lee M, Urarte AA, Kraehling JR, Genet G, Hirschi KK, Sessa WC, Canals FV, Graupera M, Yan M, Young LH, Oh PS, Eichmann A. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun 2016; 7:13650. [PMID: 27897192 PMCID: PMC5141347 DOI: 10.1038/ncomms13650] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/20/2016] [Indexed: 12/26/2022] Open
Abstract
Activin receptor-like kinase 1 (ALK1) is an endothelial serine-threonine kinase receptor for bone morphogenetic proteins (BMPs) 9 and 10. Inactivating mutations in the ALK1 gene cause hereditary haemorrhagic telangiectasia type 2 (HHT2), a disabling disease characterized by excessive angiogenesis with arteriovenous malformations (AVMs). Here we show that inducible, endothelial-specific homozygous Alk1 inactivation and BMP9/10 ligand blockade both lead to AVM formation in postnatal retinal vessels and internal organs including the gastrointestinal (GI) tract in mice. VEGF and PI3K/AKT signalling are increased on Alk1 deletion and BMP9/10 ligand blockade. Genetic deletion of the signal-transducing Vegfr2 receptor prevents excessive angiogenesis but does not fully revert AVM formation. In contrast, pharmacological PI3K inhibition efficiently prevents AVM formation and reverts established AVMs. Thus, Alk1 deletion leads to increased endothelial PI3K pathway activation that may be a novel target for the treatment of vascular lesions in HHT2.
Collapse
Affiliation(s)
- Roxana Ola
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Alexandre Dubrac
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jinah Han
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Feng Zhang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jennifer S. Fang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Bruno Larrivée
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Monica Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Ana A. Urarte
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Jan R. Kraehling
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Gael Genet
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Karen K. Hirschi
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - William C. Sessa
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Francesc V. Canals
- Translation Research Laboratory, Catalan Institute of Oncology, Idibell, Barcelona 08908, Spain
| | - Mariona Graupera
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Minhong Yan
- Molecular Oncology, Genentech, Inc., South San Francisco, California 94080-4990, USA
| | - Lawrence H. Young
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Paul S. Oh
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, PO Box 100274, Gainesville, Florida 32610, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Inserm U970, Paris Cardiovascular Research Center, Paris 75015, France
| |
Collapse
|
180
|
Takiar V, Ip CKM, Gao M, Mills GB, Cheung LWT. Neomorphic mutations create therapeutic challenges in cancer. Oncogene 2016; 36:1607-1618. [PMID: 27841866 DOI: 10.1038/onc.2016.312] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/24/2016] [Accepted: 07/17/2016] [Indexed: 02/07/2023]
Abstract
Oncogenesis is a pathologic process driven by genomic aberrations, including changes in nucleotide sequences. The majority of these mutational events fall into two broad categories: inactivation of tumor suppressor genes (hypomorph, antimorph or amorph) or activation of oncogenes (hypermorph). The recent surge in genome sequence data and functional genomics research has ushered in the discovery of aberrations in a third category: gain-of-novel-function mutation (neomorph). These neomorphic mutations, which can be found in both tumor suppressor genes and oncogenes, produce proteins with entirely different functions from their respective wild-type (WT) proteins and the other morphs. The unanticipated phenotypic outcomes elicited by neomorphic mutations imply that tumors with the neomorphic mutations may not respond to therapies designed to target the WT protein. Therefore, understanding the functional activities of each genomic aberration to be targeted is crucial in devising effective treatment strategies that will benefit specific cancer patients.
Collapse
Affiliation(s)
- V Takiar
- Departments of Radiation Oncology and Cancer Biology, University of Cincinnati College of Medicine, UC Barrett Cancer Center, OH, USA
| | - C K M Ip
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Gao
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L W T Cheung
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| |
Collapse
|
181
|
Affiliation(s)
- Vassiliki A Boussiotis
- From the Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston
| |
Collapse
|
182
|
Paranjpe S, Bowen WC, Mars WM, Orr A, Haynes MM, DeFrances MC, Liu S, Tseng GC, Tsagianni A, Michalopoulos GK. Combined systemic elimination of MET and epidermal growth factor receptor signaling completely abolishes liver regeneration and leads to liver decompensation. Hepatology 2016; 64:1711-1724. [PMID: 27397846 PMCID: PMC5074871 DOI: 10.1002/hep.28721] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED Receptor tyrosine kinases MET and epidermal growth factor receptor (EGFR) are critically involved in initiation of liver regeneration. Other cytokines and signaling molecules also participate in the early part of the process. Regeneration employs effective redundancy schemes to compensate for the missing signals. Elimination of any single extracellular signaling pathway only delays but does not abolish the process. Our present study, however, shows that combined systemic elimination of MET and EGFR signaling (MET knockout + EGFR-inhibited mice) abolishes liver regeneration, prevents restoration of liver mass, and leads to liver decompensation. MET knockout or simply EGFR-inhibited mice had distinct and signaling-specific alterations in Ser/Thr phosphorylation of mammalian target of rapamycin, AKT, extracellular signal-regulated kinases 1/2, phosphatase and tensin homolog, adenosine monophosphate-activated protein kinase α, etc. In the combined MET and EGFR signaling elimination of MET knockout + EGFR-inhibited mice, however, alterations dependent on either MET or EGFR combined to create shutdown of many programs vital to hepatocytes. These included decrease in expression of enzymes related to fatty acid metabolism, urea cycle, cell replication, and mitochondrial functions and increase in expression of glycolysis enzymes. There was, however, increased expression of genes of plasma proteins. Hepatocyte average volume decreased to 35% of control, with a proportional decrease in the dimensions of the hepatic lobules. Mice died at 15-18 days after hepatectomy with ascites, increased plasma ammonia, and very small livers. CONCLUSION MET and EGFR separately control many nonoverlapping signaling endpoints, allowing for compensation when only one of the signals is blocked, though the combined elimination of the signals is not tolerated; the results provide critical new information on interactive MET and EGFR signaling and the contribution of their combined absence to regeneration arrest and liver decompensation. (Hepatology 2016;64:1711-1724).
