1
|
Atroosh F, Al-Habori M, Al-Eryani E, Saif-Ali R. Impact of khat (Catha edulis) and oral contraceptive use on telomerase levels and tumor suppressor genes p53 and p21 in normal subjects and breast cancer patients. Sci Rep 2024; 14:16365. [PMID: 39013992 PMCID: PMC11252306 DOI: 10.1038/s41598-024-67355-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024] Open
Abstract
This study aimed to evaluate the effects of oral contraceptive (OC) use, khat chewing, and their combined effect on telomerase level and tumor suppressor genes, p53 and p21 in breast cancer (BC) patients and normal volunteers. 140 Yemeni women aged 25-40 years old enrolled, 60 newly diagnosed pretreated BC patients, and 80 control subjects. Venous blood (5 ml) was collected and the results showed BC patients to have significantly raised levels of telomerase, p53, and p21 compared to the control group. The use of OCs significantly raised telomerase in control group with no effect in BC patients; whereas p53 and p21 were significantly increased in BC patients. On the other hand, khat chewing significantly increased p53 in controls and BC patients, whereas p21 was significantly raised in BC patients. The combined use of OCs and khat chewing significantly increased telomerase and p53 in control group, and significantly increased p53 and p21 in BC patients. Telomerase was shown to be a risk factor (OR 4.4) for BC, and the use of OCs was a high-risk factor for increasing telomerase (OR 27.8) in normal subjects. In contrast, khat chewing was shown to be protective (OR 0.142), and the combined use of OCs and khat chewing decreased the risk factor of telomerase from OR 27.8 to 2.1.
Collapse
Affiliation(s)
- Fairooz Atroosh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| | - Molham Al-Habori
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen.
| | - Ekram Al-Eryani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| | - Riyadh Saif-Ali
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| |
Collapse
|
2
|
Stojkovic L, Jovanovic I, Dincic E, Djordjevic A, Kuveljic J, Djuric T, Stankovic A, Vojinovic S, Zivkovic M. Targeted RNAseq Revealed the Gene Expression Signature of Ferroptosis-Related Processes Associated with Disease Severity in Patients with Multiple Sclerosis. Int J Mol Sci 2024; 25:3016. [PMID: 38474262 DOI: 10.3390/ijms25053016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Detrimental molecular processes in multiple sclerosis (MS) lead to the cellular accumulation of lipid peroxidation products and iron in the CNS, which represents the main driving force for ferroptosis. Ferroptosis is an iron-dependent form of regulated cell death, with proposed roles in neurodegeneration, oligodendrocyte loss and neuroinflammation in the pathogenesis of MS. Ferroptosis-related gene expression signature and molecular markers, which could reflect MS severity and progression, are currently understudied in humans. To tackle these challenges, we have applied a curated approach to create and experimentally analyze a comprehensive panel of ferroptosis-related genes covering a wide range of biological processes associated with ferroptosis. We performed the first ferroptosis-related targeted RNAseq on PBMCs from highly distinctive MS phenotype groups: mild relapsing-remitting (RR) (n = 24) and severe secondary progressive (SP) (n = 24), along with protein detection of GPX4 and products of lipid peroxidation (MDA and 4-HNE). Out of 138 genes, 26 were differentially expressed genes (DEGs), indicating changes in both pro- and anti-ferroptotic genes, representing a molecular signature associated with MS severity. The top three DEGs, as non-core ferroptosis genes, CDKN1A, MAP1B and EGLN2, were replicated by qPCR to validate findings in independent patient groups (16 RR and 16 SP MS). Co-expression and interactions of DEGs were presented as additional valuable assets for deeper understanding of molecular mechanisms and key targets related to MS severity. Our study integrates a wide genetic signature and biochemical markers related to ferroptosis in easily obtainable PBMCs of MS patients with clinical data and disease severity, thus providing novel molecular markers which can complement disease-related changes in the brain and undergo further research as potential therapeutic targets.
Collapse
Affiliation(s)
- Ljiljana Stojkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Ivan Jovanovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Evica Dincic
- Clinic for Neurology, Military Medical Academy, 11000 Belgrade, Serbia
- Medical Faculty, University of Defense in Belgrade, 11042 Belgrade, Serbia
| | - Ana Djordjevic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Jovana Kuveljic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Tamara Djuric
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| | - Slobodan Vojinovic
- Department of Neurology, Medical Faculty, University of Nis, 18000 Nis, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, P.O. Box 522, 11000 Belgrade, Serbia
| |
Collapse
|
3
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
4
|
Locquet MA, Brahmi M, Blay JY, Dutour A. Radiotherapy in bone sarcoma: the quest for better treatment option. BMC Cancer 2023; 23:742. [PMID: 37563551 PMCID: PMC10416357 DOI: 10.1186/s12885-023-11232-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
Bone sarcomas are rare tumors representing 0.2% of all cancers. While osteosarcoma and Ewing sarcoma mainly affect children and young adults, chondrosarcoma and chordoma have a preferential incidence in people over the age of 40. Despite this range in populations affected, all bone sarcoma patients require complex transdisciplinary management and share some similarities. The cornerstone of all bone sarcoma treatment is monobloc resection of the tumor with adequate margins in healthy surrounding tissues. Adjuvant chemo- and/or radiotherapy are often included depending on the location of the tumor, quality of resection or presence of metastases. High dose radiotherapy is largely applied to allow better local control in case of incomplete primary tumor resection or for unresectable tumors. With the development of advanced techniques such as proton, carbon ion therapy, radiotherapy is gaining popularity for the treatment of bone sarcomas, enabling the delivery of higher doses of radiation, while sparing surrounding healthy tissues. Nevertheless, bone sarcomas are radioresistant tumors, and some mechanisms involved in this radioresistance have been reported. Hypoxia for instance, can potentially be targeted to improve tumor response to radiotherapy and decrease radiation-induced cellular toxicity. In this review, the benefits and drawbacks of radiotherapy in bone sarcoma will be addressed. Finally, new strategies combining a radiosensitizing agent and radiotherapy and their applicability in bone sarcoma will be presented.
