1
|
He J, Huang C, Guo Y, Deng R, Li L, Chen R, Wang Y, Huang J, Zheng J, Zhao X, Yu J. PTEN-mediated dephosphorylation of 53BP1 confers cellular resistance to DNA damage in cancer cells. Mol Oncol 2024; 18:580-605. [PMID: 38060346 PMCID: PMC10920079 DOI: 10.1002/1878-0261.13563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 03/09/2024] Open
Abstract
Homologous recombination (HR) repair for DNA double-strand breaks (DSBs) is critical for maintaining genome stability and conferring the resistance of tumor cells to chemotherapy. Nuclear PTEN which contains both phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and protein phosphatase plays a key role in HR repair, but the underlying mechanism remains largely elusive. We find that SUMOylated PTEN promotes HR repair but represses nonhomologous end joining (NHEJ) repair by directly dephosphorylating TP53-binding protein 1 (53BP1). During DNA damage responses (DDR), tumor suppressor ARF (p14ARF) was phosphorylated and then interacted efficiently with PTEN, thus promoting PTEN SUMOylation as an atypical SUMO E3 ligase. Interestingly, SUMOylated PTEN was subsequently recruited to the chromatin at DSB sites. This was because SUMO1 that was conjugated to PTEN was recognized and bound by the SUMO-interacting motif (SIM) of breast cancer type 1 susceptibility protein (BRCA1), which has been located to the core of 53BP1 foci on chromatin during S/G2 stage. Furthermore, these chromatin-loaded PTEN directly and specifically dephosphorylated phosphothreonine-543 (pT543) of 53BP1, resulting in the dissociation of the 53BP1 complex, which facilitated DNA end resection and ongoing HR repair. SUMOylation-site-mutated PTENK254R mice also showed decreased DNA damage repair in vivo. Blocking the PTEN SUMOylation pathway with either a SUMOylation inhibitor or a p14ARF(2-13) peptide sensitized tumor cells to chemotherapy. Our study therefore provides a new mechanistic understanding of PTEN in HR repair and clinical intervention of chemoresistant tumors.
Collapse
Affiliation(s)
- Jianfeng He
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Caihu Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Yanmin Guo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Rong Deng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Lian Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Ran Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Yanli Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Jian Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Junke Zheng
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Xian Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and InflammationShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
2
|
Smith IN, Dawson JE, Eng C. Comparative Protein Structural Network Analysis Reveals C-Terminal Tail Phosphorylation Structural Communication Fingerprint in PTEN-Associated Mutations in Autism and Cancer. J Phys Chem B 2023; 127:634-647. [PMID: 36626331 PMCID: PMC9885960 DOI: 10.1021/acs.jpcb.2c06776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/24/2022] [Indexed: 01/11/2023]
Abstract
PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a tightly regulated dual-specificity phosphatase and key regulator of the PI3K/AKT/mTOR signaling pathway. PTEN phosphorylation at its carboxy-terminal tail (CTT) serine/threonine cluster negatively regulates its tumor suppressor function by inducing a stable, closed, and inactive conformation. Germline PTEN mutations predispose individuals to PTEN hamartoma tumor syndrome (PHTS), a rare inherited cancer syndrome and, intriguingly, one of the most common causes of autism spectrum disorder (ASD). However, the mechanistic details that govern phosphorylated CTT catalytic conformational dynamics in the context of PHTS-associated mutations are unknown. Here, we utilized a comparative protein structure network (PSN)-based approach to investigate PTEN CTT phosphorylation-induced conformational dynamics specific to PTEN-ASD compared to PTEN-cancer phenotypes. Results from our study show differences in structural flexibility, inter-residue contacts, and allosteric communication patterns mediated by CTT phosphorylation, differentiating PTEN-ASD and PTEN-cancer phenotypes. Further, we identified perturbations among global metapaths and community network connections within the active site and inter-domain regions, indicating the significance of these regions in transmitting information across the PSN. Together, our studies provide a mechanistic underpinning of allosteric regulation through the coupled interplay of CTT phosphorylation conformational dynamics in PTEN-ASD and PTEN-cancer mutations. Importantly, the detailed atomistic interactions and structural consequences of PTEN variants reveal potential allosteric druggable target sites as a viable and currently unexplored treatment approach for individuals with different PHTS-associated mutations.
