1
|
Rajala A, Rajala R, Bhat MA, Eminhizer M, Hao J, Du J, Rajala RVS. Age-related retinal degeneration resulting from the deletion of Shp2 tyrosine phosphatase in photoreceptor neurons. Cell Death Dis 2024; 15:577. [PMID: 39117618 PMCID: PMC11310310 DOI: 10.1038/s41419-024-06924-y] [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: 01/08/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024]
Abstract
Shp2, a critical SH2-domain-containing tyrosine phosphatase, is essential for cellular regulation and implicated in metabolic disruptions, obesity, diabetes, Noonan syndrome, LEOPARD syndrome, and cancers. This study focuses on Shp2 in rod photoreceptor cells, revealing its enrichment, particularly in rods. Deletion of Shp2 in rods leads to age-dependent photoreceptor degeneration. Shp2 targets occludin (OCLN), a tight junction protein, and its deletion reduces OCLN expression in the retina and retinal pigment epithelium (RPE). The isolation of actively translating mRNAs from rods lacking Shp2, followed by RNA sequencing, reveals alterations in cell cycle regulation. Additionally, altered retinal metabolism is observed in retinal cells lacking Shp2. Our studies indicate that Shp2 is crucial for maintaining the structure and function of photoreceptors.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | - Rahul Rajala
- Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Mohd A Bhat
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA
| | - Mark Eminhizer
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26505, USA
| | - Jeff Hao
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26505, USA
| | - Jianhai Du
- Departments of Ophthalmology and Visual Sciences and Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, 26505, USA
| | - Raju V S Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Dean McGee Eye Institute, Oklahoma City, OK, 73104, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
2
|
Bai L, Tani T, Kobayashi T, Nouda R, Kanai Y, Sano Y, Takami K, Tomita H, Sugano E, Ozaki T, Kiyono T, Fukuda T. Establishment of immortalized Egyptian Rousettus bat cell lines. FEBS Open Bio 2024; 14:598-612. [PMID: 38373743 PMCID: PMC10988675 DOI: 10.1002/2211-5463.13781] [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: 10/03/2023] [Revised: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 02/21/2024] Open
Abstract
The Egyptian Rousettus bat (Rousettus aegyptiacus) is a common fruit bat species that is distributed mainly in Africa and the Middle East. Bats serve as reservoir hosts for numerous pathogens. Human activities, such as hunting bats for food, managing vermin, and causing habitat loss, elevate the likelihood of transmission of bat pathogens to humans and other animals. Consequently, bat cell lines play a crucial role as research materials for investigating viral pathogens. However, the inherent limitation of finite cell division in primary cells necessitates the use of immortalized cells derived from various bat tissues. Herein, we successfully established six fibroblast cell lines derived from an infant bat heart and lungs and an elderly bat heart. Three of the six cell lines, called K4DT cells, were transduced by a combination of cell cycle regulators, mutant cyclin-dependent kinase 4, cyclin D1, and human telomerase reverse transcriptase. The other three cell lines, named SV40 cells, were transfected with simian virus 40 large T antigen. Transgene protein expression was detected in the transduced cells. All three K4DT cell lines and one lung-derived SV40 cell line were virtually immortalized and nearly maintained the normal diploid karyotypes. However, the two other heart-derived SV40 cell lines had aberrant karyotypes and the young bat-derived cell line stopped proliferating at approximately 40 population doublings. These bat cell lines are valuable for studying pathogen genomics and biology.
Collapse
Affiliation(s)
- Lanlan Bai
- Graduate School of Science and EngineeringIwate UniversityJapan
| | - Tetsuya Tani
- Laboratory of Animal Reproduction, Department of AgricultureKindai UniversityNaraJapan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial DiseasesOsaka UniversityJapan
| | - Ryotaro Nouda
- Department of Virology, Research Institute for Microbial DiseasesOsaka UniversityJapan
| | - Yuta Kanai
- Department of Virology, Research Institute for Microbial DiseasesOsaka UniversityJapan
| | - Yusuke Sano
- Local Independent Administrative Agency Tennoji Zoological GardensOsakaJapan
| | - Kazutoshi Takami
- Osaka Municipal Tennoji Zoological GardensJapan
- Present address:
*Toyohashi Zoo and Botanical ParkToyohashiJapan
| | - Hiroshi Tomita
- Graduate School of Science and EngineeringIwate UniversityJapan
| | - Eriko Sugano
- Graduate School of Science and EngineeringIwate UniversityJapan
| | - Taku Ozaki
- Graduate School of Science and EngineeringIwate UniversityJapan
| | - Tohru Kiyono
- Exploratory Oncology Research & Clinical Trial CenterNational Cancer CenterChibaJapan
| | - Tomokazu Fukuda
- Graduate School of Science and EngineeringIwate UniversityJapan
| |
Collapse
|
3
|
Chen X, Keller SJ, Hafner P, Alrawashdeh AY, Avery TY, Norona J, Zhou J, Ruess DA. Tyrosine phosphatase PTPN11/SHP2 in solid tumors - bull's eye for targeted therapy? Front Immunol 2024; 15:1340726. [PMID: 38504984 PMCID: PMC10948527 DOI: 10.3389/fimmu.2024.1340726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Encoded by PTPN11, the Src-homology 2 domain-containing phosphatase 2 (SHP2) integrates signals from various membrane-bound receptors such as receptor tyrosine kinases (RTKs), cytokine and integrin receptors and thereby promotes cell survival and proliferation. Activating mutations in the PTPN11 gene may trigger signaling pathways leading to the development of hematological malignancies, but are rarely found in solid tumors. Yet, aberrant SHP2 expression or activation has implications in the development, progression and metastasis of many solid tumor entities. SHP2 is involved in multiple signaling cascades, including the RAS-RAF-MEK-ERK-, PI3K-AKT-, JAK-STAT- and PD-L1/PD-1- pathways. Although not mutated, activation or functional requirement of SHP2 appears to play a relevant and context-dependent dichotomous role. This mostly tumor-promoting and infrequently tumor-suppressive role exists in many cancers such as gastrointestinal tumors, pancreatic, liver and lung cancer, gynecological entities, head and neck cancers, prostate cancer, glioblastoma and melanoma. Recent studies have identified SHP2 as a potential biomarker for the prognosis of some solid tumors. Based on promising preclinical work and the advent of orally available allosteric SHP2-inhibitors early clinical trials are currently investigating SHP2-directed approaches in various solid tumors, either as a single agent or in combination regimes. We here provide a brief overview of the molecular functions of SHP2 and collate current knowledge with regard to the significance of SHP2 expression and function in different solid tumor entities, including cells in their microenvironment, immune escape and therapy resistance. In the context of the present landscape of clinical trials with allosteric SHP2-inhibitors we discuss the multitude of opportunities but also limitations of a strategy targeting this non-receptor protein tyrosine phosphatase for treatment of solid tumors.