Collapse
Affiliation(s)
- Marie-Anaïs Locquet
- Cell Death and Pediatric Cancer Team, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008, Lyon, France
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Leon Berard, Unicancer Lyon, 69008, Lyon, France
| | - Jean-Yves Blay
- Cell Death and Pediatric Cancer Team, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008, Lyon, France
- Department of Medical Oncology, Centre Leon Berard, Unicancer Lyon, 69008, Lyon, France
- Université Claude Bernard Lyon I, Lyon, France
| | - Aurélie Dutour
- Cell Death and Pediatric Cancer Team, Cancer Initiation and Tumor Cell Identity Department, INSERM1052, CNRS5286, Cancer Research Center of Lyon, F-69008, Lyon, France.
| |
Collapse
|
5
|
Propylene glycol, a component of electronic cigarette liquid, damages epithelial cells in human small airways. Respir Res 2022; 23:216. [PMID: 35999544 PMCID: PMC9400210 DOI: 10.1186/s12931-022-02142-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/15/2022] [Indexed: 11/10/2022] Open
Abstract
Background Electronic cigarettes (e-cigarettes) are used worldwide as a substitute for conventional cigarettes. Although they are primarily intended to support smoking cessation, e-cigarettes have been identified as a gateway to smoking habits for young people. Multiple recent reports have described the health effects of inhaling e-cigarettes. E-cigarette liquid (e-liquid) is mainly composed of propylene glycol (PG) and glycerol (Gly), and the aerosol generated by these devices primarily contains these two components. Thus, this study aimed to evaluate the effects of PG and Gly on human small airway epithelial cells (SAECs). Methods SAECs were exposed to PG or Gly, and cell proliferation, cell viability, lactate dehydrogenase (LDH) release, DNA damage, cell cycle, and apoptosis were evaluated. Additionally, SAECs derived from chronic obstructive pulmonary disease (COPD) patients (COPD-SAECs) were investigated. Results Exposure of SAECs to PG significantly inhibited proliferation (1%, PG, p = 0.021; 2–4% PG, p < 0.0001) and decreased cell viability (1–4% PG, p < 0.0001) in a concentration-dependent manner. Gly elicited similar effects but to a reduced degree as compared to the same concentration of PG. PG also increased LDH release in a concentration-dependent manner (3% PG, p = 0.0055; 4% PG, p < 0.0001), whereas Gly did not show a significant effect on LDH release. SAECs exposed to 4% PG contained more cells that were positive for phosphorylated histone H2AX (p < 0.0001), a marker of DNA damage, and an increased proportion of cells in the G1 phase (p < 0.0001) and increased p21 expression (p = 0.0005). Moreover, caspase 3/7-activated cells and cleaved poly (ADP-ribose) polymerase 1 expression were increased in SAECs exposed to 4% PG (p = 0.0054). Furthermore, comparing COPD-SAECs to SAECs without COPD in PG exposure, cell proliferation, cell viability, DNA damage and apoptosis were significantly greater in COPD-SAECs. Conclusion PG damaged SAECs more than Gly. In addition, COPD-SAECs were more susceptible to PG than SAECs without COPD. Usage of e-cigarettes may be harmful to the respiratory system, especially in patients with COPD.
Collapse
|
6
|
Ou H, Hoffmann R, González‐López C, Doherty GJ, Korkola JE, Muñoz‐Espín D. Cellular senescence in cancer: from mechanisms to detection. Mol Oncol 2021; 15:2634-2671. [PMID: 32981205 PMCID: PMC8486596 DOI: 10.1002/1878-0261.12807] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/25/2020] [Accepted: 09/22/2020] [Indexed: 01/10/2023] Open
Abstract
Senescence refers to a cellular state featuring a stable cell-cycle arrest triggered in response to stress. This response also involves other distinct morphological and intracellular changes including alterations in gene expression and epigenetic modifications, elevated macromolecular damage, metabolism deregulation and a complex pro-inflammatory secretory phenotype. The initial demonstration of oncogene-induced senescence in vitro established senescence as an important tumour-suppressive mechanism, in addition to apoptosis. Senescence not only halts the proliferation of premalignant cells but also facilitates the clearance of affected cells through immunosurveillance. Failure to clear senescent cells owing to deficient immunosurveillance may, however, lead to a state of chronic inflammation that nurtures a pro-tumorigenic microenvironment favouring cancer initiation, migration and metastasis. In addition, senescence is a response to post-therapy genotoxic stress. Therefore, tracking the emergence of senescent cells becomes pivotal to detect potential pro-tumorigenic events. Current protocols for the in vivo detection of senescence require the analysis of fixed or deep-frozen tissues, despite a significant clinical need for real-time bioimaging methods. Accuracy and efficiency of senescence detection are further hampered by a lack of universal and more specific senescence biomarkers. Recently, in an attempt to overcome these hurdles, an assortment of detection tools has been developed. These strategies all have significant potential for clinical utilisation and include flow cytometry combined with histo- or cytochemical approaches, nanoparticle-based targeted delivery of imaging contrast agents, OFF-ON fluorescent senoprobes, positron emission tomography senoprobes and analysis of circulating SASP factors, extracellular vesicles and cell-free nucleic acids isolated from plasma. Here, we highlight the occurrence of senescence in neoplasia and advanced tumours, assess the impact of senescence on tumorigenesis and discuss how the ongoing development of senescence detection tools might improve early detection of multiple cancers and response to therapy in the near future.