Collapse
Affiliation(s)
- Iris N. Smith
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio44195, United States
| | - Jennifer E. Dawson
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio44195, United States
| | - Charis Eng
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio44195, United States
- Cleveland
Clinic Lerner College of Medicine, Case
Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio44195, United
States
- Case
Comprehensive Cancer Center, Case Western
Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio44106, United States
- Taussig
Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio44195, United States
- Department
of Genetics and Genome Sciences, Case Western
Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio44106, United States
| |
Collapse
|
3
|
Smith IN, Dawson JE, Krieger J, Thacker S, Bahar I, Eng C. Structural and Dynamic Effects of PTEN C-Terminal Tail Phosphorylation. J Chem Inf Model 2022; 62:4175-4190. [PMID: 36001481 PMCID: PMC9472802 DOI: 10.1021/acs.jcim.2c00441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Indexed: 11/28/2022]
Abstract
The phosphatase and tensin homologue deleted on chromosome 10 (PTEN) tumor suppressor gene encodes a tightly regulated dual-specificity phosphatase that serves as the master regulator of PI3K/AKT/mTOR signaling. The carboxy-terminal tail (CTT) is key to regulation and harbors multiple phosphorylation sites (Ser/Thr residues 380-385). CTT phosphorylation suppresses the phosphatase activity by inducing a stable, closed conformation. However, little is known about the mechanisms of phosphorylation-induced CTT-deactivation dynamics. Using explicit solvent microsecond molecular dynamics simulations, we show that CTT phosphorylation leads to a partially collapsed conformation, which alters the secondary structure of PTEN and induces long-range conformational rearrangements that encompass the active site. The active site rearrangements prevent localization of PTEN to the membrane, precluding lipid phosphatase activity. Notably, we have identified phosphorylation-induced allosteric coupling between the interdomain region and a hydrophobic site neighboring the active site in the phosphatase domain. Collectively, the results provide a mechanistic understanding of CTT phosphorylation dynamics and reveal potential druggable allosteric sites in a previously believed clinically undruggable protein.
Collapse
Affiliation(s)
- Iris N. Smith
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio 44195, United States
| | - Jennifer E. Dawson
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio 44195, United States
| | - James Krieger
- Department
of Computational and Systems Biology, University
of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Stetson Thacker
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio 44195, United States
- Cleveland
Clinic Lerner College of Medicine, Case
Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio 44195, United
States
| | - Ivet Bahar
- Department
of Computational and Systems Biology, University
of Pittsburgh, 800 Murdoch Building, 3420 Forbes Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Charis Eng
- Genomic
Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE-50, Cleveland, Ohio 44195, United States
- Cleveland
Clinic Lerner College of Medicine, Case
Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio 44195, United
States
- Case
Comprehensive Cancer Center, Case Western
Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Taussig
Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, United States
- Department
of Genetics and Genome Sciences, Case Western
Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
4
|
Matreyek KA, Stephany JJ, Ahler E, Fowler DM. Integrating thousands of PTEN variant activity and abundance measurements reveals variant subgroups and new dominant negatives in cancers. Genome Med 2021; 13:165. [PMID: 34649609 PMCID: PMC8518224 DOI: 10.1186/s13073-021-00984-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background PTEN is a multi-functional tumor suppressor protein regulating cell growth, immune signaling, neuronal function, and genome stability. Experimental characterization can help guide the clinical interpretation of the thousands of germline or somatic PTEN variants observed in patients. Two large-scale mutational datasets, one for PTEN variant intracellular abundance encompassing 4112 missense variants and one for lipid phosphatase activity encompassing 7244 variants, were recently published. The combined information from these datasets can reveal variant-specific phenotypes that may underlie various clinical presentations, but this has not been comprehensively examined, particularly for somatic PTEN variants observed in cancers. Methods Here, we add to these efforts by measuring the intracellular abundance of 764 new PTEN variants and refining abundance measurements for 3351 previously studied variants. We use this expanded and refined PTEN abundance dataset to explore the mutational patterns governing PTEN intracellular abundance, and then incorporate the phosphatase activity data to subdivide PTEN variants into four functionally distinct groups. Results This analysis revealed a set of highly abundant but lipid phosphatase defective variants that could act in a dominant-negative fashion to suppress PTEN activity. Two of these variants were, indeed, capable of dysregulating Akt signaling in cells harboring a WT PTEN allele. Both variants were observed in multiple breast or uterine tumors, demonstrating the disease relevance of these high abundance, inactive variants. Conclusions We show that multidimensional, large-scale variant functional data, when paired with public cancer genomics datasets and follow-up assays, can improve understanding of uncharacterized cancer-associated variants, and provide better insights into how they contribute to oncogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00984-x.