Collapse
Affiliation(s)
- Shirish Paranjpe
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - William C Bowen
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Wendy M Mars
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Anne Orr
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Meagan M Haynes
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Marie C DeFrances
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Silvia Liu
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - George C Tseng
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Anastasia Tsagianni
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | |
Collapse
|
183
|
Xu X, Chen G, Wu L, Liu L. Association of genetic polymorphisms in PTEN and additional gene-gene interaction with risk of esophageal squamous cell carcinoma in Chinese Han population. Dis Esophagus 2016; 29:944-949. [PMID: 26541596 DOI: 10.1111/dote.12428] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study aims to investigate the association of five single nucleotide polymorphisms (SNPs) in the phosphatase and tensin homologue (PTEN) gene and additional role of gene-gene interaction with esophageal squamous cell carcinoma (ESCC), based on a Chinese case-control study. A total of 871 subjects (420 males and 451 females) were selected, including 425 ESCC cases and 446 controls. Five SNPs were selected for genotyping in the case-control study: rs2735343, rs555895, rs2299939, rs17431184 and rs701848. Logistic regression model was used to examine the association between five SNP and ESCC, and additional interaction among five SNP, odds ratio (OR) and 95% confident interval (95%CI) were calculated. All genotypes were distributed according to Hardy-Weinberg equilibrium in controls. The carriers of homozygous mutant of rs2735343 and rs701848 polymorphism revealed increased ESCC risk than those with wild-type homozygotes, and OR (95%CI) were 1.27 (1.09-2.08) and 1.45 (1.17-1.98), respectively. We also found a potential gene-gene interaction between rs2735343 and rs701848 (P = 0.0010), and a potential gene-gene interaction among all five SNP (P = 0.0107) after covariates adjustment. Subjects with TC or CC of rs2735343 and TC or CC of rs701848 genotype have highest ESCC risk, compared to subjects with TT of rs2735343 and TT of rs701848 genotype, OR (95% CI) was 2.76 (1.37-3.45) after covariates adjustment. The carriers of homozygous mutant of rs2735343 and rs701848 polymorphism revealed increased ESCC risk. We also found a potential gene-gene interaction between rs2735343 and rs701848 and a potential gene-gene interaction among all five SNPs.
Collapse
Affiliation(s)
- X Xu
- Department of Gastroenterology, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - G Chen
- Department of Gastroenterology, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - L Wu
- Department of Gastroenterology, Yixing People's Hospital, Wuxi, Jiangsu, China
| | - L Liu
- Department of Gastroenterology, Yixing People's Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
184
|
The tobacco-specific carcinogen-operated calcium channel promotes lung tumorigenesis via IGF2 exocytosis in lung epithelial cells. Nat Commun 2016; 7:12961. [PMID: 27666821 PMCID: PMC5052689 DOI: 10.1038/ncomms12961] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) binding to the tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) induces Ca2+ signalling, a mechanism that is implicated in various human cancers. In this study, we investigated the role of NNK-mediated Ca2+ signalling in lung cancer formation. We show significant overexpression of insulin-like growth factors (IGFs) in association with IGF-1R activation in human preneoplastic lung lesions in smokers. NNK induces voltage-dependent calcium channel (VDCC)-intervened calcium influx in airway epithelial cells, resulting in a rapid IGF2 secretion via the regulated pathway and thus IGF-1R activation. Silencing nAChR, α1 subunit of L-type VDCC, or various vesicular trafficking curators, including synaptotagmins and Rabs, or blockade of nAChR/VDCC-mediated Ca2+ influx significantly suppresses NNK-induced IGF2 exocytosis, transformation and tumorigenesis of lung epithelial cells. Publicly available database reveals inverse correlation between use of calcium channel blockers and lung cancer diagnosis. Our data indicate that NNK disrupts the regulated pathway of IGF2 exocytosis and promotes lung tumorigenesis. The binding of tobacco-specific carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) to nicotinic acetylcholine receptors (nAChRs) induces calcium signalling. Here the authors show that NKK-induced calcium influx in airway epithelial cells triggers IGF2 secretion and tumourigenesis.
Collapse
|
185
|
Wang L, Yu X, Dong J, Meng Y, Yang Y, Wang H, Wang C, Zhang Y, Zhao Y, Zhao J, Wang H, Lu C, Li B. Combined SRC inhibitor saracatinib and anti-ErbB2 antibody H2-18 produces a synergistic antitumor effect on trastuzumab-resistant breast cancer. Biochem Biophys Res Commun 2016; 479:563-570. [PMID: 27666484 DOI: 10.1016/j.bbrc.2016.09.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/21/2016] [Indexed: 10/21/2022]
Abstract
Despite of the effectiveness of the anti-ErbB2 humanized antibody trastuzumab, trastuzumab resistance emerges as a major and common clinical problem. Thus, circumventing trastuzumab resistance has become an urgent need. Recently, Src inhibitor saracatinib has drawn great attention for its key role in trastuzumab response. As shown in our previous study, H2-18, an anti-ErbB2 antibody, could potently induce programmed cell death (PCD) in trastuzumab-resistant breast cancer cells. Here we combined H2-18 and a Src inhibitor, saracatinib, and studied the antitumor activity of this drug combination in trastuzumab-resistant breast cancer cell lines. The results showed that H2-18 and saracatinib could synergistically inhibit cell proliferation of BT-474, SKBR-3, HCC-1954 and HCC-1419 breast cancer cell lines in vitro. H2-18 plus saracatinib could also inhibit the HCC-1954 tumor growth more effectively in vivo than each drug alone. H2-18 plus saracatinib showed a significantly more potent PCD-inducing activity compared with either H2-18 or saracatinib alone. We conclude that enhanced PCD may contribute to the superior antitumor efficacy of this combination therapy. The combination of H2-18 and SRC inhibitor has the potential to be translated into clinic.