Collapse
Affiliation(s)
- Hui‐Ling Ou
- CRUK Cambridge Centre Early Detection ProgrammeDepartment of OncologyHutchison/MRC Research CentreUniversity of CambridgeUK
| | - Reuben Hoffmann
- Department of Biomedical EngineeringKnight Cancer InstituteOHSU Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandORUSA
| | - Cristina González‐López
- CRUK Cambridge Centre Early Detection ProgrammeDepartment of OncologyHutchison/MRC Research CentreUniversity of CambridgeUK
| | - Gary J. Doherty
- Department of OncologyCambridge University Hospitals NHS Foundation TrustCambridge Biomedical CampusUK
| | - James E. Korkola
- Department of Biomedical EngineeringKnight Cancer InstituteOHSU Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandORUSA
| | - Daniel Muñoz‐Espín
- CRUK Cambridge Centre Early Detection ProgrammeDepartment of OncologyHutchison/MRC Research CentreUniversity of CambridgeUK
| |
Collapse
|
7
|
Porosnicu I, Butnaru CM, Tiseanu I, Stancu E, Munteanu CVA, Bita BI, Duliu OG, Sima F. Y 2O 3 Nanoparticles and X-ray Radiation-Induced Effects in Melanoma Cells. Molecules 2021; 26:molecules26113403. [PMID: 34199757 PMCID: PMC8200002 DOI: 10.3390/molecules26113403] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
The innovative strategy of using nanoparticles in radiotherapy has become an exciting topic due to the possibility of simultaneously improving local efficiency of radiation in tumors and real-time monitoring of the delivered doses. Yttrium oxide (Y2O3) nanoparticles (NPs) are used in material science to prepare phosphors for various applications including X-ray induced photodynamic therapy and in situ nano-dosimetry, but few available reports only addressed the effect induced in cells by combined exposure to different doses of superficial X-ray radiation and nanoparticles. Herein, we analyzed changes induced in melanoma cells by exposure to different doses of X-ray radiation and various concentrations of Y2O3 NPs. By evaluation of cell mitochondrial activity and production of intracellular reactive oxygen species (ROS), we estimated that 2, 4, and 6 Gy X-ray radiation doses are visibly altering the cells by inducing ROS production with increasing the dose while at 6 Gy the mitochondrial activity is also affected. Separately, high-concentrated solutions of 25, 50, and 100 µg/mL Y2O3 NPs were also found to affect the cells by inducing ROS production with the increase of concentration. Additionally, the colony-forming units assay evidenced a rather synergic effect of NPs and radiation. By adding the NPs to cells before irradiation, a decrease of the number of proliferating cell colonies was observed with increase of X-ray dose. DNA damage was evidenced by quantifying the γ-H2AX foci for cells treated with Y2O3 NPs and exposed to superficial X-ray radiation. Proteomic profile confirmed that a combined effect of 50 µg/mL Y2O3 NPs and 6 Gy X-ray dose induced mitochondria alterations and DNA changes in melanoma cells.
Collapse
Affiliation(s)
- Ioana Porosnicu
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
- Faculty of Physics, Doctoral School on Physics, University of Bucharest, 405 Atomistilor Street, 077125 Magurele-Ilfov, Romania;
| | - Cristian M. Butnaru
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
- Correspondence: (C.M.B.); (F.S.)
| | - Ion Tiseanu
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
| | - Elena Stancu
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
| | - Cristian V. A. Munteanu
- Institute of Biochemistry, Romanian Academy, 296 Splaiul Independentei, 060031 Bucharest, Romania;
| | - Bogdan I. Bita
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
| | - Octavian G. Duliu
- Faculty of Physics, Doctoral School on Physics, University of Bucharest, 405 Atomistilor Street, 077125 Magurele-Ilfov, Romania;
| | - Felix Sima
- National Institute of Laser Plasma and Radiation Physics, P.O. Box MG-36, 76900 Bucharest-Magurele, Romania; (I.P.); (I.T.); (E.S.); (B.I.B.)
- Correspondence: (C.M.B.); (F.S.)
| |
Collapse
|
8
|
de Pedro I, Galán-Vidal J, Freije A, de Diego E, Gandarillas A. p21CIP1 controls the squamous differentiation response to replication stress. Oncogene 2020; 40:152-162. [PMID: 33097856 DOI: 10.1038/s41388-020-01520-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 11/09/2022]
Abstract
The control of cell fate is critical to homeostasis and cancer. Cell cycle cdk inhibitor p21CIP1 has a central and paradoxical role in the regulatory crossroads leading to senescence, apoptosis, or differentiation. p21 is an essential target of tumor suppressor p53, but it also is regulated independently. In squamous self-renewal epithelia continuously exposed to mutagenesis, p21 controls cell fate by mechanisms still intriguing. We previously identified a novel epidermoid DNA damage-differentiation response. We here show that p21 intervenes in the mitosis block that is required for the squamous differentiation response to cell cycle deregulation and replication stress. The inactivation of endogenous p21 in human primary keratinocytes alleviated the differentiation response to oncogenic loss of p53 or overexpression of the DNA replication major regulator Cyclin E. The bypass of p21-induced mitotic block involving upregulation of Cyclin B allowed DNA damaged cells to escape differentiation and continue to proliferate. In addition, loss of p21 drove keratinocytes from differentiation to apoptosis upon moderate UV irradiation. The results show that p21 is required to drive keratinocytes towards differentiation in response to genomic stress and shed light into its dual and paradoxical role in carcinogenesis.
Collapse
Affiliation(s)
- Isabel de Pedro
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Jesús Galán-Vidal
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Ana Freije
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain
| | - Ernesto de Diego
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain.,Paediatric Surgery, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Alberto Gandarillas
- Cell Cycle, Stem Cell Fate and Cancer Laboratory, Institute for Research Marqués de Valdecilla (IDIVAL), 39011, Santander, Spain. .,INSERM, Languedoc-Roussillon, 34394, Montpellier, France.
| |
Collapse
|
9
|
Choi J, Kim G, Cho SB, Im HJ. Radiosensitizing high-Z metal nanoparticles for enhanced radiotherapy of glioblastoma multiforme. J Nanobiotechnology 2020; 18:122. [PMID: 32883290 PMCID: PMC7470617 DOI: 10.1186/s12951-020-00684-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential step during the treatment of glioblastoma multiforme (GBM), one of the most lethal malignancies. The survival in patients with GBM was improved by the current standard of care for GBM established in 2005 but has stagnated since then. Since GBM is a radioresistant malignancy and the most of GBM recurrences occur in the radiotherapy field, increasing the effectiveness of radiotherapy using high-Z metal nanoparticles (NPs) has recently attracted attention. This review summarizes the progress in radiotherapy approaches for the current treatment of GBM, the physical and biological mechanisms of radiosensitization through high-Z metal NPs, and the results of studies on radiosensitization in the in vitro and in vivo GBM models using high-Z metal NPs to date.