Collapse
Affiliation(s)
- Kenneth A Matreyek
- Department of Genome Sciences, University of Washington, Seattle, WA, USA. .,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Jason J Stephany
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Ethan Ahler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.,Present Address: Revolution Medicines, Redwood City, CA, 94063, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Jang H, Smith IN, Eng C, Nussinov R. The mechanism of full activation of tumor suppressor PTEN at the phosphoinositide-enriched membrane. iScience 2021; 24:102438. [PMID: 34113810 PMCID: PMC8169795 DOI: 10.1016/j.isci.2021.102438] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/15/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor suppressor PTEN, the second most highly mutated protein in cancer, dephosphorylates signaling lipid PIP3 produced by PI3Ks. Excess PIP3 promotes cell proliferation. The mechanism at the membrane of this pivotal phosphatase is unknown hindering drug discovery. Exploiting explicit solvent simulations, we tracked full-length PTEN trafficking from the cytosol to the membrane. We observed its interaction with membranes composed of zwitterionic phosphatidylcholine, anionic phosphatidylserine, and phosphoinositides, including signaling lipids PIP2 and PIP3. We tracked its moving away from the zwitterionic and getting absorbed onto anionic membrane that harbors PIP3. We followed it localizing on microdomains enriched in signaling lipids, as PI3K does, and observed PIP3 allosterically unfolding the N-terminal PIP2 binding domain, positioning it favorably for the polybasic motif interaction with PIP2. Finally, we determined PTEN catalytic action at the membrane, all in line with experimental observations, deciphering the mechanisms of how PTEN anchors to the membrane and restrains cancer. PTEN localizes on membrane microdomains enriched in phosphoinositides, as PI3K does Full PTEN activation requires both signaling lipids, PIP2 and PIP3 Strong salt bridge interactions sustain stable PTEN membrane localization Substrate-induced P loop conformational change implicates PTEN catalytic activity
Collapse
Affiliation(s)
- Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA
| | - Iris Nira Smith
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA.,Center for Personalized Genetic Healthcare, Cleveland Clinic Community Care and Population Health, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Germline High Risk Cancer Focus Group, Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD 21702, USA.,Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
6
|
Smith SL, Pitt AR, Spickett CM. Approaches to Investigating the Protein Interactome of PTEN. J Proteome Res 2020; 20:60-77. [PMID: 33074689 DOI: 10.1021/acs.jproteome.0c00570] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor suppressor phosphatase and tensin homologue (PTEN) is a redox-sensitive dual specificity phosphatase with an essential role in the negative regulation of the PI3K-AKT signaling pathway, affecting metabolic and cell survival processes. PTEN is commonly mutated in cancer, and dysregulation in the metabolism of PIP3 is implicated in other diseases such as diabetes. PTEN interactors are responsible for some functional roles of PTEN beyond the negative regulation of the PI3K pathway and are thus of great importance in cell biology. Both high-data content proteomics-based approaches and low-data content PPI approaches have been used to investigate the interactome of PTEN and elucidate further functions of PTEN. While low-data content approaches rely on co-immunoprecipitation and Western blotting, and as such require previously generated hypotheses, high-data content approaches such as affinity pull-down proteomic assays or the yeast 2-hybrid system are hypothesis generating. This review provides an overview of the PTEN interactome, including redox effects, and critically appraises the methods and results of high-data content investigations into the global interactome of PTEN. The biological significance of findings from recent studies is discussed and illustrates the breadth of cellular functions of PTEN that can be discovered by these approaches.