Collapse
Affiliation(s)
- Lingfei Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Xiaojie Yu
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jian Dong
- Department of Vascular Surgery, Changhai Hospital, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Yang Yang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Huajing Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Chao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yajun Zhang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Yirong Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jian Zhao
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Hao Wang
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China.
| | - Cuihua Lu
- Department of Gastroenterology, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, People's Republic of China.
| | - Bohua Li
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
186
|
Wang IX, Grunseich C, Chung YG, Kwak H, Ramrattan G, Zhu Z, Cheung VG. RNA-DNA sequence differences in Saccharomyces cerevisiae. Genome Res 2016; 26:1544-1554. [PMID: 27638543 PMCID: PMC5088596 DOI: 10.1101/gr.207878.116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 09/15/2016] [Indexed: 01/06/2023]
Abstract
Alterations of RNA sequences and structures, such as those from editing and alternative splicing, result in two or more RNA transcripts from a DNA template. It was thought that in yeast, RNA editing only occurs in tRNAs. Here, we found that Saccharomyces cerevisiae have all 12 types of RNA–DNA sequence differences (RDDs) in the mRNA. We showed these sequence differences are propagated to proteins, as we identified peptides encoded by the RNA sequences in addition to those by the DNA sequences at RDD sites. RDDs are significantly enriched at regions with R-loops. A screen of yeast mutants showed that RDD formation is affected by mutations in genes regulating R-loops. Loss-of-function mutations in ribonuclease H, senataxin, and topoisomerase I that resolve RNA–DNA hybrids lead to increases in RDD frequency. Our results demonstrate that RDD is a conserved process that diversifies transcriptomes and proteomes and provide a mechanistic link between R-loops and RDDs.
Collapse
Affiliation(s)
- Isabel X Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Youree G Chung
- College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Hojoong Kwak
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Girish Ramrattan
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| | - Zhengwei Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Vivian G Cheung
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA.,Departments of Pediatrics and Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
187
|
Yang Z, Xie C, Xu W, Liu G, Cao X, Li W, Chen J, Zhu Y, Luo S, Luo Z, Lu N. Phosphorylation and inactivation of PTEN at residues Ser380/Thr382/383 induced by Helicobacter pylori promotes gastric epithelial cell survival through PI3K/Akt pathway. Oncotarget 2016; 6:31916-26. [PMID: 26376616 PMCID: PMC4741650 DOI: 10.18632/oncotarget.5577] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023] Open
Abstract
Phosphorylation of PTEN at residues Ser380/Thr382/383 leads to loss of phosphatase activity and tumor suppressor function. Here, we found that phosphorylation of PTEN at residues Ser380/Thr382/383 was increased with gastric carcinogenesis, and more importantly, Helicobacter pylori was a trigger of this modification in chronic non-atrophic gastritis. H. pylori could phosphorylate and inactivate PTEN in vivo and in vitro, resulting in survival of gastric epithelial cells. Furthermore, stable expression of dominant-negative mutant PTEN or inhibition of Akt prevented the enhanced survival induced by H. pylori. These results indicate that PTEN phosphorylation at residues Ser380/Thr382/383 is a novel mechanism of PTEN inactivation in gastric carcinogenesis, and H. pylori triggers this modification, resulting in activation of the PI3K/Akt pathway and promotion of cell survival.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wenting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Gongmeizi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ximei Cao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhijun Luo
- The Medical College of Nanchang University, Nanchang, Jiangxi, China.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
188
|
Mazzolini L, Broban A, Froment C, Burlet-Schiltz O, Besson A, Manenti S, Dozier C. Phosphorylation of CDC25A on SER283 in late S/G2 by CDK/cyclin complexes accelerates mitotic entry. Cell Cycle 2016; 15:2742-52. [PMID: 27580187 DOI: 10.1080/15384101.2016.1220455] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Cdc25A phosphatase is an essential activator of CDK-cyclin complexes at all steps of the eukaryotic cell cycle. The activity of Cdc25A is itself regulated in part by positive and negative feedback regulatory loops performed by its CDK-cyclin substrates that occur in G1 as well as during the G1/S and G2/M transitions. However, the regulation of Cdc25A during G2 phase progression before mitotic entry has not been intensively characterized. Here, we identify by mass spectrometry analysis a new phosphorylation event of Cdc25A on Serine283. Phospho-specific antibodies revealed that the phosphorylation of this residue appears in late S/G2 phase of an unperturbed cell cycle and is performed by CDK-cyclin complexes. Overexpression studies of wild-type and non-phosphorylatable mutant forms of Cdc25A indicated that Ser283 phosphorylation increases the G2/M-promoting activity of the phosphatase without impacting its stability or subcellular localization. Our results therefore identify a new positive regulatory loop between Cdc25A and its CDK-cyclin substrates which contributes to accelerate entry into mitosis through the regulation of Cdc25A activity in G2.