Collapse
Affiliation(s)
- Jinyeong Choi
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Gaeun Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Su Bin Cho
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyung-Jun Im
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Yin Z, Jin H, Huang S, Qu G, Meng Q. REG γ knockdown suppresses proliferation by inducing apoptosis and cell cycle arrest in osteosarcoma. PeerJ 2020; 8:e8954. [PMID: 32328351 PMCID: PMC7166046 DOI: 10.7717/peerj.8954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/21/2020] [Indexed: 11/20/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common malignant bone tumor with high mortality in children and adolescents. REG γ is overexpressed and plays oncogenic roles in various types of human cancers. However, the expression and potential roles of REG γ in osteosarcoma are elusive. This study aims at exploring possible biological functions of REG γ in the pathogenesis of osteosarcoma and its underlying mechanism. Methods Quantitativereverse transcription-polymerase chain reaction (qRT-PCR), western blotting andimmunohistochemistry (IHC)were performed to detect the expression levels of REG γ in OS tissues and cell lines. Then, the effects of REG γ expression on OS cell proliferation in vitro were analyzed by Cell Counting Kit-8 (CCK-8), ethylene deoxyuridine (EdU), colony formation, flow cytometry. The protein levels of apoptosis and cell-cycle related proteins were evaluated using western blotting. Results In present study, we found for the first time that REG γ is overexpressed in osteosarcoma tissues and cell lines and knockdown of REG γ significantly inhibits cell proliferation and induces apoptosis and cell cycle arrest in osteosarcoma cells. Furthermore, we observed that p21, caspase-3 and cleaved caspase-3 are increased while the expression of cycinD1 and bcl-2 are decreased after REG γ depletion in osteosarcoma cells. In conclusion, REG γ may be involved in the proliferation of osteosarcoma and serve as a novel therapeutic target in patients with osteosarcoma.
Collapse
Affiliation(s)
- Zhiqiang Yin
- Department of Orthopedics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Jin
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shibo Huang
- Department of General Surgery, Heilongjiang Agricultural Reclamation General Hospital, Harbin, China
| | - Guofan Qu
- The Affiliated Tumor Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilong, China
| | - Qinggang Meng
- Department of Medical Science Institute of Harbin, The Fourth Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilong, China
| |
Collapse
|
11
|
Pandey S, Osman TA, Sharma S, Vallenari EM, Shahdadfar A, Pun CB, Gautam DK, Uhlin-Hansen L, Rikardsen O, Johannessen AC, Costea DE, Sapkota D. Loss of S100A14 expression at the tumor-invading front correlates with poor differentiation and worse prognosis in oral squamous cell carcinoma. Head Neck 2020; 42:2088-2098. [PMID: 32202693 DOI: 10.1002/hed.26140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/23/2020] [Accepted: 03/05/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND We previously showed a tumor-suppressive function of S100A14 in oral squamous cell carcinoma (OSCC). This study aimed to examine the prognostic significance and differentiation-related function of S100A14 in OSCC. METHODS S100A14 expression was examined in 170 OSCCs from Norwegian and Nepalese populations using immunohistochemistry. Pro-differentiation function was investigated by overexpressing and silencing S100A14 expression in OSCC-derived cells. External transcriptomic datasets were used to validate association between S100A14 and differentiation markers in OSCC. RESULT Loss of S100A14 expression at the invading tumor fronts significantly correlated with poor differentiation and reduced 10-years survival of OSCC-patients. Multivariate Cox analysis identified S100A14 to be an independent prognostic factor. Modulation of S100A14 expression in OSCC-derived cells positively correlated with the expression of differentiation markers. Analysis of external datasets supported the pro-differentiation function of S100A14. CONCLUSION These results indicate that S100A14 is a pro-differentiation protein and its expression might be useful as a prognostic marker in OSCC.