Collapse
Affiliation(s)
- Sarah L Smith
- School of Life and Health Sciences, Aston Triangle, Aston University, B4 7ET, Birmingham, U.K
| | - Andrew R Pitt
- School of Life and Health Sciences, Aston Triangle, Aston University, B4 7ET, Birmingham, U.K.,Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester, M1 7DN, U.K
| | - Corinne M Spickett
- School of Life and Health Sciences, Aston Triangle, Aston University, B4 7ET, Birmingham, U.K
| |
Collapse
|
7
|
Mills RD, Liang LY, Lio DSS, Mok YF, Mulhern TD, Cao G, Griffin M, Kenche VB, Culvenor JG, Cheng HC. The Roc-COR tandem domain of leucine-rich repeat kinase 2 forms dimers and exhibits conventional Ras-like GTPase properties. J Neurochem 2019; 147:409-428. [PMID: 30091236 DOI: 10.1111/jnc.14566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/18/2022]
Abstract
The Parkinson's disease (PD)-causative leucine-rich repeat kinase 2 (LRRK2) belongs to the Roco family of G-proteins comprising a Ras-of-complex (Roc) domain followed by a C-terminal of Roc (COR) domain in tandem (called Roc-COR domain). Two prokaryotic Roc-COR domains have been characterized as 'G proteins activated by guanine nucleotide-dependent dimerization' (GADs), which require dimerization for activation of their GTPase activity and bind guanine nucleotides with relatively low affinities. Additionally, LRRK2 Roc domain in isolation binds guanine nucleotides with relatively low affinities. As such, LRRK2 GTPase domain was predicted to be a GAD. Herein, we describe the design and high-level expression of human LRRK2 Roc-COR domain (LRRK2 Roc-COR). Biochemical analyses of LRRK2 Roc-COR reveal that it forms homodimers, with the C-terminal portion of COR mediating its dimerization. Furthermore, it co-purifies and binds Mg2+ GTP/GDP at 1 : 1 stoichiometry, and it hydrolyzes GTP with Km and kcat of 22 nM and 4.70 × 10-4 min-1 , respectively. Thus, even though LRRK2 Roc-COR forms GAD-like homodimers, it exhibits conventional Ras-like GTPase properties, with high-affinity binding of Mg2+ -GTP/GDP and low intrinsic catalytic activity. The PD-causative Y1699C mutation mapped to the COR domain was previously reported to reduce the GTPase activity of full-length LRRK2. In contrast, this mutation induces no change in the GTPase activity, and only slight perturbations in the secondary structure contents of LRRK2 Roc-COR. As this mutation does not directly affect the GTPase activity of the isolated Roc-COR tandem, it is possible that the effects of this mutation on full-length LRRK2 occur via other functional domains. Open Practices Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Ryan D Mills
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Lung-Yu Liang
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia.,Cell Signaling Research Laboratories, University of Melbourne, Parkville, Victoria, Australia
| | - Daisy Sio-Seng Lio
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia.,Cell Signaling Research Laboratories, University of Melbourne, Parkville, Victoria, Australia
| | - Yee-Foong Mok
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Terrence D Mulhern
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - George Cao
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael Griffin
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Vijaya B Kenche
- Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia.,Florey Neuroscience Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Janetta G Culvenor
- Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia.,Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia.,Cell Signaling Research Laboratories, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
8
|
Yan R, Wang Y, Shi M, Xiao Y, Liu L, Liu L, Guo B. Regulation of PTEN/AKT/FAK pathways by PPARγ impacts on fibrosis in diabetic nephropathy. J Cell Biochem 2019; 120:6998-7014. [PMID: 30652342 DOI: 10.1002/jcb.27937] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023]