Collapse
Affiliation(s)
- Laurent Mazzolini
- a Centre de Recherche en Cancérologie de Toulouse, INSERM UMR1037, CNRS ERL5294 , Université Toulouse III Paul Sabatier , Toulouse , France.,b Equipe labellisée LIGUE contre le Cancer , CNRS ERL5294 , Toulouse , France
| | - Anaïs Broban
- a Centre de Recherche en Cancérologie de Toulouse, INSERM UMR1037, CNRS ERL5294 , Université Toulouse III Paul Sabatier , Toulouse , France
| | - Carine Froment
- c Institut de Pharmacologie et de Biologie Structurale , Université Toulouse III Paul Sabatier Toulouse , CNRS UMR5089 , Toulouse , France
| | - Odile Burlet-Schiltz
- c Institut de Pharmacologie et de Biologie Structurale , Université Toulouse III Paul Sabatier Toulouse , CNRS UMR5089 , Toulouse , France
| | - Arnaud Besson
- a Centre de Recherche en Cancérologie de Toulouse, INSERM UMR1037, CNRS ERL5294 , Université Toulouse III Paul Sabatier , Toulouse , France.,b Equipe labellisée LIGUE contre le Cancer , CNRS ERL5294 , Toulouse , France
| | - Stéphane Manenti
- a Centre de Recherche en Cancérologie de Toulouse, INSERM UMR1037, CNRS ERL5294 , Université Toulouse III Paul Sabatier , Toulouse , France.,b Equipe labellisée LIGUE contre le Cancer , CNRS ERL5294 , Toulouse , France
| | - Christine Dozier
- a Centre de Recherche en Cancérologie de Toulouse, INSERM UMR1037, CNRS ERL5294 , Université Toulouse III Paul Sabatier , Toulouse , France.,b Equipe labellisée LIGUE contre le Cancer , CNRS ERL5294 , Toulouse , France
| |
Collapse
|
189
|
Mizerska-Kowalska M, Bojarska-Junak A, Jakubowicz-Gil J, Kandefer-Szerszeń M. Neutral endopeptidase (NEP) is differentially involved in biological activities and cell signaling of colon cancer cell lines derived from various stages of tumor development. Tumour Biol 2016; 37:13355-13368. [PMID: 27460083 PMCID: PMC5097095 DOI: 10.1007/s13277-016-5248-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/15/2016] [Indexed: 01/08/2023] Open
Abstract
The presented studies were aimed at exploring the role of neutral endopeptidase (NEP) in the function of colon cancer cell lines LS 180 and SW 620, derived from different grades and stages of tumor development. NEP silencing by siRNA resulted in decreased viability and proliferation accompanied by increased apoptosis in both cell lines. Additionally, cell cycle arrest at the G2/M phase was observed, but only in LS 180 cells. Opposite to these results, serum-stimulated migration was increased in both cell lines. Furthermore, NEP silencing influenced the invasive activity of LS 180 and SW 620 cells in an opposite manner: while LS 180 cells showed an enhanced invasiveness, SW 620 cells exerted a reduced activity. An exploration of the activity of signaling molecules responsible for the function of tumor cells—Akt, PTEN, and FAK—after NEP silencing indicated that the endopeptidase is involved in their regulation. The increased phosphorylation level of Akt was accompanied by a decrease in PTEN in the presence of a high concentration of serum. A reduced concentration of serum did not change the phosphorylation status of Akt. Enhanced autophosphorylation of FAK was observed in LS 180 and SW 620 cells cultivated in a medium with a high concentration of serum. Taken together, these results confirm that NEP is implicated in the regulation of the survival, growth, and motile activity of colon cancer. This is also the first report which shows that NEP mediates cancer cell migration and invasiveness, but not growth and survival, through Akt/FAK signaling pathways.
Collapse
Affiliation(s)
- Magdalena Mizerska-Kowalska
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Faculty of Biology and Biotechnology, Akademicka 19 Street, 20-033, Lublin, Poland.
| | - Agnieszka Bojarska-Junak
- Chair and Department of Clinical Immunology, Medical University of Lublin, W.Chodźki 4a Street, Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Faculty of Biology and Biotechnology, Akademicka 19 Street, 20-033, Lublin, Poland
| | - Martyna Kandefer-Szerszeń
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Faculty of Biology and Biotechnology, Akademicka 19 Street, 20-033, Lublin, Poland
| |
Collapse
|
190
|
Zhang L, Kondo H, Kamikubo H, Kataoka M, Sakamoto W. VIPP1 Has a Disordered C-Terminal Tail Necessary for Protecting Photosynthetic Membranes against Stress. PLANT PHYSIOLOGY 2016; 171:1983-95. [PMID: 27208228 PMCID: PMC4936581 DOI: 10.1104/pp.16.00532] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 05/09/2016] [Indexed: 05/19/2023]
Abstract
Integrity of biomembranes is vital to living organisms. In bacteria, PspA is considered to act as repairing damaged membrane by forming large supercomplexes in Arabidopsis (Arabidopsis thaliana). Vulnerable to oxidative stress, photosynthetic organisms also contain a PspA ortholog called VIPP1, which has an additional C-terminal tail (Vc). In this study, Vc was shown to coincide with an intrinsically disordered region, and the role of VIPP1 in membrane protection against stress was investigated. We visualized VIPP1 by fusing it to GFP (VIPP1-GFP that fully complemented lethal vipp1 mutations), and investigated its behavior in vivo with live imaging. The intrinsically disordered nature of Vc enabled VIPP1 to form what appeared to be functional particles along envelopes, whereas the deletion of Vc caused excessive association of the VIPP1 particles, preventing their active movement for membrane protection. Expression of VIPP1 lacking Vc complemented vipp1 mutation, but exhibited sensitivity to heat shock stress. Conversely, transgenic plants over-expressing VIPP1 showed enhanced tolerance against heat shock, suggesting that Vc negatively regulates VIPP1 particle association and acts in maintaining membrane integrity. Our data thus indicate that VIPP1 is involved in the maintenance of photosynthetic membranes. During evolution, chloroplasts have acquired enhanced tolerance against membrane stress by incorporating a disordered C-terminal tail into VIPP1.