Collapse
Affiliation(s)
- Sushma Pandey
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Tarig A Osman
- Department of Clinical Medicine, the Gade Laboratory for Pathology, University of Bergen, Haukeland University Hospital, Bergen, Norway
| | - Sunita Sharma
- Department of Clinical Dentistry, Centre for Clinical Dental Research, University of Bergen, Bergen, Norway
| | - Evan M Vallenari
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Aboulghassem Shahdadfar
- Centre for Eye Research, Department of Ophthalmology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Chin B Pun
- Department of Pathology, B.P. Koirala Memorial Cancer Hospital, Bharatpur, Nepal
| | - Dej K Gautam
- Department of Surgical Oncology, B.P. Koirala Memorial Cancer Hospital, Bharatpur, Nepal
| | - Lars Uhlin-Hansen
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway.,Department of Medical Biology-Tumor Biology Research Group, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Oddveig Rikardsen
- Department of Otorhinolaryngology, University Hospital of North Norway, Tromsø, Norway
| | - Anne C Johannessen
- Department of Clinical Medicine, the Gade Laboratory for Pathology, University of Bergen, Haukeland University Hospital, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Daniela E Costea
- Department of Clinical Medicine, the Gade Laboratory for Pathology, University of Bergen, Haukeland University Hospital, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway.,Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Turesson I, Simonsson M, Hermansson I, Book M, Sigurdadottir S, Thunberg U, Qvarnström F, Johansson KA, Fessé P, Nyman J. Epidermal Keratinocyte Depletion during Five Weeks of Radiotherapy is Associated with DNA Double-Strand Break Foci, Cell Growth Arrest and Apoptosis: Evidence of Increasing Radioresponsiveness and Lack of Repopulation; the Number of Melanocytes Remains Unchanged. Radiat Res 2020; 193:481-496. [PMID: 32196412 DOI: 10.1667/rr15417.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
During fractionated radiotherapy, epithelial cell populations are thought to decrease initially, followed by accelerated repopulation to compensate cell loss. However, previous findings in skin with daily 1.1 Gy dose fractions indicate continued and increasing cell depletion. Here we investigated epidermal keratinocyte response with daily 2 Gy fractions as well as accelerated and hypofractionation. Epidermal interfollicular melanocytes were also assessed. Skin-punch biopsies were collected from breast cancer patients before, during and after mastectomy radiotherapy to the thoracic wall with daily 2 Gy fractions for 5 weeks. In addition, 2.4 Gy radiotherapy four times per week and 4 Gy fractions twice per week for 5 weeks, and two times 2 Gy daily for 2.5 weeks, were used. Basal keratinocyte density of the interfollicular epidermis was determined and immunostainings of keratinocytes for DNA double-strand break (DSB) foci, growth arrest, apoptosis and mitosis were quantified. In addition, interfollicular melanocytes were counted. Initially minimal keratinocyte loss was observed followed by pronounced depletion during the second half of treatment and full recovery at 2 weeks post treatment. DSB foci per cell peaked towards the end of treatment. p21-stained cell counts increased during radiotherapy, especially the second half. Apoptotic frequency was low throughout radiotherapy but increased at treatment end. Mitotic cell count was significantly suppressed throughout radiotherapy and did not recover during weekend treatment gaps, but increased more than threefold compared to unexposed skin 2 weeks post-radiotherapy. The number of melanocytes remained constant over the study period. Germinal keratinocyte loss rate increased gradually during daily 2 Gy fractions for 5 weeks, and similarly for hypofractionation. DSB foci number after 2 Gy irradiation revealed an initial radioresistance followed by increasing radiosensitivity. Growth arrest mediated by p21 strongly suggests that cells within or recruited into the cell cycle during treatment are at high risk of loss and do not contribute significantly to repopulation. It is possible that quiescent (G0) cells at treatment completion accounted for the accelerated post-treatment repopulation. Recent knowledge of epidermal tissue regeneration and cell cycle progression during genotoxic and mitogen stress allows for a credible explanation of the current finding. Melanocytes were radioresistant regarding cell depletion.
Collapse
Affiliation(s)
- Ingela Turesson
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Martin Simonsson
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | | | - Majlis Book
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Sunna Sigurdadottir
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Ulf Thunberg
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Fredrik Qvarnström
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Karl-Axel Johansson
- Department of Radiophysics, University of Göteborg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Fessé
- Department of Centre for Research and Development, Uppsala University/Region Gävleborg, Gävle, Sweden
| | | |
Collapse
|
13
|
Cellular Stress Responses in Radiotherapy. Cells 2019; 8:cells8091105. [PMID: 31540530 PMCID: PMC6769573 DOI: 10.3390/cells8091105] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy is one of the major cancer treatment strategies. Exposure to penetrating radiation causes cellular stress, directly or indirectly, due to the generation of reactive oxygen species, DNA damage, and subcellular organelle damage and autophagy. These radiation-induced damage responses cooperatively contribute to cancer cell death, but paradoxically, radiotherapy also causes the activation of damage-repair and survival signaling to alleviate radiation-induced cytotoxic effects in a small percentage of cancer cells, and these activations are responsible for tumor radio-resistance. The present study describes the molecular mechanisms responsible for radiation-induced cellular stress response and radioresistance, and the therapeutic approaches used to overcome radioresistance.
Collapse
|
14
|
Qi L, He W. REGγ Is Associated with Lymph Node Metastasis and T-Stage in Papillary Thyroid Carcinoma. Med Sci Monit 2018; 24:1373-1378. [PMID: 29509725 PMCID: PMC5849352 DOI: 10.12659/msm.905664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND This paper aimed to evaluate the expression of REGg and characterize its clinical significance in papillary thyroid carcinoma (PTC). MATERIAL AND METHODS In total, 54 patients with PTC who underwent partial or total thyroidectomy and cervical node dissection for PTC from February 2009 to September 2011 were retrospectively reviewed. Thyroid specimens and metastatic lymph nodes from 54 patients and normal thyroid tissues obtained from 13 volunteers were collected and analyzed. Tumor size, T-stage, and lymph nodes metastasis were recorded based on surgical pathology. Immunohistochemical (IHC) technology was performed to analyze REGg protein expression level. Corrections between the expression of REGγ and the clinicopathological factors were analyzed. RESULTS All the normal thyroid tissues were REGg-negative. REGγ was positive in 75.9% (41/54) of PTC tissues, of which 29 cases (29/42, 69.0%) were in T1-T2 stage and 12 cases (12/12,100%) were in T3-T4 stage. Positive REGγ was found in 21 cases (21/24, 87.5%) in T1-T2 stage with lymph nodes metastasis, while 11 cases were in T3-T4 stage with metastases to lymph nodes (11/11, 100%). High level of REGγ expression was significantly correlated with T-stage (P<0.05) and lymph node metastases (P<0.05). In addition, there was no statistically significant difference between the expression of REGγ and age, sex, tumor size, or tumor multiplicity (P>0.05). Using binary logistic regression model, positive REGγ was identified as a significant independent predictor factor of lymph node metastasis in PTC. CONCLUSIONS High expression of REGg seemed positively correlated with T-stage and lymph node metastasis in PTC tissues.