Collapse
Affiliation(s)
- Xun Chen
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Steffen Johannes Keller
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Philipp Hafner
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Asma Y. Alrawashdeh
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Thomas Yul Avery
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Johana Norona
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Dietrich Alexander Ruess
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Notarstefano V, Belloni A, Mariani P, Orilisi G, Orsini G, Giorgini E, Byrne HJ. Multivariate curve Resolution-Alternating least squares coupled with Raman microspectroscopy: new insights into the kinetic response of primary oral squamous carcinoma cells to cisplatin. Analyst 2023; 148:4365-4372. [PMID: 37548234 DOI: 10.1039/d3an01182h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Raman MicroSpectroscopy (RMS) is a powerful label-free tool to probe the effects of drugs at a cellular/subcellular level. It is important, however, to be able to extract relevant biochemical and kinetic spectroscopic signatures of the specific cellular responses. In the present study, a combination of Multivariate Curve Resolution-Alternating Least Squares (MCR-ALS) and Principal Component Analysis (PCA) is used to analyse the RMS data for the example of exposure of primary Oral Squamous Carcinoma Cells (OSCC) to the chemotherapeutic agent cisplatin. Dosing regimens were established by cytotoxicity assays, and the effects of the drug on cellular spectral profiles were monitored from 16 to 72 hours post-exposure using an apoptosis assay, to establish the relative populations of viable (V), early (EA) and late apoptotic/dead (LA/D) cells after the drug treatment. Based on a kinetic model of the progression from V > EA > D, MCR-ALS regression analysis of the RMS responses was able to extract spectral profiles associated with each stage of the cellular responses, enabling a quantitative comparison of the response rates for the respective drug treatments. Moreover, PCA was used to compare the spectral profiles of the viable cells exposed to the drug. Spectral differences were highlighted in the early stages (16 hours exposure), indicative of the initial cellular response to the drug treatment, and also in the late stages (48-72 hours exposure), representing the cell death pathway. The study demonstrates that RMS coupled with multivariate analysis can be used to quantitatively monitor the progression of cellular responses to different drugs, towards future applications for label-free, in vitro, pre-clinical screening.
Collapse
Affiliation(s)
- Valentina Notarstefano
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Alessia Belloni
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Paolo Mariani
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Giulia Orilisi
- Department of Clinical Sciences and Stomatology, Università Politecnica delle Marche, Via Brecce Bianche, 60126 Ancona, Italy
| | - Giovanna Orsini
- Department of Clinical Sciences and Stomatology, Università Politecnica delle Marche, Via Brecce Bianche, 60126 Ancona, Italy
| | - Elisabetta Giorgini
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy.
| | - Hugh J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Dublin, Ireland
| |
Collapse
|
5
|
Chemoresistant Cancer Cell Lines Are Characterized by Migratory, Amino Acid Metabolism, Protein Catabolism and IFN1 Signalling Perturbations. Cancers (Basel) 2022; 14:cancers14112763. [PMID: 35681748 PMCID: PMC9179525 DOI: 10.3390/cancers14112763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary While chemoresistance remains a major barrier to improving the outcomes for patients with ovarian cancer, the molecular features, and associated biological functions, which underpin chemoresistance in ovarian cancer remain poorly understood. In this study we aimed to provide insight into the proteins and metabolites, and their associated biological pathways, which play a role in conferring chemoresistance to ovarian cancer. Through mass spectrometry analysis comparing the proteome and metabolome of chemosensitive vs chemoresistant ovarian cancer cell lines we revealed numerous perturbations in signalling and metabolic pathways in chemoresistant cells. Further comparison to primary cells taken from patients with chemoresistant or chemosensitive disease identified a shared dysregulation in cytokine and type 1 interferon signalling. Our research sets the foundation for a deeper understanding of the proteomic and metabolomic features of chemoresistance and identifies type 1 interferon signalling as a common feature of chemoresistance. Abstract Chemoresistance remains the major barrier to effective ovarian cancer treatment. The molecular features and associated biological functions of this phenotype remain poorly understood. We developed carboplatin-resistant cell line models using OVCAR5 and CaOV3 cell lines with the aim of identifying chemoresistance-specific molecular features. Chemotaxis and CAM invasion assays revealed enhanced migratory and invasive potential in OVCAR5-resistant, compared to parental cell lines. Mass spectrometry analysis was used to analyse the metabolome and proteome of these cell lines, and was able to separate these populations based on their molecular features. It revealed signalling and metabolic perturbations in the chemoresistant cell lines. A comparison with the proteome of patient-derived primary ovarian cancer cells grown in culture showed a shared dysregulation of cytokine and type 1 interferon signalling, potentially revealing a common molecular feature of chemoresistance. A comprehensive analysis of a larger patient cohort, including advanced in vitro and in vivo models, promises to assist with better understanding the molecular mechanisms of chemoresistance and the associated enhancement of migration and invasion.
Collapse
|
6
|
Notarstefano V, Sabbatini S, Pro C, Belloni A, Orilisi G, Rubini C, Byrne HJ, Vaccari L, Giorgini E. Exploiting fourier transform infrared and Raman microspectroscopies on cancer stem cells from oral squamous cells carcinoma: new evidence of acquired cisplatin chemoresistance. Analyst 2021; 145:8038-8049. [PMID: 33063801 DOI: 10.1039/d0an01623c] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oral Squamous Cells Carcinoma (OSCC) is characterised by the risk of recurrence and the onset of a refractoriness response to chemotherapy drugs. These phenomena have been recently related to a subpopulation of Cancer Stem Cells (CSCs), which have either an innate or acquired drug resistance, triggered by chemotherapy treatments. In this light, to precisely target chemotherapy regimens, it is essential to improve knowledge on CSCs, with a particular focus on their molecular features. In this work, a subpopulation of CSCs, isolated by tumour sphere formation from primary OSCC cells, were treated with cisplatin for 16, 24 and 48 hours and analysed by infrared absorption and Raman microspectroscopies. CSC spectral data were compared with those obtained in previous work, for primary OSCC cells treated under the same conditions. Routine viability/apoptosis cell-based assays evidenced in CSCs and primary OSCCs, a similar degree of sensitivity to the drug at 24 hours, while a reversion of the conventional monotonic time response exhibited by OSCCs was shown by CSCs at 48 hours. This peculiar time response was also supported by the analysis of IR and Raman data, which pinpointed alterations in the lipid composition and DNA conformation in CSCs. The results obtained suggest that CSCs, although sharing with OSCC cells a similar sensitivity to cisplatin, display the onset of a mechanism of chemoresistance and enrichment of resistant CSCs as a result of drug treatment, shedding new light on the severe issue of refractoriness of some patients to chemotherapy conventionally used for OSCC.
Collapse
Affiliation(s)
- Valentina Notarstefano
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Identification of new proteins related with cisplatin resistance in Saccharomyces cerevisiae. Appl Microbiol Biotechnol 2021; 105:1965-1977. [PMID: 33576883 DOI: 10.1007/s00253-021-11137-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 10/22/2022]
Abstract
The aim of this study is to select a cisplatin-resistant Saccharomyces cerevisiae strain to look for new molecular markers of resistance and the identification of mechanisms/interactions involved. A resistant strain was obtained after 80 days of cisplatin exposure. Then, total protein extraction, purification, and identification were carried out, in wild-type (wt) and resistant strains, by tandem mass spectrometry using a "nano HPLC-ESI-MS/MS" ion trap system. The increase in the exponentially modified protein abundance index (emPAI) (resistant vs wt strains) was calculated to study the increase in protein expression. "Genemania" software ( http://www.Genemania.org/ ) was used to compare the effects, functions, and protein interactions. KEGG tool was used for metabolic pathway analysis. Data are available via ProteomeXchange with identifier PXD020665. The cisplatin-resistant strain showed 2.5 times more resistance than the wt strain for the inhibitory dose 50% (ID50) value (224 μg/ml vs 89.68 μg/ml) and 2.78 times more resistant for the inhibitory dose 90% (ID90) value (735.2 μg/ml vs 264.04 μg/ml). Multiple deregulated proteins were found in the glutathione and carbon metabolism, oxidative phosphorylation, proteasome, glycolysis and gluconeogenesis, glyoxylate metabolism, fatty acid degradation pathway, citric acid cycle, and ribosome. The most overexpressed proteins in the cisplatin-resistant strain were related to growth and metabolism (QCR2, QCR1, ALDH4, ATPB, ATPA, ATPG, and PCKA), cell structure (SCW10), and thermal shock (HSP26). The results suggest that these proteins could be involved in cisplatin resistance. The resistance acquisition process is complex and involves the activation of multiple mechanisms that interact together. KEY POINTS: • Identification of new proteins/genes related to cisplatin resistance • Increased expression of QCR2/QCR1/ALDH4/ATPB/ATPA/SCW10/HSP26/ATPG and PCKA proteins • Multiple molecular mechanisms that interact together are involved in resistance.