Collapse
Affiliation(s)
- Lingang Zhang
- Institute of Plant Science and Resources, Okayama University, Kurashiki, Okayama 710-0046, Japan (L.Z., H.Ko., W.S.); School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China (L.Z.); and Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan (H.Ka., M.K.)
| | - Hideki Kondo
- Institute of Plant Science and Resources, Okayama University, Kurashiki, Okayama 710-0046, Japan (L.Z., H.Ko., W.S.); School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China (L.Z.); and Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan (H.Ka., M.K.)
| | - Hironari Kamikubo
- Institute of Plant Science and Resources, Okayama University, Kurashiki, Okayama 710-0046, Japan (L.Z., H.Ko., W.S.); School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China (L.Z.); and Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan (H.Ka., M.K.)
| | - Mikio Kataoka
- Institute of Plant Science and Resources, Okayama University, Kurashiki, Okayama 710-0046, Japan (L.Z., H.Ko., W.S.); School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China (L.Z.); and Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan (H.Ka., M.K.)
| | - Wataru Sakamoto
- Institute of Plant Science and Resources, Okayama University, Kurashiki, Okayama 710-0046, Japan (L.Z., H.Ko., W.S.); School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China (L.Z.); and Graduate School of Materials Science, Nara Institute of Science and Technology, Ikoma, Nara 630-0101, Japan (H.Ka., M.K.)
| |
Collapse
|
191
|
Kaliszczak M, Trousil S, Ali T, Aboagye EO. AKT activation controls cell survival in response to HDAC6 inhibition. Cell Death Dis 2016; 7:e2286. [PMID: 27362804 PMCID: PMC5108334 DOI: 10.1038/cddis.2016.180] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/11/2016] [Accepted: 05/23/2016] [Indexed: 01/08/2023]
Abstract
HDAC6 is emerging as an important therapeutic target for cancer. We investigated mechanisms responsible for survival of tumor cells treated with a HDAC6 inhibitor. Expression of the 20 000 genes examined did not change following HDAC6 treatment in vivo. We found that HDAC6 inhibition led to an increase of AKT activation (P-AKT) in vitro, and genetic knockdown of HDAC6 phenocopied drug-induced AKT activation. The activation of AKT was not observed in PTEN null cells; otherwise, PTEN/PIK3CA expression per se did not predict HDAC6 inhibitor sensitivity. Interestingly, HDAC6 inhibitor treatment led to inactivating phosphorylation of PTEN (P-PTEN Ser380), which likely led to the increased P-AKT in cells that express PTEN. Synergy was observed with phosphatidylinositol 3'-kinases (PI3K) inhibitor treatment in vitro, accompanied by increased caspase 3/7 activity. Furthermore, combination of HDAC6 inhibitor with a PI3K inhibitor caused substantial tumor growth inhibition in vivo compared with either treatment alone, also detectable by Ki-67 immunostaining and (18)F-FLT positron emission tomography (PET). In aggregate AKT activation appears to be a key survival mechanism for HDAC6 inhibitor treatment. Our findings indicate that dual inhibition of HDAC6 and P-AKT may be necessary to substantially inhibit growth of solid tumors.
Collapse
Affiliation(s)
- M Kaliszczak
- Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - S Trousil
- Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - T Ali
- Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - E O Aboagye
- Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
192
|
Human Cytomegalovirus Induces an Atypical Activation of Akt To Stimulate the Survival of Short-Lived Monocytes. J Virol 2016; 90:6443-6452. [PMID: 27147739 DOI: 10.1128/jvi.00214-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/26/2016] [Indexed: 02/02/2023] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is a pervasive herpesvirus responsible for significant morbidity and mortality among immunodeficient/naive hosts. Following a primary HCMV infection, circulating blood monocytes mediate the systemic spread of the virus. Extending the short 48-h life span of monocytes is critical to the viral dissemination process, as these blood-borne cells are nonpermissive for virus replication until they are fully differentiated into macrophages. Here, we show that HCMV glycoprotein gB binding to cellular epidermal growth factor receptor (EGFR) during HCMV entry initiated a rapid (within 15 min) activation of the apoptosis suppressor Akt, which was maintained through 72 h. The virus-induced activation of Akt was more robust than that with the normal myeloid growth factor macrophage colony-stimulating factor (M-CSF) and was essential for infected monocytes to bypass the 48-h viability checkpoint. Activation of phosphoinositide 3-kinase (PI3K) following EGFR engagement by HCMV mediated the phosphorylation of Akt. Moreover, HCMV entry drove a switch away from the PI3K p110δ isoform, which was required for the viability of uninfected monocytes, to the p110β isoform in order to facilitate the Akt-dependent prosurvival state within infected cells. Simultaneously, in contrast to M-CSF, HCMV promoted a rapid increase in SH2 domain-containing inositol 5-phosphatase 1 (SHIP1) expression, leading to signaling through a noncanonical Akt activation pathway. To ensure maximum Akt activity, HCMV also induced an early phosphorylation-dependent inactivation of the negative regulator phosphatase and tensin homolog. Overall, our data indicate that HCMV hijacks the upstream Akt signaling network to induce a nontraditional activation of Akt and subsequently a prosurvival decision at the 48-h cell fate checkpoint, a vital step for HCMV's dissemination and persistence strategy. IMPORTANCE HCMV is found throughout the world with a prevalence of 55 to 100% within the human population. HCMV infection is generally asymptomatic in immunocompetent or naive individuals but is a significant cause of morbidity and mortality among the immunocompromised. Widespread organ inflammation is associated with symptomatic infections, which is a direct consequence of the viral dissemination strategy. Inflammatory peripheral blood monocytes facilitate the spread of HCMV. However, HCMV must subvert the naturally short life span of monocytes. In this work, we demonstrate that HCMV induces the activation of Akt, an antiapoptotic protein, in a manner distinct from that of normal myeloid growth factors. Moreover, we decipher how HCMV dysregulates the upstream Akt signaling network during viral entry to promote an Akt-dependent prosurvival state following infection. Delineation of the virus-specific mechanisms that regulate cellular prosurvival pathways in order to drive the survival of HCMV-infected monocytes is important to identifying new anti-HCMV therapeutic targets.