Collapse
Affiliation(s)
- Long Qi
- Department of Nuclear Medicine, Huadong Hospital, Fudan University, Shanghai, China (mainland)
| | - Wei He
- Department of Nuclear Medicine, Huadong Hospital, Fudan University, Shanghai, China (mainland)
| |
Collapse
|
15
|
Li H, Zou T, Meng S, Peng YZ, Yang JF. p21 protects cardiomyocytes against ischemia-reperfusion injury by inhibiting oxidative stress. Mol Med Rep 2018; 17:4665-4671. [PMID: 29328456 DOI: 10.3892/mmr.2018.8382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 01/27/2017] [Indexed: 11/06/2022] Open
Abstract
Ischemic heart disease is a major health threat, resulting in a large number of mortalities annually worldwide. Oxidative stress is one of the main causes of cell death during ischemia‑reperfusion (IR) injury. Cyclin dependent kinase inhibitor 1A (known as p21) is important in protecting tissues against IR injury, however the mechanism remains unknown. In the present study, oxygen‑glucose deprivation and subsequent reoxygenation (OGD/R) in H9c2 heart‑derived myocytes was used as a model to study myocardial IR injury in vitro. mRNA and protein expression levels were determined by reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. The levels of reactive oxygen species were measured using the fluorescence dye 2',7'‑dichlorodihydrofluorescein diacetate. The present data demonstrated that p21 expression was upregulated by tumor protein p53 (p53) in H9c2 cells exposed to OGD/R. p21 protected H9c2 cells against OGD/R‑induced oxidative stress. In addition, p21 mediated upregulation of NF‑E2‑related factor‑2 (Nrf2), a regulator of antioxidant responses, which in turn suppressed cell death in H9c2 cells subjected to OGD/R. Thus, activation of the p53/p21/Nrf2 signaling pathway may be an important adaptive response that limits oxidative injury during IR.
Collapse
Affiliation(s)
- Hong Li
- Department of Emergency Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Tong Zou
- Department of Cardiology, Beijing Hospital, Beijing 100730, P.R. China
| | - Shuai Meng
- Department of Emergency Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yun-Zhu Peng
- Department of Cardiology, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jie-Fu Yang
- Department of Cardiology, Beijing Hospital, Beijing 100730, P.R. China
| |
Collapse
|
16
|
He G, Di X, Sun X, Yan J, Zhang S. Analysis of radio-sensitization patents in China from 2006 to 2015. Expert Opin Ther Pat 2017. [PMID: 28621575 DOI: 10.1080/13543776.2017.1344222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Radiotherapy is by means of ionizing radiation to kill tumor cells, inhibit and control the growth, metastasis and diffusion of tumor cells. During the last few decades, application of radiotherapy combined with chemotherapy and surgery are clinical mainstream treatments. However, little is known what radio-sensitization agents have been patented in China and what the potential drug candidates for patents are in China. Areas covered: This reviews covers research and patent literature of the last 10 years dealing with the discovery and development of novel radio-sensitization patents in China. Expert opinion: The 94 radio-sensitization patents granted from 2006 to 2015 mainly focus on six types of products. They are: traditional Chinese medicines (TCM), synthetic compounds, combinations of synthetic compounds and TCM, biological products, medical apparatus and others. In the course of tumor treatment, radiotherapy occupies an irreplaceable position. Previously believed that due to the prevalence of hypoxic cells in solid tumors, most of the tumor exist a certain degree of radiation resistance. To find effective ways to improve the sensitivity of tumor cells to radiation therapy has become a focus in scientific research and clinical treatment. So radiation sensitivity has been proposed and widely studied.
Collapse
Affiliation(s)
- Guofeng He
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Xiaoke Di
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Xinchen Sun
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Jingjing Yan
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| | - Shu Zhang
- a Department of Radiation Oncology , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China.,b Clinical Research Center , the First Affiliated Hospital of Nanjing Medical University , Nanjing , P. R. China
| |
Collapse
|
17
|
Li H, Xu W, Ma Y, Zhou S. Separation and purification of the bovine milk fat globule membrane protein and its effect on improvement of C2C12mouse skeletal muscle cell proliferation. NEW J CHEM 2017. [DOI: 10.1039/c7nj00560a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A novel method to improve the proliferation activity of C2C12cells by the bovine milk fat globule membrane (MFGM) protein was established in this study.
Collapse
Affiliation(s)
- He Li
- School of Chemistry and Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- P. R. China
| | - Weili Xu
- School of Chemistry and Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- P. R. China
| | - Ying Ma
- School of Chemistry and Chemical Engineering
- Harbin Institute of Technology
- Harbin 150090
- P. R. China
| | - Shaobo Zhou
- School of Life Sciences
- Institute of Biomedical and Environmental Science and Technology
- University of BedfordshireLuton
- UK
| |
Collapse
|
18
|
Cellular Pathways in Response to Ionizing Radiation and Their Targetability for Tumor Radiosensitization. Int J Mol Sci 2016; 17:ijms17010102. [PMID: 26784176 PMCID: PMC4730344 DOI: 10.3390/ijms17010102] [Citation(s) in RCA: 273] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/22/2015] [Accepted: 12/25/2015] [Indexed: 12/20/2022] Open
Abstract
During the last few decades, improvements in the planning and application of radiotherapy in combination with surgery and chemotherapy resulted in increased survival rates of tumor patients. However, the success of radiotherapy is impaired by two reasons: firstly, the radioresistance of tumor cells and, secondly, the radiation-induced damage of normal tissue cells located in the field of ionizing radiation. These limitations demand the development of drugs for either radiosensitization of tumor cells or radioprotection of normal tissue cells. In order to identify potential targets, a detailed understanding of the cellular pathways involved in radiation response is an absolute requirement. This review describes the most important pathways of radioresponse and several key target proteins for radiosensitization.