Collapse
|
8
|
Tripathi RKP, Ayyannan SR. Emerging chemical scaffolds with potential SHP2 phosphatase inhibitory capabilities - A comprehensive review. Chem Biol Drug Des 2020; 97:721-773. [PMID: 33191603 DOI: 10.1111/cbdd.13807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
The drug discovery panorama is cluttered with promising therapeutic targets that have been deserted because of inadequate authentication and screening failures. Molecular targets formerly tagged as "undruggable" are nowadays being more cautiously cross-examined, and whilst they stay intriguing, numerous targets are emerging more accessible. Protein tyrosine phosphatases (PTPs) excellently exemplifies a class of molecular targets that have transpired as druggable, with several small molecules and antibodies recently turned available for further development. In this respect, SHP2, a PTP, has emerged as one of the potential targets in the current pharmacological research, particularly for cancer, due to its critical role in various signalling pathways. Recently, few molecules with excellent potency have entered clinical trials, but none could reach the clinic. Consequently, search for novel, non-toxic, and specific SHP2 inhibitors are on purview. In this review, general aspects of SHP2 including its structure and mechanistic role in carcinogenesis have been presented. It also sheds light on the development of novel molecular architectures belonging to diverse chemical classes that have been proposed as SHP2-specific inhibitors along with their structure-activity relationships (SARs), stemming from chemical, mechanism-based and computer-aided studies reported since January 2015 to July 2020 (excluding patents), focusing on their potency and selectivity. The encyclopedic facts and discussions presented herein will hopefully facilitate researchers to design new ligands with better efficacy and selectivity against SHP2.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Department of Pharmaceutical Science, Sushruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar, Assam, India.,Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
9
|
Wang R, Lu X, Yu R. lncRNA MALAT1 Promotes EMT Process and Cisplatin Resistance of Oral Squamous Cell Carcinoma via PI3K/AKT/m-TOR Signal Pathway. Onco Targets Ther 2020; 13:4049-4061. [PMID: 32494159 PMCID: PMC7231756 DOI: 10.2147/ott.s251518] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/22/2020] [Indexed: 11/29/2022] Open
Abstract
Background Cisplatin (DDP) is the first-line chemotherapy agent for the treatment of oral squamous cell carcinoma (OSCC). The emergence of DDP resistance leads to diminished drug efficacy and survival benefit. lncRNA MALAT1 has been considered as one of the most important factors in OSCC. It has also been reported to enhance chemo-resistance in other kinds of carcinomas. However, little is known about the role of lncRNA MALAT1 in DDP resistance of OSCC. Materials and Methods Two kinds of human DDP-resistant cell lines (CAL-27R and SCC-9R) were developed from cisplatin-naïve cell lines (CAL-27 and SCC-9, respectively) as in vitro cell models. Cell transfection was performed to overexpress or knockdown MALAT1 in these cells. Mouse xenograft models were also established. The following measurements were performed: cell proliferation, colony formation, wound healing, transwell, and TUNEL assays, as well as Western blot and immunofluorescence staining. Results DDP-resistant cells showed higher expression level of MALAT1 compared to cisplatin-naïve cells. The overexpression of MALAT1 in cisplatin-naïve cells enhanced DDP resistance and suppressed apoptosis in OSCC cells. However, the knockdown of MALAT1 in DDP-resistance cells induced apoptotic cell death and restored the sensitivity to DDP. Further analyses suggested that MALAT1 might promote DDP resistance via regulating P-glycoprotein expression, epithelial–mesenchymal transition process, and the activation of PI3K/AKT/m-TOR signaling pathway. Conclusion MALAT1 might be a potential therapeutic target for the treatment of DDP-resistant OSCC.
Collapse
Affiliation(s)
- Ran Wang
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinxing Lu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Riyue Yu
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
10
|
Chen J, Chen Z, Huang Z, Yu H, Li Y, Huang W. Formiminotransferase Cyclodeaminase Suppresses Hepatocellular Carcinoma by Modulating Cell Apoptosis, DNA Damage, and Phosphatidylinositol 3-Kinases (PI3K)/Akt Signaling Pathway. Med Sci Monit 2019; 25:4474-4484. [PMID: 31203308 PMCID: PMC6592141 DOI: 10.12659/msm.916202] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Formiminotransferase cyclodeaminase (FTCD) is a candidate tumor suppressor gene in hepatocellular carcinoma (HCC). However, the mechanism for reduced expression of FTCD and its functional role in HCC remains unclear. In this study, we explored the biological functions of FTCD in HCC. Material/Methods The expression and clinical correlation of FTCD in HCC tissue were analyzed using TCGA (The Cancer Genome Atlas) and a cohort of 60 HCC patients. The MEXPRESS platform was accessed to identify the methylation level in promoter region FTCD. CCK-8 assay and flow cytometry analysis were used to explore the proliferation, cell apoptosis proportion, and DNA damage in HCC cells with FTCD overexpression. Western blot analysis was performed to identify the downstream target of FTCD. Results FTCD is significantly downregulated in HCC tissues and cell lines. Low FTCD expression is correlated with a poor prognosis (P<0.001) and an aggressive tumor phenotype, including AFP levels (P=0.009), tumor size (P=0.013), vascular invasion (P=0.001), BCLC stage (P=0.024), and pTNM stage (P<0.001). Bioinformatics analysis indicated promoter hypermethylation can result in decreased expression of FTCD. FTCD overexpression suppressed cell proliferation by promoting DNA damage and inducing cell apoptosis in HCC cells. FTCD overexpression resulted in increased level of PTEN protein, but a decrease in PI3K, total Akt, and phosphorylated Akt protein in HCC cells, suggesting involvement of the PI3K/Akt pathway. Conclusions FTCD acts as a tumor suppressor gene in HCC pathogenesis and progression and is a candidate prognostic marker and a possible therapeutic target for this disease.
Collapse
Affiliation(s)
- Jiajia Chen
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Department of General Surgery, Affiliated Chaozhou Central Hospital, Southern Medical University, Chaozhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Zemian Chen
- Department of Medical Oncology, Affiliated Chaozhou Central Hospital, Southern Medical University, Chaozhou, Guangdong, China (mainland)
| | - Zhentian Huang
- Department of General Surgery, Affiliated Chaozhou Central Hospital, Southern Medical University, Chaozhou, Guangdong, China (mainland)
| | - Hongrong Yu
- Department of Human Anatomy, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yanbing Li
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| | - Wenhua Huang
- National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland).,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
11
|
Aghaalikhani N, Rashtchizadeh N, Shadpour P, Allameh A, Mahmoodi M. Cancer stem cells as a therapeutic target in bladder cancer. J Cell Physiol 2018; 234:3197-3206. [PMID: 30471107 DOI: 10.1002/jcp.26916] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Bladder cancer is one of the most prevalent genitourinary cancers responsible for about 150,000 deaths per year worldwide. Currently, several treatments, such as endoscopic and open surgery, appended by local or systemic immunotherapy, chemotherapy, and radiotherapy are used to treat this malignancy. However, the differences in treatment outcome among patients suffering from bladder cancer are considered as one of the important challenges. In recent years, cancer stem cells, representing a population of undifferentiated cells with stem-cell like properties, have been eyed as a major culprit for the high recurrence rate in superficial papillary bladder cancer. Cancer stem cells have been reported to be resistant to conventional treatments, such as chemotherapy, radiation, and immunotherapy, which induce selective pressure on tumoral populations resulting in selection and growth of the resistant cells. Therefore, targeting the therapeutic aspects of cancer stem cells in bladder cancer may be promising. In this study, we briefly discuss the biology of bladder cancer and then address the possible relationship between molecular biology of bladder cancer and cancer stem cells. Subsequently, the mechanisms of resistance applied by cancer stem cells against the conventional therapeutic tools, especially chemotherapy, are discussed. Moreover, by emphasizing the biomarkers described for cancer stem cells in bladder cancer, we have provided, described, and proposed targets on cancer stem cells for therapeutic interventions and, finally, reviewed some immunotargeting strategies against bladder cancer stem cells.