Abstract
Renal tubular epithelial-to-mesenchymal transition (EMT) and tubulointerstitial fibrosis (TIF) are important pathological features of diabetic nephropathy (DN). However, the regulatory mechanism underlying EMT and TIF are still unclear. Previous studies showed that the decrease in the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was closely related to the aggravation of DN, but no published study showed how PTEN participated in the regulation of EMT and TIF. In this study, the rat proximal tubular epithelial cells (NRK52E) and C57BL mice and human kidney tissues were used as the research objects to investigate the mechanism underlying the regulatory effect of peroxisome proliferator-activated receptors γ (PPARγ) on PTEN and its influence on EMT and TIF, the regulation of PTEN's dual activity of lipid phosphatase/protein phosphatase by the serine threonine protein kinase B(AKT)/focal adhesion kinase (FAK) signaling pathway, and the role of PTEN in EMT and TIF. The results showed that PPARγ regulated the expression of PTEN at a transcriptional level and further regulated EMT and TIF. This dual activity could regulate the phosphorylation level of AKT and FAK and also affect FAK transcription. However, the 129 mutant of PTEN (PTEN-G129E) lost the lipid phosphatase activity, and its protein phosphatase activity was involved only in EMT and renal fibrosis through regulating FAK phosphorylation. This study systematically elucidated the role of PPARγ/PTEN/AKT/FAK signaling pathway in EMT and TIF during the pathogenesis of DN.
Collapse
Affiliation(s)
- Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Lirong Liu
- Department of Clinical Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
9
|
Smith IN, Thacker S, Jaini R, Eng C. Dynamics and structural stability effects of germline PTEN mutations associated with cancer versus autism phenotypes. J Biomol Struct Dyn 2018; 37:1766-1782. [PMID: 29663862 DOI: 10.1080/07391102.2018.1465854] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Individuals with germline mutations in the tumor suppressor gene phosphatase and tensin homolog (PTEN), irrespective of clinical presentation, are diagnosed with PTEN hamartoma tumor syndrome (PHTS). PHTS confers a high risk of breast, thyroid, and other cancers or autism spectrum disorder (ASD) with macrocephaly. It remains unclear why mutations in one gene can lead to seemingly disparate phenotypes. Thus, we sought to identify differences in ASD vs. cancer-associated germline PTEN missense mutations by investigating putative structural effects induced by each mutation. We utilized a theoretical computational approach combining in silico structural analysis and molecular dynamics (MD) to interrogate 17 selected mutations from our patient population: six mutations were observed in patients with ASD (only), six mutations in patients with PHTS-associated cancer (only), four mutations shared across both phenotypes, and one mutation with both ASD and cancer. We demonstrate structural stability changes where all six cancer-associated mutations showed a global decrease in structural stability and increased dynamics across the domain interface with a proclivity to unfold, mediating a closed (inactive) active site. In contrast, five of the six ASD-associated mutations showed localized destabilization that contribute to the partial opening of the active site. Our results lend insight into distinctive structural effects of germline PTEN mutations associated with PTEN-ASD vs. those associated with PTEN-cancer, potentially aiding in identification of the shared and separate molecular features that contribute to autism or cancer, thus, providing a deeper understanding of genotype-phenotype relationships for germline PTEN mutations.