Collapse
|
19
|
Zhao Y, Ma X, Wang J, Chen S, Yuan H, Xu A, Hang H, Wu L. The Roles of p21(Waf1/CIP1) and Hus1 in Generation and Transmission of Damage Signals Stimulated by Low-Dose Alpha-Particle Irradiation. Radiat Res 2015; 184:578-85. [PMID: 26600172 DOI: 10.1667/rr4165.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Previously reported studies have demonstrated the involvement of p21(Waf1/CIP1) in radiation-induced bystander effects (RIBE). Mouse embryonic fibroblasts (MEFs) lacking Hus1 fail to proliferate in vitro, but inactivation of p21 allows for the continued growth of Hus1-deficient cells, indicating the close connection between p21 and Hus1 cells. In this study, wild-type MEFs, Hus1(+/+)p21(-/-) MEFs and p21(-/-)Hus1(-/-) MEFs were used in a series of radiation-induced bystander effect experiments, the roles of p21 and Hus1 in the induction and transmission of radiation-induced damage signals were investigated. Our results showed that after 5 cGy α particle irradiation, wild-type MEFs induced significant increases in γ-H2AX foci and micronuclei formation in bystander cells, whereas the bystander effects were not detectable in p21(-/-)Hus1(+/+) MEFs and were restored again in p21(-/-)Hus1(-/-) MEFs. Media transfer experiments showed that p21(-/-)Hus1(+/+) MEFs were deficient in the production bystander signals, but could respond to bystander signals. We further investigated the mitogen-activated protein kinases (MAPKs) that might be involved in the bystander effects. It was found that although knocking out p21 did not affect the expression of connexin43 and its phosphorylation, it did result in inactivation of some MAPK signal pathway kinases, including JNK1/2, ERK1/2 and p38, as well as a decrease in reactive oxygen species (ROS) levels in irradiated cells. However, the activation of MAPK kinases and the ROS levels in irradiated cells were restored in the cell line by knocking out Hus1. These results suggest that p21(Waf1/CIP1) and Hus1 play crucial roles in the generation and transmission of bystander damage signals after low-dose α-particle irradiation.
Collapse
Affiliation(s)
- Ye Zhao
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China
| | - Xiaoyan Ma
- b National Laboratory of Biomacromolecules and Center for Computational and Systems Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Jun Wang
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China;,c Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, P.R. China; and
| | - Shaopeng Chen
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China;,c Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, P.R. China; and
| | - Hang Yuan
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China;,c Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, P.R. China; and
| | - An Xu
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China;,c Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, P.R. China; and
| | - Haiying Hang
- b National Laboratory of Biomacromolecules and Center for Computational and Systems Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Lijun Wu
- a Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P.R. China;,c Key Laboratory of Environmental Toxicology and Pollution Control Technology of Anhui Province, Hefei, Anhui 230031, P.R. China; and.,d School of Nuclear Science and Technology, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| |
Collapse
|
20
|
Zhang H, Davies KJA, Forman HJ. Oxidative stress response and Nrf2 signaling in aging. Free Radic Biol Med 2015; 88:314-336. [PMID: 26066302 PMCID: PMC4628850 DOI: 10.1016/j.freeradbiomed.2015.05.036] [Citation(s) in RCA: 596] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/29/2015] [Accepted: 05/31/2015] [Indexed: 12/20/2022]
Abstract
Increasing oxidative stress, a major characteristic of aging, has been implicated in a variety of age-related pathologies. In aging, oxidant production from several sources is increased, whereas antioxidant enzymes, the primary lines of defense, are decreased. Repair systems, including the proteasomal degradation of damaged proteins, also decline. Importantly, the adaptive response to oxidative stress declines with aging. Nrf2/EpRE signaling regulates the basal and inducible expression of many antioxidant enzymes and the proteasome. Nrf2/EpRE activity is regulated at several levels, including transcription, posttranslation, and interactions with other proteins. This review summarizes current studies on age-related impairment of Nrf2/EpRE function and discusses the changes in Nrf2 regulatory mechanisms with aging.
Collapse
Affiliation(s)
- Hongqiao Zhang
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology
| | - Kelvin J A Davies
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology; Division of Molecular & Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, CA 90089-0191, USA
| | - Henry Jay Forman
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology; School of Natural Science, University of California at Merced, Merced, CA 95344, USA.
| |
Collapse
|
21
|
Yang H, Yin P, Shi Z, Ma Y, Zhao C, Zheng J, Chen T. Sinomenine, a COX-2 inhibitor, induces cell cycle arrest and inhibits growth of human colon carcinoma cells in vitro and in vivo. Oncol Lett 2015; 11:411-418. [PMID: 26870226 DOI: 10.3892/ol.2015.3838] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 09/22/2015] [Indexed: 11/06/2022] Open
Abstract
Certain non-steroidal anti-inflammatory drugs may possess anti-tumorigenic effects in certain cancer cell types. Sinomenine (SIN) is an alkaloid from Sinomenium acutum, a Chinese medicinal plant that inhibits inflammatory reactions and that has been used in the treatment of neuralgia and rheumatic diseases. In this study, we investigated the anticancer effects of SIN against colorectal cancer in vitro and in vivo, as well as the underlying mechanisms. The effects of SIN on proliferation, cell cycle progression and cyclooxygenase (COX)-2 expression were examined in human colorectal cancer-derived SW1116 cells. The in vivo effects of SIN were examined in a model of SW1116 tumor xenograft growth in athymic nude mice. Changes in COX-2 expression induced by the biological effects of SIN were analyzed by western blot analysis. The effects of SIN treatment on G1 phase cell cycle regulators in xenografts were analyzed by immunohistochemistry. Our findings demonstrate that SIN inhibits the proliferation of SW1116 cells by promoting their accumulation in the G1 phase, with concomitant suppression of COX-2 expression. Time- and dose-dependent inhibition of tumor growth and reduced toxicity were observed in nude mice administered daily intraperitoneal injections of SIN at doses of 25, 50 and 100 mg/kg. SIN-treated tumors also exhibited reduced COX-2 expression, a marked increase in Cip1/p21 protein levels and a decrease in the levels of cyclin D1 and cyclin E. SIN may be an effective chemopreventive agent against colorectal cancer. The growth inhibitory properties of SIN against colorectal cancer may be mediated via a COX-2 inhibitory effect and cell cycle arrest in the G1 phase.