Collapse
Affiliation(s)
- Nazi Aghaalikhani
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadereh Rashtchizadeh
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pejman Shadpour
- Hasheminejad Kidney Centre (HKC), Hospital Management Research Centre (HMRC), University of Medical Sciences (IUMS), Tehran, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marzieh Mahmoodi
- Faculty of Health and Nutrition, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
12
|
Takahashi S. Molecular functions of SIRPα and its role in cancer. Biomed Rep 2018; 9:3-7. [PMID: 29930800 DOI: 10.3892/br.2018.1102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/21/2018] [Indexed: 01/17/2023] Open
Abstract
Signal regulatory protein α (SIRPα), also known as cluster of differentiation (CD)172a or Src homology 2 domain-containing phosphatase substrate-1, is a cell surface receptor expressed on myeloid and hematopoietic stem cells and neurons. Accumulating data suggests an important role of SIRPα in cell signaling as a negative regulator of the phosphatidylinositol 3-kinase signaling and mitogen-activated protein kinase pathways. In various cancers, including prostate, breast and liver, as well as astrocytoma and myeloid malignancies, downregulation of SIRPα is frequently observed, resulting in activation of these downstream signaling pathways. In turn, cell proliferation, transformation, migration and invasion may occur. Recently, it has been reported that blocking CD47, an anti-phagocytic signal expressed on tumor cells and an SIRPα ligand, may serve as a promising therapeutic approach, particular for the treatment of acute myeloid leukemia. In the present review, the current findings on SIRPα are summarized, with particular focus on its role in cancer.
Collapse
Affiliation(s)
- Shinichiro Takahashi
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagino-ku, Sendai 983-8536, Japan
| |
Collapse
|
13
|
Busch M, Papior D, Stephan H, Dünker N. Characterization of etoposide- and cisplatin-chemoresistant retinoblastoma cell lines. Oncol Rep 2017; 39:160-172. [PMID: 29192327 PMCID: PMC5783599 DOI: 10.3892/or.2017.6100] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022] Open
Abstract
Retinoblastoma (RB) is the most common malignant intraocular tumor in early childhood. Imminent chemotherapy resistance diminishes the clinical-therapeutic options and emphasizes the necessity for new therapeutic approaches. The present study aimed at characterizing and comparing etoposide and cisplatin-resistant human RB cell lines with regard to changes in proliferation and apoptosis levels, anchorage independent growth behavior in vitro as well as tumor formation capacity in vivo. The proliferation rates were significantly increased in the etoposide-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 reflecting significantly higher growth kinetics compared to the parental controls. In line with these findings in in vivo chicken chorioallantoic (CAM) assays, etoposide-resistant cell lines generated significantly increased numbers of tumors with higher tumor weights compared to their parental counterparts. In contrast to etoposide, the cisplatin-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 displayed significantly increased apoptosis rates and reduced proliferation rates resulting in significantly decreased growth kinetics. Tumor formation capacity of cisplatin-resistant cell lines did not significantly change, and in comparison with parental controls cisplatin-resistant Y-79 cells displayed significantly reduced tumor weight. Soft agarose assays indicated that anchorage-independent growth of all chemotherapy-resistant cell lines analyzed was significantly decreased. Summarizing, one can state that etoposide-resistant RB cells behave more aggressively than the tumor cells of origin and potentially represent a risk factor for local relapse, while cisplatin-resistant cells show a significantly decreased tumorigenic potential.
Collapse
Affiliation(s)
- Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| | - David Papior
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Harald Stephan
- Division of Haematology and Oncology, Children's Hospital, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| |
Collapse
|
14
|
Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, Santos LL. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist Updat 2016; 24:34-54. [DOI: 10.1016/j.drup.2015.11.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023]
|
15
|
Peng G, Cao RB, Li YH, Zou ZW, Huang J, Ding Q. Alterations of cell cycle control proteins SHP‑1/2, p16, CDK4 and cyclin D1 in radioresistant nasopharyngeal carcinoma cells. Mol Med Rep 2014; 10:1709-16. [PMID: 25109634 PMCID: PMC4148372 DOI: 10.3892/mmr.2014.2463] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 05/21/2014] [Indexed: 12/12/2022] Open
Abstract
The primary treatment for nasopharyngeal carcinoma (NPC) is radiotherapy, with or without concurrent chemotherapy. However, resistance to radiotherapy is not uncommon. The aim of the present study was to establish a radioresistant NPC cell line to study the molecular mechanisms of radioresistance by measuring the expression of cell cycle control proteins src homology 2 domain-containing phosphatase (SHP)-1/2, p16, CDK4 and cyclin D1. Human nasopharyngeal carcinoma CNE-2 cells were cultured, divided into two groups (CNE-2S1 and CNE-2S2) and irradiated with a dose of 6 Gy x5 or 2 Gy x15, respectively. The cells were subcultured between doses of irradiation. The surviving sublines (CNE-2S1 and CNE-2S2 clones) were then passaged for three months and their radiosensitivity was determined. The cell cycle distribution and protein expression of SHP-1/2, p16, CDK4 and cyclin D1 in parental and progenitor cell lines were measured. Small interfering (si)RNA-mediated knockdown of SHP-1 and SHP-2 in the NPC cells was used to further examine their roles in radiosensitivity and cell cycle distribution. CNE-2S1, a radio-resistant cell line, had a significantly higher percentage of cells in S phase and a lower percentage of cells in G1 phase, enhanced expression levels of SHP-1, CDK4 and cyclin D1, and reduced expression of p16, respectively, as compared with the parent cells. Stable suppression of SHP-1 mRNA in CNE-2 cells resulted in increased radiosensitivity compared with the parental cells, a decrease in the number of cells in S phase and an increase in the expression of p16. The results suggested that the SHP-1/p16/cyclin D1/CDK4 pathway may have a role in regulating radiosensitivity and cell cycle distribution in nasopharyngeal cells.
Collapse
Affiliation(s)
- Gang Peng
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ru-Bo Cao
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yue-Hua Li
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhen-Wei Zou
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Huang
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qian Ding
- Department of Head and Neck Cancer, Cancer Center of Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
16
|
Schleimann MH, Hoberg S, Solhøj Hansen A, Bundgaard B, Witt CT, Kofod-Olsen E, Höllsberg P. The DR6 protein from human herpesvirus-6B induces p53-independent cell cycle arrest in G2/M. Virology 2014; 452-453:254-63. [PMID: 24606703 DOI: 10.1016/j.virol.2014.01.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/09/2014] [Accepted: 01/30/2014] [Indexed: 11/24/2022]
Abstract
HHV-6B infection inhibits cell proliferation in G2/M, but no protein has so far been recognized to exert this function. Here we identify the protein product of direct repeat 6, DR6, as an inhibitor of G2/M cell-cycle progression. Transfection of DR6 reduced the total number of cells compared with mock-transfected cells. Lentiviral transduction of DR6 inhibited host cell DNA synthesis in a p53-independent manner, and this inhibition was DR6 dose-dependent. A deletion of 66 amino acids from the N-terminal part of DR6 prevented efficient nuclear translocation and the ability to inhibit DNA synthesis. DR6-induced accumulation of cells in G2/M was accompanied by an enhanced expression of cyclin B1 that accumulated predominantly in the cytoplasm. Pull-down of cyclin B1 brought down pCdk1 with the inactivating phosphorylation at Tyr15. Together, DR6 delays cell cycle with an accumulation of cells in G2/M and thus might be involved in HHV-6B-induced cell-cycle arrest.