Collapse
|
193
|
Chen Z, Thomas SN, Bolduc DM, Jiang X, Zhang X, Wolberger C, Cole PA. Enzymatic Analysis of PTEN Ubiquitylation by WWP2 and NEDD4-1 E3 Ligases. Biochemistry 2016; 55:3658-66. [PMID: 27295432 DOI: 10.1021/acs.biochem.6b00448] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PTEN is a lipid phosphatase that converts phosphatidylinositol 3,4,5-phosphate (PIP3) to phosphatidylinositol 4,5-phosphate (PIP2) and plays a critical role in the regulation of tumor growth. PTEN is subject to regulation by a variety of post-translational modifications, including phosphorylation on a C-terminal cluster of four Ser/Thr residues (380, 382, 383, and 385) and ubiquitylation by various E3 ligases, including NEDD4-1 and WWP2. It has previously been shown that C-terminal phosphorylation of PTEN can increase its cellular half-life. Using in vitro ubiquitin transfer assays, we show that WWP2 is more active than NEDD4-1 in ubiquitylating unphosphorylated PTEN. The mapping of ubiquitylation sites in PTEN by mass spectrometry showed that both NEDD4-1 and WWP2 can target a broad range of Lys residues in PTEN, although NEDD4-1 versus WWP2 showed a stronger preference for ubiquitylating PTEN's C2 domain. Whereas tetraphosphorylation of PTEN did not significantly affect its ubiquitylation by NEDD4-1, it inhibited PTEN ubiquitylation by WWP2. Single-turnover and pull-down experiments suggested that tetraphosphorylation of PTEN appears to weaken its interaction with WWP2. These studies reveal how the PTEN E3 ligases WWP2 and NEDD4-1 exhibit distinctive properties in Lys selectivity and sensitivity to PTEN phosphorylation. Our findings also provide a molecular mechanism for the connection between PTEN Ser/Thr phosphorylation and PTEN's cellular stability.
Collapse
Affiliation(s)
- Zan Chen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| | - Stefani N Thomas
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| | - David M Bolduc
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center , New York, New York 10065, United States
| | - Xiangbin Zhang
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| | - Cynthia Wolberger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| | - Philip A Cole
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine , Baltimore, Maryland 21205, United States
| |
Collapse
|
194
|
Pten regulates spindle pole movement through Dlg1-mediated recruitment of Eg5 to centrosomes. Nat Cell Biol 2016; 18:814-21. [PMID: 27240320 DOI: 10.1038/ncb3369] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/29/2016] [Indexed: 12/18/2022]
Abstract
Phosphatase and tensin homologue (Pten) suppresses neoplastic growth by negatively regulating PI(3)K signalling through its phosphatase activity. To gain insight into the actions of non-catalytic Pten domains in normal physiological processes and tumorigenesis, we engineered mice lacking the PDZ-binding domain (PDZ-BD). Here, we show that the PDZ-BD regulates centrosome movement and that its heterozygous or homozygous deletion promotes aneuploidy and tumour formation. We found that Pten is recruited to pre-mitotic centrosomes in a Plk1-dependent fashion to create a docking site for protein complexes containing the PDZ-domain-containing protein Dlg1 (also known as Sap97) and Eg5 (also known as Kif11), a kinesin essential for centrosome movement and bipolar spindle formation. Docking of Dlg1-Eg5 complexes to Pten depended on Eg5 phosphorylation by the Nek9-Nek6 mitotic kinase cascade and Cdk1. PDZ-BD deletion or Dlg1 ablation impaired loading of Eg5 onto centrosomes and spindle pole motility, yielding asymmetrical spindles that are prone to chromosome missegregation. Collectively, these data demonstrate that Pten, through the Dlg1-binding ability of its PDZ-BD, accumulates phosphorylated Eg5 at duplicated centrosomes to establish symmetrical bipolar spindles that properly segregate chromosomes, and suggest that this function contributes to tumour suppression.