Collapse
Affiliation(s)
- Iris Nira Smith
- a Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic , Cleveland , OH , USA
| | - Stetson Thacker
- a Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic , Cleveland , OH , USA.,e Cleveland Clinic Lerner College of Medicine , Cleveland , OH , USA
| | - Ritika Jaini
- a Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic , Cleveland , OH , USA.,d Germline High Risk Cancer Focus Group , Comprehensive Cancer Center, Case Western Reserve University School of Medicine , Cleveland , OH , USA.,e Cleveland Clinic Lerner College of Medicine , Cleveland , OH , USA
| | - Charis Eng
- a Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic , Cleveland , OH , USA.,b Taussig Cancer Institute, Cleveland Clinic , Cleveland , OH , USA.,c Department of Genetics and Genome Sciences, Case Westren Reserve University School of Medicine , Cleveland , OH , USA.,d Germline High Risk Cancer Focus Group , Comprehensive Cancer Center, Case Western Reserve University School of Medicine , Cleveland , OH , USA.,e Cleveland Clinic Lerner College of Medicine , Cleveland , OH , USA
| |
Collapse
|
10
|
Mighell TL, Evans-Dutson S, O'Roak BJ. A Saturation Mutagenesis Approach to Understanding PTEN Lipid Phosphatase Activity and Genotype-Phenotype Relationships. Am J Hum Genet 2018; 102:943-955. [PMID: 29706350 PMCID: PMC5986715 DOI: 10.1016/j.ajhg.2018.03.018] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/16/2018] [Indexed: 12/19/2022] Open
Abstract
Phosphatase and tensin homolog (PTEN) is a tumor suppressor frequently mutated in diverse cancers. Germline PTEN mutations are also associated with a range of clinical outcomes, including PTEN hamartoma tumor syndrome (PHTS) and autism spectrum disorder (ASD). To empower new insights into PTEN function and clinically relevant genotype-phenotype relationships, we systematically evaluated the effect of PTEN mutations on lipid phosphatase activity in vivo. Using a massively parallel approach that leverages an artificial humanized yeast model, we derived high-confidence estimates of functional impact for 7,244 single amino acid PTEN variants (86% of possible). We identified 2,273 mutations with reduced cellular lipid phosphatase activity, which includes 1,789 missense mutations. These data recapitulated known functional findings but also uncovered new insights into PTEN protein structure, biochemistry, and mutation tolerance. Several residues in the catalytic pocket showed surprising mutational tolerance. We identified that the solvent exposure of wild-type residues is a critical determinant of mutational tolerance. Further, we created a comprehensive functional map by leveraging correlations between amino acid substitutions to impute functional scores for all variants, including those not present in the assay. Variant functional scores can reliably discriminate likely pathogenic from benign alleles. Further, 32% of ClinVar unclassified missense variants are phosphatase deficient in our assay, supporting their reclassification. ASD-associated mutations generally had less severe fitness scores relative to PHTS-associated mutations (p = 7.16 × 10-5) and a higher fraction of hypomorphic mutations, arguing for continued genotype-phenotype studies in larger clinical datasets that can further leverage these rich functional data.
Collapse
Affiliation(s)
- Taylor L Mighell
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR 97239, USA; Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sara Evans-Dutson
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brian J O'Roak
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
11
|
Smith IN, Briggs JM. Structural mutation analysis of PTEN and its genotype-phenotype correlations in endometriosis and cancer. Proteins 2016; 84:1625-1643. [PMID: 27481051 DOI: 10.1002/prot.25105] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/17/2016] [Accepted: 07/05/2016] [Indexed: 12/19/2022]
Abstract
The phosphatase and tensin homolog deleted on chromosome ten (PTEN) gene encodes a tumor suppressor phosphatase that has recently been found to be frequently mutated in patients with endometriosis, endometrial cancer and ovarian cancer. Here, we present the first computational analysis of 13 somatic missense PTEN mutations associated with these phenotypes. We found that a majority of the mutations are associated in conserved positions within the active site and are clustered within the signature motif, which contain residues that play a crucial role in loop conformation and are essential for catalysis. In silico analyses were utilized to identify the putative effects of these mutations. In addition, coarse-grained models of both wild-type (WT) PTEN and mutants were constructed using elastic network models to explore the interplay of the structural and global dynamic effects that the mutations have on the relationship between genotype and phenotype. The effects of the mutations reveal that the local structure and interactions affect polarity, protein structure stability, electrostatic surface potential, and global dynamics of the protein. Our results offer new insight into the role in which PTEN missense mutations contribute to the molecular mechanism and genotypic-phenotypic correlation of endometriosis, endometrial cancer, and ovarian cancer. Proteins 2016; 84:1625-1643. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iris N Smith
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, 77204-5001
| | - James M Briggs
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, 77204-5001.