Collapse
Affiliation(s)
| | - Søren Hoberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | | | | | - Per Höllsberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
17
|
Xu D, Zheng H, Yu WM, Qu CK. Activating mutations in protein tyrosine phosphatase Ptpn11 (Shp2) enhance reactive oxygen species production that contributes to myeloproliferative disorder. PLoS One 2013; 8:e63152. [PMID: 23675459 PMCID: PMC3651249 DOI: 10.1371/journal.pone.0063152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/29/2013] [Indexed: 12/25/2022] Open
Abstract
Gain of function (GOF) mutations in protein tyrosine phosphatase Ptpn11 have been identified in childhood leukemias, and these mutations are sufficient to drive the development of myeloproliferative disorder and malignant leukemias in mice. However, the molecular mechanisms by which Ptpn11 mutations induce these malignancies are not completely understood. Here we report that Ptpn11 GOF mutations cause cytokine hypersensitivity in hematopoietic cells partly by enhancing the production of reactive oxygen species (ROS). GOF mutations D61G or E76K in Ptpn11 increased ROS levels in myeloid progenitors but not in hematopoietic stem cells. Increased ROS enhanced cellular responses to cytokines by promoting cytokine signaling. Treatment with an antioxidant partially corrected cytokine hypersensitivity in Ptpn11 mutant progenitors. Further analyses demonstrated that Ptpn11 mutations increased mitochondrial aerobic metabolism by interacting with a novel substrate in the mitochondria. This study provides new insights into the pathogenic effects of GOF mutations of Ptpn11 and implies that antioxidants may have a therapeutic benefit for the leukemic patients with these mutations.
Collapse
Affiliation(s)
- Dan Xu
- Department of Medicine, Division of Hematology and Oncology, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Hong Zheng
- Department of Medicine, Division of Hematology and Oncology, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Wen-Mei Yu
- Department of Medicine, Division of Hematology and Oncology, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Cheng-Kui Qu
- Department of Medicine, Division of Hematology and Oncology, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
18
|
Tang S, Bai C, Yang P, Chen X. 14-3-3ε boosts bleomycin-induced DNA damage response by inhibiting the drug-resistant activity of MVP. J Proteome Res 2013; 12:2511-24. [PMID: 23590642 DOI: 10.1021/pr301085c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Major vault protein (MVP) is the predominant constituent of the vault particle, the largest known ribonuclear protein complex. Although emerging evidence have been establishing the links between MVP (vault) and multidrug resistance (MDR), little is known regarding exactly how the MDR activity of MVP is modulated during cellular response to drug-induced DNA damage (DDR). Bleomycin (BLM), an anticancer drug, induces DNA double-stranded breaks (DSBs) and consequently triggers the cellular DDR. Due to its physiological implications in hepatocellular carcinoma (HCC) and cell fate decision, 14-3-3ε was chosen as the pathway-specific bait protein to identify the critical target(s) responsible for HCC MDR. By using an LC-MS/MS-based proteomic approach, MVP was first identified in the BLM-induced 14-3-3ε interactome formed in HCC cells. Biological characterization revealed that MVP possesses specific activity to promote the resistance to the BLM-induced DDR. On the other hand, 14-3-3ε enhances BLM-induced DDR by interacting with MVP. Mechanistic investigation further revealed that 14-3-3ε, in a phosphorylation-dependent manner, binds to the phosphorylated sites at both Thr52 and Ser864 of the monomer of MVP. Consequently, the phosphorylation-dependent binding between 14-3-3ε and MVP inhibits the drug-resistant activity of MVP for an enhanced DDR to BLM treatment. Our findings provide an insight into the mechanism underlying how the BLM-induced interaction between 14-3-3ε and MVP modulates MDR, implicating novel strategy to overcome the chemotherapeutic resistance through interfering specific protein-protein interactions.
Collapse
Affiliation(s)
- Siwei Tang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
19
|
Tsang YH, Han X, Man WY, Lee N, Poon RYC. Novel functions of the phosphatase SHP2 in the DNA replication and damage checkpoints. PLoS One 2012. [PMID: 23189174 PMCID: PMC3506573 DOI: 10.1371/journal.pone.0049943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Replication stress- and DNA damage-induced cell cycle checkpoints are critical for maintaining genome stability. To identify protein phosphatases involved in the activation and maintenance of the checkpoints, we have carried out RNA interference-based screens with a human phosphatome shRNA library. Several phosphatases, including SHP2 (also called PTPN11) were found to be required for cell survival upon hydroxyurea-induced replicative stress in HeLa cells. More detailed studies revealed that SHP2 was also important for the maintenance of the checkpoint after DNA damage induced by cisplatin or ionizing radiation in HeLa cells. Furthermore, SHP2 was activated after replicative stress and DNA damage. Although depletion of SHP2 resulted in a delay in cyclin E accumulation and an extension of G1 phase, these cell cycle impairments were not responsible for the increase in apoptosis after DNA damage. Depletion of SHP2 impaired CHK1 activation, checkpoint-mediated cell cycle arrest, and DNA repair. These effects could be rescued with a shRNA-resistant SHP2. These results underscore the importance of protein phosphatases in checkpoint control and revealed a novel link between SHP2 and cell cycle checkpoints.
Collapse
Affiliation(s)
- Yiu Huen Tsang
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Xianxian Han
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Wing Yu Man
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Nelson Lee
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y. C. Poon
- Division of Life Science and Center for Cancer Research, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- * E-mail:
| |
Collapse
|
20
|
Liu X, Zheng H, Qu CK. Protein tyrosine phosphatase Shp2 (Ptpn11) plays an important role in maintenance of chromosome stability. Cancer Res 2012; 72:5296-306. [PMID: 22890240 DOI: 10.1158/0008-5472.can-12-1495] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Both activating and inactivating mutations in protein tyrosine phosphatase Ptpn11 (encoding Shp2) are associated with tumorigenesis. However, the underlying mechanisms remain unclear. Here, we show that Shp2 plays an important role in mitosis, dysregulation of which results in chromosome instability and cancer predisposition. Depletion of Shp2 compromised the mitotic checkpoint. Shp2-depleted cells exhibited a delay in mitotic entry and an earlier mitotic exit. Moreover, Shp2 deficiency caused defective kinetochore-microtubule attachment, chromosome misalignment, chromosomal congression defects, lagging chromosomes, and chromosome missegregation. Reintroduction of wild-type Shp2, but not a catalytically deficient mutant, restored the checkpoint function and chromosome alignment at metaphase in Shp2-deficient cells, establishing a requirement for the catalytic activity of Shp2 during mitosis. Further analyses revealed that Shp2 was required for the optimal activation of the mitotic kinases PLK1 and Aurora B and thereby the proper kinetochore localization and phosphorylation of BubR1, a core mitotic checkpoint protein that is also critical for chromosome alignment. Together, our findings show a previously unrecognized role for Shp2 in the maintenance of chromosome stability and suggest a new mechanism by which dysregulation of Shp2 signaling contributes to malignancy development.
Collapse
Affiliation(s)
- Xia Liu
- Division of Hematology-Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
21
|
Liu X, Qu CK. Protein Tyrosine Phosphatase SHP-2 (PTPN11) in Hematopoiesis and Leukemogenesis. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:195239. [PMID: 21799948 PMCID: PMC3135119 DOI: 10.1155/2011/195239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 04/01/2011] [Indexed: 01/28/2023]
Abstract
SHP-2 (PTPN11), a ubiquitously expressed protein tyrosine phosphatase, is critical for hematopoietic cell development and function owing to its essential role in growth factor/cytokine signaling. More importantly, germline and somatic mutations in this phosphatase are associated with Noonan syndrome, Leopard syndrome, and childhood hematologic malignancies. The molecular mechanisms by which SHP-2 mutations induce these diseases are not fully understood, as the biochemical bases of SHP-2 functions still remain elusive. Further understanding SHP-2 signaling activities and identification of its interacting proteins/substrates will shed light on the pathogenesis of PTPN11-associated hematologic malignancies, which, in turn, may lead to novel therapeutics for these diseases.