Collapse
|
195
|
Zhou J, Ling J, Song J, Wang Y, Feng B, Ping F. Interleukin 10 protects primary melanocyte by activation of Stat-3 and PI3K/Akt/NF-κB signaling pathways. Cytokine 2016; 83:275-281. [PMID: 27186967 DOI: 10.1016/j.cyto.2016.05.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/25/2016] [Accepted: 05/11/2016] [Indexed: 01/16/2023]
Abstract
Vitiligo is a common melanocytopenic disorder of the skin, with acquired focal depigmentation. Normal human skin relies on melanocytes to provide photoprotection and thermoregulation by producing melanin. Interleukin 10 (IL-10) is a pleiotropic immunoregulatory cytokine drawing more and more researchers' attention. The present study was conducted to investigate the effects of IL-10 on melanocytes and elucidate the underlying mechanisms. We proved that IL-10 play no role in regulating melanogenesis of normal human foreskin-derived epidermal melanocytes (NHEM). IL-10 stimulation activated the JAK/Stat-3 and PI3K/Akt signaling pathways. Moreover, IL-10 treatment increased translocation of p65 NF-κB into the nuclear compartment, and up-regulated expression of the pro-survival proteins Bcl-2 and Bcl-xL. IL-10 restored anti-apoptotic proteins expression and suppressed cytochrome c release in H2O2-induced apoptosis. In conclusion, IL-10 may provide pro-survival cues to melanocytes and be applied in the treatment of vitiligo and other depigmenting disorders.
Collapse
Affiliation(s)
- Jia Zhou
- School of Pharmaceutical Science, Jiangnan University, Wuxi 214122, PR China
| | - Jingjing Ling
- Department of Pharmacy, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, PR China
| | - Jing Song
- Laboratory Animal Center, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Yong Wang
- Department of Urological Surgical, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, PR China
| | - Bainian Feng
- School of Pharmaceutical Science, Jiangnan University, Wuxi 214122, PR China
| | - Fengfeng Ping
- Department of Clinical Laboratory Science, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, PR China.
| |
Collapse
|
196
|
Chen Z, Dempsey DR, Thomas SN, Hayward D, Bolduc DM, Cole PA. Molecular Features of Phosphatase and Tensin Homolog (PTEN) Regulation by C-terminal Phosphorylation. J Biol Chem 2016; 291:14160-14169. [PMID: 27226612 DOI: 10.1074/jbc.m116.728980] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Indexed: 12/27/2022] Open
Abstract
PTEN is a tumor suppressor that functions to negatively regulate the PI3K/AKT pathway as the lipid phosphatase for phosphatidylinositol 3,4,5-triphosphate. Phosphorylation of a cluster of Ser/Thr residues (amino acids 380-385) on the C-terminal tail serves to alter the conformational state of PTEN from an open active state to a closed inhibited state, resulting in a reduction of plasma membrane localization and inhibition of enzyme activity. The relative contribution of each phosphorylation site to PTEN autoinhibition and the structural basis for the conformational closure is still unclear. To further the structural understanding of PTEN regulation by C-terminal tail phosphorylation, we used protein semisynthesis to insert stoichiometric and site-specific phospho-Ser/Thr(s) in the C-terminal tail of PTEN. Additionally, we employed photo-cross-linking to map the intramolecular PTEN interactions of the phospho-tail. Systematic evaluation of the PTEN C-tail phospho-cluster showed autoinhibition, and conformational closure was influenced by the aggregate effect of multiple phospho-sites rather than dominated by a single phosphorylation site. Moreover, photo-cross-linking suggested a direct interaction between the PTEN C-tail and a segment in the N-terminal region of the catalytic domain. Mutagenesis experiments provided additional insights into how the PTEN phospho-tail interacts with both the C2 and catalytic domains.
Collapse
Affiliation(s)
- Zan Chen
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Daniel R Dempsey
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Stefani N Thomas
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Dawn Hayward
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - David M Bolduc
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Philip A Cole
- Department of Pharmacology and Molecular Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205.
| |
Collapse
|
197
|
Stumpf M, Blokzijl-Franke S, den Hertog J. Fine-Tuning of Pten Localization and Phosphatase Activity Is Essential for Zebrafish Angiogenesis. PLoS One 2016; 11:e0154771. [PMID: 27138341 PMCID: PMC4854392 DOI: 10.1371/journal.pone.0154771] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/19/2016] [Indexed: 02/06/2023] Open
Abstract
The lipid- and protein phosphatase PTEN is an essential tumor suppressor that is highly conserved among all higher eukaryotes. As an antagonist of the PI3K/Akt cell survival and proliferation pathway, it exerts its most prominent function at the cell membrane, but (PIP3-independent) functions of nuclear PTEN have been discovered as well. PTEN subcellular localization is tightly controlled by its protein conformation. In the closed conformation, PTEN localizes predominantly to the cytoplasm. Opening up of the conformation of PTEN exposes N-terminal and C-terminal regions of the protein that are required for both interaction with the cell membrane and translocation to the nucleus. Lack of Pten leads to hyperbranching of the intersegmental vessels during zebrafish embryogenesis, which is rescued by expression of exogenous Pten. Here, we observed that expression of mutant PTEN with an open conformation rescued the hyperbranching phenotype in pten double homozygous embryos and suppressed the increased p-Akt levels that are characteristic for embryos lacking Pten. In addition, in pten mutant and wild type embryos alike, open conformation PTEN induced stalled intersegmental vessels, which fail to connect with the dorsal longitudinal anastomotic vessel. Functional hyperactivity of open conformation PTEN in comparison to wild type PTEN seems to result predominantly from its enhanced recruitment to the cell membrane. Enhanced recruitment of phosphatase inactive mutants to the membrane did not induce the stalled vessel phenotype nor did it rescue the hyperbranching phenotype in pten double homozygous embryos, indicating that PTEN phosphatase activity is indispensable for its regulatory function during angiogenesis. Taken together, our data suggest that PTEN phosphatase activity needs to be carefully fine-tuned for normal embryogenesis and that the control of its subcellular localization is a key mechanism in this process.