| |
Collapse
|
12
|
Chia YCJ, Catimel B, Lio DSS, Ang CS, Peng B, Wu H, Zhu HJ, Cheng HC. The C-terminal tail inhibitory phosphorylation sites of PTEN regulate its intrinsic catalytic activity and the kinetics of its binding to phosphatidylinositol-4,5-bisphosphate. Arch Biochem Biophys 2015; 587:48-60. [PMID: 26471078 DOI: 10.1016/j.abb.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/04/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
Abstract
Dephosphorylation of four major C-terminal tail sites and occupancy of the phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2]-binding site of PTEN cooperate to activate its phospholipid phosphatase activity and facilitate its recruitment to plasma membrane. Our investigation of the mechanism by which phosphorylation of these C-terminal sites controls the PI(4,5)P2-binding affinity and catalytic activity of PTEN resulted in the following findings. First, dephosphorylation of all four sites leads to full activation; and phosphorylation of any one site significantly reduces the intrinsic catalytic activity of PTEN. These findings suggest that coordinated inhibition of the upstream protein kinases and activation of the protein phosphatases targeting the four sites are needed to fully activate PTEN phosphatase activity. Second, PI(4,5)P2 cannot activate the phosphopeptide phosphatase activity of PTEN, suggesting that PI(4,5)P2 can only activate the phospholipid phosphatase activity but not the phosphoprotein phosphatase activity of PTEN. Third, dephosphorylation of all four sites significantly decreases the affinity of PTEN for PI(4,5)P2. Since PI(4,5)P2 is a major phospholipid co-localizing with the phospholipid- and phosphoprotein-substrates in plasma membrane, we hypothesise that the reduced affinity facilitates PTEN to "hop" on the plasma membrane to dephosphorylate these substrates.
Collapse
Affiliation(s)
- Yeong-Chit Joel Chia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Bruno Catimel
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Daisy Sio Seng Lio
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Benjamin Peng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hong Wu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hong-Jian Zhu
- Department of Surgery, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria 3052, Australia
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
13
|
Abstract
Phosphatases are a heterogeneous group of enzymes catalyzing dephosphorylation of diverse substrates ranging from small organic molecules to large phosphorylated multiprotein complexes. A wide variety of biochemical approaches for measuring phosphatase activity exists. Spectrophotometric methods utilizing artificial chromogenic, fluorogenic, and luminogenic substrates and taking advantage of the optical properties of dephosphorylated products are broadly used by research community. Another major assay type is based on quantitation of the second product of any phosphatase reactions, inorganic phosphate, using a variety of phosphate detection methods. Although, in theory, compatible with any phosphatase substrate, these assays often are unable to provide acceptable high-throughput screening adaptations of native phosphatase reactions. Conversely, phosphatase assays with artificial substrates frequently are incapable to mirror the intricacies of substrate binding and catalysis of the native reaction and, as a result, unable to deliver biologically relevant phosphatase modulators. Utilization of comprehensive phosphatase assay panels, employing honed biochemical assays and cell-based model systems, in conjunction with novel approaches for screening phosphatases may aid in identification of potent, selective, and biologically active phosphatase modulators.