Collapse
Affiliation(s)
- Xia Liu
- Division of Hematology and Oncology, Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Cheng-Kui Qu
- Division of Hematology and Oncology, Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
22
|
Phosphatase-dependent and -independent functions of Shp2 in neural crest cells underlie LEOPARD syndrome pathogenesis. Dev Cell 2010; 18:750-62. [PMID: 20493809 DOI: 10.1016/j.devcel.2010.03.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Revised: 02/11/2010] [Accepted: 03/12/2010] [Indexed: 01/19/2023]
Abstract
The tyrosine phosphatase SHP2 (PTPN11) regulates cellular proliferation, survival, migration, and differentiation during development. Germline mutations in PTPN11 cause Noonan and LEOPARD syndromes, which have overlapping clinical features. Paradoxically, Noonan syndrome mutations increase SHP2 phosphatase activity, while LEOPARD syndrome mutants are catalytically impaired, raising the possibility that SHP2 has phosphatase-independent roles. By comparing shp2-deficient zebrafish embryos with those injected with mRNA encoding LEOPARD syndrome point mutations, we identify a phosphatase- and Erk-dependent role for Shp2 in neural crest specification and migration. We also identify an unexpected phosphatase- and Erk-independent function, mediated through its SH2 domains, which is evolutionarily conserved and prevents p53-mediated apoptosis in the brain and neural crest. Our results indicate that previously enigmatic aspects of LEOPARD syndrome pathogenesis can be explained by the combined effects of loss of Shp2 catalytic function and retention of an SH2 domain-mediated role that is essential for neural crest cell survival.
Collapse
|
23
|
Zhan Y, Counelis GJ, O'Rourke DM. The protein tyrosine phosphatase SHP-2 is required for EGFRvIII oncogenic transformation in human glioblastoma cells. Exp Cell Res 2009; 315:2343-57. [PMID: 19427850 DOI: 10.1016/j.yexcr.2009.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 02/03/2023]
Abstract
Oncogenic EGFRvIII is a naturally occurring oncoprotein and is expressed in about 40-50% of human glioblastomas, particularly those that arise de novo. To understand the molecular mechanisms by which this oncoprotein alters transforming phenotypes, and since our previous work indicated that SHP-2 protein tyrosine phosphatase activity modulated EGFRvIII activation and downstream signaling, we examined whether SHP-2 plays a role in EGFRvIII-induced oncogenesis by using both PTEN-deficient U87MG.EGFRvIII and PTEN-intact LN229.EGFRvIII cells. Inhibition of SHP-2 expression by Shp-2 siRNA inhibited cell growth, transformation and altered morphology of these EGFRvIII transformed GBM cells. Ectopic expression of a PTPase-inactive form of SHP-2, SHP-2 C459S, but not its wild-type SHP-2 or either of two SH2 domain mutants, abrogated transformation of EGFRvIII-expressing glioblastomas in soft agar and in nude mice. SHP-2 C459S cells grew slower and exhibited a more flattened morphology with more organized actin stress fibers under both full growth and low serum conditions. Furthermore, shp-2+/- and -/- mouse embryonic fibroblasts (MEFs) could not be transformed by EGFRvIII while shp-2+/+ MEFs displayed a fully transformed phenotype upon introduction of EGFRvIII, again indicating a requirement for functional SHP-2 in EGFRvIII transformation. Moreover, the SHP-2 PTPase activity inhibitor NSC-87877 inhibited endogenous SHP-2 activity, Erk phosphorylation and transformation in both GBM cell lines. EGFRvIII expression recruited SHP-2 to the receptor complex to transduce signals and also increased SHP-2 phosphorylation at Tyr542. Inhibition of EGFRvIII-induced cell growth and transformation by SHP-2 C459S or shp-2 siRNA was mediated by its ability to block cell cycle progression at different phases in these GBM cells. These data indicate that differential activation of SHP-2 phosphorylation at Tyr542 in these two GBM cell lines likely results in increased different PTPase activity and distinct mechanisms of cell cycle progression and SHP-2, in particular its PTPase activity, plays a critical role in EGFRvIII-mediated transformation.
Collapse
Affiliation(s)
- Yi Zhan
- Department of Neurosurgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
24
|
Langdon YG, Goetz SC, Berg AE, Swanik JT, Conlon FL. SHP-2 is required for the maintenance of cardiac progenitors. Development 2007; 134:4119-30. [PMID: 17928416 DOI: 10.1242/dev.009290] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The isolation and culturing of cardiac progenitor cells has demonstrated that growth factor signaling is required to maintain cardiac cell survival and proliferation. In this study, we demonstrate in Xenopus that SHP-2 activity is required for the maintenance of cardiac precursors in vivo. In the absence of SHP-2 signaling, cardiac progenitor cells downregulate genes associated with early heart development and fail to initiate cardiac differentiation. We further show that this requirement for SHP-2 is restricted to cardiac precursor cells undergoing active proliferation. By demonstrating that SHP-2 is phosphorylated on Y542/Y580 and that it binds to FRS-2, we place SHP-2 in the FGF pathway during early embryonic heart development. Furthermore, we demonstrate that inhibition of FGF signaling mimics the cellular and biochemical effects of SHP-2 inhibition and that these effects can be rescued by constitutively active/Noonan-syndrome-associated forms of SHP-2. Collectively, these results show that SHP-2 functions within the FGF/MAPK pathway to maintain survival of proliferating populations of cardiac progenitor cells.
Collapse
Affiliation(s)
- Yvette G Langdon
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
25
|
Kuo PL, Chen CY, Hsu YL. Isoobtusilactone A induces cell cycle arrest and apoptosis through reactive oxygen species/apoptosis signal-regulating kinase 1 signaling pathway in human breast cancer cells. Cancer Res 2007; 67:7406-20. [PMID: 17671211 DOI: 10.1158/0008-5472.can-07-1089] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study is the first to investigate the anticancer effect of isoobtusilactone A (IOA) in two human breast cancer cell lines, MCF-7 and MDA-MB-231. IOA exhibited effective cell growth inhibition by inducing cancer cells to undergo G(2)-M phase arrest and apoptosis. Further investigation revealed that IOA's inhibition of cell growth was also evident in a nude mice model. Cell cycle blockade was associated with increased levels of p21 and reduced amounts of cyclin B1, cyclin A, cdc2, and cdc25C. IOA also enhanced the levels of inactivated phosphorylated cdc2 and cdc25C. IOA triggered the mitochondrial apoptotic pathway, as indicated by a change in Bax/Bcl-2 ratios, resulting in mitochondrial membrane potential loss, cytochrome c release, and caspase-9 activation. We also found that the generation of reactive oxygen species (ROS) is a critical mediator in IOA-induced cell growth inhibition. Enhancement of ROS by IOA activated apoptosis signal-regulating kinase 1 (ASK1) resulted in the increased activation of c-Jun NH(2)-terminal kinase and p38. Antioxidants EUK8 and N-acetyl cystenine significantly decreased apoptosis by inhibiting the ASK1 dephosphorylation at Ser(967) and subsequently increased the interaction of ASK1 with thioredoxin or 14-3-3 proteins. Moreover, blocking ASK1 by small interfering RNA inhibition completely suppressed IOA-induced apoptosis. Taken together, these results imply a critical role for ROS and ASK1 in IOA's anticancer activity.