Collapse
Affiliation(s)
- Miriam Stumpf
- Hubrecht Institute–Koninklijke Nederlandse Akademie van Wetenschappen (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sasja Blokzijl-Franke
- Hubrecht Institute–Koninklijke Nederlandse Akademie van Wetenschappen (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute–Koninklijke Nederlandse Akademie van Wetenschappen (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
198
|
Mandato E, Manni S, Zaffino F, Semenzato G, Piazza F. Targeting CK2-driven non-oncogene addiction in B-cell tumors. Oncogene 2016; 35:6045-6052. [PMID: 27041560 DOI: 10.1038/onc.2016.86] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/15/2016] [Accepted: 02/15/2016] [Indexed: 12/14/2022]
Abstract
Genetic mutations of oncogenes often underlie deranged cell growth and altered differentiation pathways leading to malignant transformation of B-lymphocytes. However, addiction to oncogenes is not the only drive to lymphoid tumor pathogenesis. Dependence on non-oncogenes, which act by propelling basic mechanisms of cell proliferation and survival, has also been recognized in the pathobiology of lymphoid leukemias, lymphomas and multiple myeloma. Among the growing number of molecules that may uphold non-oncogene addiction, a key place is increasingly being recognized to the serine-threonine kinase CK2. This enzyme is overexpressed and overactive in B-acute lymphoblastic leukemia, multiple myeloma, chronic lymphocytic leukemia and non-Hodgkin lymphomas, such as mantle cell, follicular, Burkitt's and diffuse large B-cell lymphomas. In these tumors, CK2 may serve the activity of oncogenes, similar to BCR-ABL and c-MYC, control the activation of critical signaling cascades, such as NF-κB (nuclear factor-κB), STAT3 (signal transducer and activator of transcription 3) and PTEN/PI3K/AKT (phosphatase and tensin homolog protein/phosphoinositide 3-kinase/AKR thymoma), and sustain multiple cellular stress-elicited pathways, such as the proteotoxic stress, unfolded protein and DNA-damage responses. CK2 has also been shown to have an essential role in tuning signals derived from the stromal tumor microenvironment. Not surprisingly, targeting CK2 in lymphoid tumor cell lines or mouse xenograft models can boost the cytotoxic effects of both conventional chemotherapeutics and novel agents, similar to heat-shock protein 90, proteasome and tyrosine kinases inhibitors. In this review, we summarize the evidence indicating how CK2 embodies most of the features of a cancer growth-promoting non-oncogene, focusing on lymphoid tumors. We further discuss the preclinical data of the use of small ATP-competitive CK2 inhibitors, which hold the promise to be additional options in novel drug combinations for the therapy of lymphoid and plasmacellular malignancies.
Collapse
Affiliation(s)
- E Mandato
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - S Manni
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Zaffino
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - G Semenzato
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - F Piazza
- Department of Medicine, Hematology Branch, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
199
|
Jang H, Lee OH, Lee Y, Yoon H, Chang EM, Park M, Lee JW, Hong K, Kim JO, Kim NK, Ko JJ, Lee DR, Yoon TK, Lee WS, Choi Y. Melatonin prevents cisplatin-induced primordial follicle loss via suppression of PTEN/AKT/FOXO3a pathway activation in the mouse ovary. J Pineal Res 2016; 60:336-47. [PMID: 26882203 DOI: 10.1111/jpi.12316] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 02/09/2016] [Indexed: 12/25/2022]
Abstract
Premature ovarian failure (POF) is a major side effect of chemotherapy in young cancer patients. To develop pharmaceutical agents for preserving fertility, it is necessary to understand the mechanisms responsible for chemotherapy-induced follicle loss. Here, we show that treatment with cisplatin, a widely used anticancer drug, depleted the dormant follicle pool in mouse ovaries by excessive activation of the primordial follicles, without inducing follicular apoptosis. Moreover, we show that co-treatment with the antioxidant melatonin prevented cisplatin-induced disruption of the follicle reserve. We quantified the various stages of growing follicles, including primordial, primary, secondary, and antral, to demonstrate that cisplatin treatment alone significantly decreased, whereas melatonin co-treatment preserved, the number of primordial follicles in the ovary. Importantly, analysis of the PTEN/AKT/FOXO3a pathway demonstrated that melatonin significantly decreased the cisplatin-mediated inhibitory phosphorylation of PTEN, a key negative regulator of dormant follicle activation. Moreover, melatonin prevented the cisplatin-induced activating phosphorylation of AKT, GSK3β, and FOXO3a, all of which trigger follicle activation. Additionally, we show that melatonin inhibited the cisplatin-induced inhibitory phosphorylation and nuclear export of FOXO3a, which is required in the nucleus to maintain dormancy of the primordial follicles. These findings demonstrate that melatonin attenuates cisplatin-induced follicle loss by preventing the phosphorylation of PTEN/AKT/FOXO3a pathway members; thus, melatonin is a potential therapeutic agent for ovarian protection and fertility preservation during chemotherapy in female cancer patients.
Collapse
Affiliation(s)
- Hoon Jang
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Ok-Hee Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Youngeun Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Hyemin Yoon
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Eun Mi Chang
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Miseon Park
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Jeong-Woong Lee
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
- Functional Genomics, School of Engineering, University of Science and Technology (UST), Daejeon, Korea
| | - Kwonho Hong
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, Korea
| | - Jung Oh Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Nam Keun Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Jung Jae Ko
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
| | - Dong Ryul Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Tae Ki Yoon
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Woo Sik Lee
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Youngsok Choi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Korea
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, Korea
| |
Collapse
|
200
|
Inherited PTEN mutations and the prediction of phenotype. Semin Cell Dev Biol 2016; 52:30-8. [DOI: 10.1016/j.semcdb.2016.01.030] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/21/2015] [Accepted: 01/21/2016] [Indexed: 12/19/2022]
|