Collapse
|
14
|
Phin S, Moore MW, Cotter PD. Genomic Rearrangements of PTEN in Prostate Cancer. Front Oncol 2013; 3:240. [PMID: 24062990 PMCID: PMC3775430 DOI: 10.3389/fonc.2013.00240] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/30/2013] [Indexed: 12/21/2022] Open
Abstract
The phosphatase and tensin homolog gene (PTEN) on chromosome 10q23.3 is a negative regulator of the PIK3/Akt survival pathway and is the most frequently deleted tumor suppressor gene in prostate cancer. Monoallelic loss of PTEN is present in up to 60% of localized prostate cancers and complete loss of PTEN in prostate cancer is linked to metastasis and androgen-independent progression. Studies on the genomic status of PTEN in prostate cancer initially used a two-color fluorescence in situ hybridization (FISH) assay for PTEN copy number detection in formalin fixed paraffin embedded tissue preparations. More recently, a four-color FISH assay containing two additional control probes flanking the PTEN locus with a lower false-positive rate was reported. Combined with the detection of other critical genomic biomarkers for prostate cancer such as ERG, androgen receptor, and MYC, the evaluation of PTEN genomic status has proven to be invaluable for patient stratification and management. Although less frequent than allelic deletions, point mutations in the gene and epigenetic silencing are also known to contribute to loss of PTEN function, and ultimately to prostate cancer initiation. Overall, it is clear that PTEN is a powerful biomarker for prostate cancer. Used as a companion diagnostic for emerging therapeutic drugs, FISH analysis of PTEN is promisingly moving human prostate cancer closer to more effective cancer management and therapies.
Collapse
|
15
|
Rodríguez-Escudero I, Oliver MD, Andrés-Pons A, Molina M, Cid VJ, Pulido R. A comprehensive functional analysis of PTEN mutations: implications in tumor- and autism-related syndromes. Hum Mol Genet 2011; 20:4132-42. [PMID: 21828076 DOI: 10.1093/hmg/ddr337] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The PTEN (phosphatase and tensin homolog) phosphatase is unique in mammals in terms of its tumor suppressor activity, exerted by dephosphorylation of the lipid second messenger PIP(3) (phosphatidylinositol 3,4,5-trisphosphate), which activates the phosphoinositide 3-kinase/Akt/mTOR (mammalian target of rapamycin) oncogenic pathway. Loss-of-function mutations in the PTEN gene are frequent in human cancer and in the germline of patients with PTEN hamartoma tumor-related syndromes (PHTSs). In addition, PTEN is mutated in patients with autism spectrum disorders (ASDs), although no functional information on these mutations is available. Here, we report a comprehensive in vivo functional analysis of human PTEN using a heterologous yeast reconstitution system. Ala-scanning mutagenesis at the catalytic loops of PTEN outlined the critical role of residues within the P-catalytic loop for PIP(3) phosphatase activity in vivo. PTEN mutations that mimic the P-catalytic loop of mammalian PTEN-like proteins (TPTE, TPIP, tensins and auxilins) affected PTEN function variably, whereas tumor- or PHTS-associated mutations targeting the PTEN P-loop produced complete loss of function. Conversely, Ala-substitutions, as well as tumor-related mutations at the WPD- and TI-catalytic loops, displayed partial activity in many cases. Interestingly, a tumor-related D92N mutation was partially active, supporting the notion that the PTEN Asp92 residue might not function as the catalytic general acid. The analysis of a panel of ASD-associated hereditary PTEN mutations revealed that most of them did not substantially abrogate PTEN activity in vivo, whereas most of PHTS-associated mutations did. Our findings reveal distinctive functional patterns among PTEN mutations found in tumors and in the germline of PHTS and ASD patients, which could be relevant for therapy.
Collapse
Affiliation(s)
- Isabel Rodríguez-Escudero
- Departamento de Microbiología II, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto Ramón y Cajal de Investigaciones Sanitarias, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|