Collapse
Affiliation(s)
- Po-Lin Kuo
- Cell Biology Laboratory, Department of Biotechnology, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | | | | |
Collapse
|
26
|
Xue J, Zhou D, Yao H, Gavrialov O, McConnell MJ, Gelb BD, Haddad GG. Novel functional interaction between Na+/H+exchanger 1 and tyrosine phosphatase SHP-2. Am J Physiol Regul Integr Comp Physiol 2007; 292:R2406-16. [PMID: 17289818 DOI: 10.1152/ajpregu.00859.2006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Besides being a intracellular pH (pHi) regulator, Na+/H+exchanger (NHE)1 has recently been postulated as a membrane scaffold that assembles protein complexes and coordinates various signaling pathways. The aim of the present study was to uncover NHE1 interactive partners and study their functional implications. NHE1 interactive partners were screened in the mouse brain with a signal transduction AntibodyArray. Ten of 400 tested proteins appeared to be potentially associated with NHE1. These partners have been shown to be involved in either cell proliferative or apoptotic pathways. The interactions between NHE1 and Src homology 2 domain-containing protein tyrosine phosphatase (SHP-2), Bin1, and heat shock protein (HSP)70 were reciprocally confirmed by coimmunoprecipitation. Moreover, in vitro binding data have shown that NHE1 COOH terminus interacts directly with SHP-2. The functional significance of the association between NHE1 and SHP-2 was further investigated by measuring pHi, cell proliferation, and cell death with the fluorescent dye BCECF, [3H]thymidine incorporation, and medium lactate dehydrogenase activity, respectively. Our results revealed that cells with SHP-2 overexpression exhibited a higher steady-state pHiand a faster, NHE1-dependent pHirecovery rate from acid load in HEPES buffer. In addition, SHP-2 overexpression diminished the HOE-642-induced inhibition of cell proliferation and protected cells from hypoxic injury, especially in the presence of HOE-642. Together, our findings demonstrate that SHP-2 not only is physically associated with NHE1 but also modulates NHE1 functions such as pHiregulation, cell proliferation, and cell death under hypoxia.
Collapse
Affiliation(s)
- Jin Xue
- Department of Pediatrics, University of California San Diego, San Diego, California 92093-0735, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Amin ARMR, Thakur VS, Paul RK, Feng GS, Qu CK, Mukhtar H, Agarwal ML. SHP-2 tyrosine phosphatase inhibits p73-dependent apoptosis and expression of a subset of p53 target genes induced by EGCG. Proc Natl Acad Sci U S A 2007; 104:5419-24. [PMID: 17369354 PMCID: PMC1838507 DOI: 10.1073/pnas.0700642104] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Green tea polyphenol, epigallocatechin-3-gallate (EGCG) differentially regulates the cellular growth of cancer cells in a p53-dependent manner through apoptosis and/or cell cycle arrest. In an effort to further elucidate the mechanism of differential growth regulation by EGCG, we have investigated the role of the tyrosine phosphatase, SHP-2. Comparing the responses of mouse embryonic fibroblasts (MEFs), expressing either WT or functionally inactive/truncated SHP-2, we find that inactivation of SHP-2 remarkably sensitizes cells to EGCG-mediated killing. MEFs lacking functional SHP-2 undergo massive apoptosis upon treatment with EGCG. By comparing gene expression profiles, we have identified a set of transcriptional targets of p53 that are differentially modulated in cells undergoing apoptosis. Western blot and real-time PCR analyses of a select group of genes further confirm that the expression is SHP-2-dependent. Similar observations were made in MEFs lacking p53, confirming that the expression of these "p53 target genes" is p53-independent. In addition, EGCG treatment induced the expression of p73 mRNA and protein in both cell types, but not p63. Inactivation of p73 in cells expressing nonfunctional SHP-2 markedly inhibited apoptosis and p53 target gene expression. Although phosphorylation of JNK is differentially regulated by SHP2, it was found to be dispensable for EGCG-induced apoptosis and p53 target gene expression. Our results have identified SHP-2 as a negative regulator of EGCG-induced-apoptosis and have identified a subset of p53 target genes whose expression is paradoxically not mediated by p53 but by one of its family members, p73.
Collapse
Affiliation(s)
| | | | | | | | - Cheng-Kui Qu
- Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106
| | | | - Munna L. Agarwal
- Departments of *Genetics and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
28
|
Chan RJ, Li Y, Hass MN, Walter A, Voorhorst CS, Shelley WC, Yang Z, Orschell CM, Yoder MC. Shp-2 heterozygous hematopoietic stem cells have deficient repopulating ability due to diminished self-renewal. Exp Hematol 2006; 34:1230-9. [PMID: 16939816 DOI: 10.1016/j.exphem.2006.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 04/14/2006] [Accepted: 04/17/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Improved understanding of hematopoietic stem cell (HSC) differentiation, proliferation, and self-renewal is sought to develop improved stem cell-based therapies as well as to define novel therapies for stem cell-based diseases such as leukemia. Shp-2 is a widely expressed nonreceptor protein tyrosine phosphatase that participates early in hematopoietic development. The following study was performed to examine the role of Shp-2 in HSC function. METHODS Bone marrow low-density mononuclear cells were isolated from WT and Shp-2(+/-) littermate controls and utilized in competitive repopulation studies, homing analysis, cell-cycle analysis, and serial transplantation studies. RESULTS Haploinsufficiency of Shp-2 causes a threefold reduction in HSC repopulating units following transplantation into lethally irradiated recipients. Homing of Shp-2(+/-) and WT cells to the bone marrow and spleen compartments was equal. Cell-cycle analysis studies revealed that the Shp-2(+/-) lin(-)Sca-1(+)c-kit(+) cells are less quiescent than WT cells, providing a potential etiology for the observed reduced engraftment of the Shp-2(+/-) cells. Consistently, in serial transplantation studies, we observed a significant reduction of Shp-2(+/-) self-renewal compared to that of WT cells. CONCLUSION These data demonstrate that Shp-2 is required for the physiologic homeostasis of the HSC compartment and potentially provide insight into how oncogenic Shp-2 may contribute to the pathogenesis of myeloproliferative disorders and leukemias.
Collapse
Affiliation(s)
- Rebecca J Chan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chen J, Yu WM, Daino H, Broxmeyer HE, Druker BJ, Qu CK. SHP-2 phosphatase is required for hematopoietic cell transformation by Bcr-Abl. Blood 2006; 109:778-85. [PMID: 17003374 PMCID: PMC1785089 DOI: 10.1182/blood-2006-04-019141] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
SHP-2 phosphatase forms a stable protein complex with and is heavily tyrosine-phosphorylated by the oncogenic tyrosine kinase Bcr-Abl. However, the role of SHP-2 in Bcr-Abl-mediated leukemogenesis is unclear. In the present report, we provide evidence that SHP-2 is required for hematopoietic cell transformation by Bcr-Abl. In vitro biological effects of Bcr-Abl transduction were diminished in SHP-2Delta/Delta hematopoietic cells, and the leukemic potential of Bcr-Abl-transduced SHP-2Delta/Delta cells in recipient animals was compromised. Further analyses showed that Bcr-Abl protein (p210) was degraded, and its oncogenic signaling was greatly decreased in SHP-2Delta/Delta cells. Treatment with proteasome inhibitors or reintroduction of SHP-2 restored p210 level in Bcr-Abl-transduced SHP-2Delta/Delta cells. Subsequent investigation revealed that SHP-2 interacted with heat shock protein 90, an important chaperone protein protecting p210 from proteasome-mediated degradation. The role of SHP-2 in the stability of p210 is independent of its catalytic activity. Blockade of SHP-2 expression in p210-expressing cells by antisense or small-interfering RNA approaches decreased p210 level, causing cell death. Inhibition of SHP-2 enzymatic activity by overexpression of catalytically inactive SHP-2 mutant did not destabilize p210 but enhanced serum starvation-induced apoptosis, suggesting that SHP-2 also plays an important role in downstream signaling of p210 kinase. These studies identified a novel function of SHP-2 and suggest that SHP-2 might be a useful target for controlling Bcr-Abl-positive leukemias.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Wen-Mei Yu
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Hanako Daino
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Hal E. Broxmeyer
- Walther Oncology Center and Department of Immunology and Microbiology, Indiana University School of Medicine, Indianapolis, IN
| | - Brian J. Druker
- Howard Hughes Medical Institute, Oregon Health & Science University Cancer Institute, Portland, OR
| | - Cheng-Kui Qu
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
- Correspondence: Cheng-Kui Qu,
Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106; e-mail:
| |
Collapse
|
30
|
Zou GM, Chan RJ, Shelley WC, Yoder MC. Reduction of Shp-2 Expression by Small Interfering RNA Reduces Murine Embryonic Stem Cell-Derived In Vitro Hematopoietic Differentiation. Stem Cells 2006; 24:587-94. [PMID: 16269528 DOI: 10.1634/stemcells.2005-0272] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Shp-2 is a member of a small family of cytoplasmic Src homology 2 (SH2) domain-containing protein tyrosine phosphatases. Although Shp-2 has been shown to be necessary for hematopoiesis using a mouse model expressing a mutant residual protein (Shp-2(delta/delta)), we used small interfering RNA (siRNA) to reduce Shp-2 expression and examined the consequences on embryonic stem cell (ESC)-derived hemangioblast, primitive, and definitive hematopoietic development. We found that at a concentration of 50 nM, Shp-2 siRNA effectively diminished Shp-2 expression in differentiating embryoid bodies. Hemangioblast, primitive, and definitive hematopoietic progenitor formation was decreased significantly after transfection with Shp-2 siRNA but not with scrambled siRNA. Because Shp-2 is involved in signals emanating from the basic fibroblast growth factor (bFGF) receptor, we asked whether Shp-2 functions in bFGF-mediated hemangioblast development. Reduction of Shp-2 expression using siRNA, but not scrambled siRNA, blocked the bFGF-induced increase in hemangioblast development. Using siRNA as an independent method of reducing Shp-2 function, in contrast to the mutant mouse model (Shp-2(delta/delta)) previously used, we demonstrate that Shp-2 is required in hemangioblast, primitive, and definitive progenitor hematopoietic development and that Shp-2 is integrally necessary for bFGF-mediated hemangioblast production.
Collapse
Affiliation(s)
- Gang-Ming Zou
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut St., R4-402E, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
31
|
Yuan L, Yu WM, Xu M, Qu CK. SHP-2 Phosphatase Regulates DNA Damage-induced Apoptosis and G2/M Arrest in Catalytically Dependent and Independent Manners, Respectively. J Biol Chem 2005; 280:42701-6. [PMID: 16260787 DOI: 10.1074/jbc.m506768200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SHP-2, a tyrosine phosphatase implicated in diverse signaling pathways induced by growth factors and cytokines, is also involved in DNA damage-triggered signaling and cellular responses. We previously demonstrated that SHP-2 played an important role in DNA damage-induced apoptosis and G2/M cell cycle checkpoint. In the present studies, we have provided evidence that SHP-2 functions in DNA damage apoptosis and G2/M arrest in catalytically dependent and independent manners, respectively. Mutant embryonic fibroblasts with the Exon 3 deletion mutation in SHP-2 showed decreased apoptosis and diminished G2/M arrest in response to cisplatin treatment. Wild type (WT), but not catalytically inactive mutant SHP-2 (SHP-2 C459S), rescued the apoptotic response of the mutant cells. Interestingly, both WT and SHP-2 C459S efficiently restored the G2/M arrest response. Furthermore, inhibition of the catalytic activity of endogenous SHP-2 in WT cells by overexpression of SHP-2 C459S greatly decreased cell death but not G2/M arrest induced by cisplatin. Biochemical analyses revealed that activation of c-Abl kinase was decreased in SHP-2 C459S-overexpressing cells. However, DNA damage-induced translocation of Cdc25C from the nucleus to the cytoplasm was fully restored in both WT and SHP-2 C459S "rescued" cells. Additionally, we demonstrated that the role of SHP-2 in DNA damage-induced cellular responses was independent of the tumor suppressor p53. Embryonic stem cells with the SHP-2 deletion mutation showed markedly decreased sensitivity to cisplatin-induced apoptosis, attributed to impaired induction of p73 but not p53. In agreement with these results, DNA damage-induced apoptosis and G2/M arrest were also decreased in SHP-2/p53 double mutant embryonic fibroblasts. Collectively, these studies have further defined the mechanisms by which SHP-2 phosphatase regulates DNA damage responses.
Collapse
Affiliation(s)
- Liangping Yuan
- Department of Medicine, Division of Hematology/Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
32
|
SHI QIANG, AIDA KEIKO, VANDEBERG JOHNL, WANG XINGLI. Passage-dependent changes in baboon endothelial cells--relevance to in vitro aging. DNA Cell Biol 2004; 23:502-9. [PMID: 15307953 PMCID: PMC1350950 DOI: 10.1089/1044549041562294] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In vitro cell culture system is a useful model for aging-related changes in a wide spectrum of biomedical research. In this study, we explored the passage and donor age-dependent changes in baboon macrovascular endothelial cells that are relevant to both in vitro cell culture aging models and experiments using cell culture techniques. We collected baboon femoral arterial samples from nine baboons ranging in age from 6 months to 30 years (equivalent to humans approximately 18 months to 90 years of age). We then cultured baboon femoral artery endothelial cells (BFAECs) in standard DMEM medium with 20% fetal calf serum with 1:3 split for subculture. Endothelial functions were documented by morphology, Dil-LDL uptake and expression of eNOS, MCP-1, vWF, VCAM-1, ICAM-1, and E-Selectin with or without cytokine stimulation. Most of the cells became nonmitotic after 30 population doublings, or 10 passages, when they became flattened, enlarged, and senescent. While it took approximately 3 days to reach confluence from three-dilution seeding at early passages (<6), confluence was not achieved even after 7 days of culture for cells after the 9th or 10th passage. There was a linear decline in eNOS expression with passage. However, this decline was significantly less in endothelial cells from a young baboon (6 months) than those from an old baboon (30 years). While basal expression of adhesion molecules was not changed with passaging, responses to cytokine stimulation appeared to be increased in later passaged cells. Our study has provided evidence for passage-related changes in key endothelial functions. The donor age-related differences in this in vitro aging process suggests that in vitro endothelial culture can serve as a biomarker for in vivo aging. Nonhuman primates can provide a model for investigating such aging-related biological characteristics.
Collapse
Affiliation(s)
- QIANG SHI
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas
| | - KEIKO AIDA
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas
| | - JOHN L. VANDEBERG
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas
- Southwest National Primate Research Center, San Antonio, Texas
| | - XING LI WANG
- Department of Genetics, Southwest Foundation for Biomedical Research, San Antonio, Texas
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Address reprint requests to: Xing Li Wang, M.D., Ph.D., MS NAB 2010, Baylor College of Medicine, One Baylor Plaza, Houston TX 77030, E-mail:
| |
Collapse
|
33
|
Li JG, Chen YK, Wang YM. Splicing of SV40T gene exons and construction of a retroviral vector pLLTSN. Shijie Huaren Xiaohua Zazhi 2004; 12:1104-1107. [DOI: 10.11569/wcjd.v12.i5.1104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct an immortalization vector for hepatocytes immortalization, and two exons of simian virus 40 large T antigen gene (SV40T) were spliced and a retroviral vector pLLTSN without intron was constructed.
METHODS: The two exons of SV40T were amplified respectively by high fidelity polymerase chain reaction (PCR) by using the plasmid pUC19-SV40T as the template. Then SV40T gene was spliced by overlapping extension (SOE), and cloned into theEcoRⅠand BamHⅠsites of the retroviral vector pLXSN. The positive recombinant clones were screened and identified by PCR by using colonies directly as templates, and by restriction endonuclease digestion analysis, and DNA sequence analysis.
RESULTS: The 2.1 kb SV40T gene was spliced. Among the ten colonies randomly screened, four were proved positive, and one of them was verified by plasmid DNA sequencing.
CONCLUSION: The retroviral vector pLLTSN containing SV40T without intron is successfully constructed.
Collapse
|