Collapse
Affiliation(s)
- Haibo Yang
- Department of Colorectal Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Peihao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhan Shi
- Department of Colorectal Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yanchun Ma
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Chenggen Zhao
- Department of Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Jampu Zheng
- Experimental Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Teng Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|
22
|
Li T, Cheng Y, Wang P, Wang W, Hu F, Mo X, Lv H, Xu T, Han W. CMTM4 is frequently downregulated and functions as a tumour suppressor in clear cell renal cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:122. [PMID: 26474560 PMCID: PMC4609138 DOI: 10.1186/s13046-015-0236-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/05/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND Chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing family (CMTM) is a gene family involved in multiple malignancies. CMTM4 is a member of this family and is located at chromosome 16q22.1, a locus that harbours a number of tumour suppressor genes. It has been defined as a regulator of cell cycle and division in HeLa cells; however, its roles in tumourigenesis remain poorly studied. METHODS An integrated bioinformatics analysis based on the array data from the GEO database was conducted to view the differential expression of CMTM4 across multiple cancers and their corresponding control tissues. Primary clear cell renal cell carcinoma (ccRCC) and the paired adjacent non-tumour tissues were then collected to examine the expression of CMTM4 by western blotting, immunohistochemistry, and quantitative RT-PCR. The ccRCC cell lines A498 and 786-O and the normal renal tubular epithelial cell line HK-2 were also tested for CMTM4 expression by western blotting. Cell Counting Kit-8 (CCK-8) and viable cell counting assays were used to delineate the growth curves of 786-O cells after CMTM4 overexpression or knockdown. Wound healing and transwell assays were performed to assess the cells' ability to migrate. The effects of CMTM4 on cellular apoptosis and cell cycle progression were analysed by flow cytometry, and cell cycle hallmarks were detected by western blotting and RT-PCR. The xenograft model in nude mice was used to elucidate the function of CMTM4 in tumourigenesis ex vivo. RESULTS By omic data analysis, we found a substantial downregulation of CMTM4 in ccRCC. Western blotting then confirmed that CMTM4 was dramatically reduced in 86.9 % (53/61) of ccRCC tissues compared with the paired adjacent non-tumour tissues, as well as in the 786-O and A498 ccRCC cell lines. Restoration of CMTM4 significantly suppressed 786-O cell growth by inducing G2/M cell cycle arrest and p21 upregulation, and cell migration was also inhibited. However, knockdown of CMTM4 led to a completely opposite effect on these cell behaviours. Overexpression of CMTM4 also markedly inhibited the tumour xenograft growth in nude mice. CONCLUSIONS CMTM4 is downregulated and exhibits tumour-suppressor activities in ccRCC, and could be exploited as a target for ccRCC treatment.
Collapse
Affiliation(s)
- Ting Li
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Yingying Cheng
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Pingzhang Wang
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Wenyan Wang
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Fengzhan Hu
- Department of Urology, Peking University People's Hospital, 11 Xi-Zhi-Men South Street, Beijing, 100044, China.
| | - Xiaoning Mo
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Hongxia Lv
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, 11 Xi-Zhi-Men South Street, Beijing, 100044, China.
| | - Wenling Han
- Peking University Center for Human Disease Genomics, Department of Immunology, Key Laboratory of Medical Immunology, Ministry of Health, School of Basic Medical Sciences, Peking University Center for Human Disease Genomics, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
23
|
REGγ is critical for skin carcinogenesis by modulating the Wnt/β-catenin pathway. Nat Commun 2015; 6:6875. [PMID: 25908095 DOI: 10.1038/ncomms7875] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/09/2015] [Indexed: 12/17/2022] Open
Abstract
Here we report that mice deficient for the proteasome activator, REGγ, exhibit a marked resistance to TPA (12-O-tetradecanoyl-phorbol-13-acetate)-induced keratinocyte proliferation, epidermal hyperplasia and onset of papillomas compared with wild-type counterparts. Interestingly, a massive increase of REGγ in skin tissues or cells resulting from TPA induces activation of p38 mitogen-activated protein kinase (MAPK/p38). Blocking p38 MAPK activation prevents REGγ elevation in HaCaT cells with TPA treatment. AP-1, the downstream effector of MAPK/p38, directly binds to the REGγ promoter and activates its transcription in response to TPA stimulation. Furthermore, we find that REGγ activates Wnt/β-catenin signalling by degrading GSK-3β in vitro and in cells, increasing levels of CyclinD1 and c-Myc, the downstream targets of β-catenin. Conversely, MAPK/p38 inactivation or REGγ deletion prevents the increase of cyclinD1 and c-Myc by TPA. This study demonstrates that REGγ acts in skin tumorigenesis mediating MAPK/p38 activation of the Wnt/β-catenin pathway.
Collapse
|
24
|
Kato H, Izumi K, Uenoyama A, Shiomi A, Kuo S, Feinberg SE. Hypoxia induces an undifferentiated phenotype of oral keratinocytes in vitro. Cells Tissues Organs 2015; 199:393-404. [PMID: 25720390 DOI: 10.1159/000371342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2014] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to determine the effects of hypoxia on the proliferating potential and phenotype of primary human oral keratinocytes cultured at ambient oxygen tension (20%) or at different levels of hypoxia (2 and 0.5% O2). The effects of oxygen tensions on cellular metabolic activity, cell proliferation, clonogenicity and proliferation heterogeneity were measured. Cell cycle profiles were analyzed by a fluorescent-activated cell sorter, and p21(WAF1/CIP1) expression in the G0/G1 phase was also concomitantly quantitated. The expression levels of cell cycle regulatory proteins were examined by immunoblotting, and the cellular senescence was assessed by senescence-associated β-galactosidase staining. Basal and suprabasal keratinocyte phenotypes were determined by the expression levels of 14-3-3σ, p75(NTR) and α6 integrin. Despite having a lower metabolism, the proliferation rate and clonogenic potential were remarkably enhanced in hypoxic cells. The significantly higher percentage of cells in the G0/G1 phase under hypoxia and the expression patterns of cell cycle regulatory proteins in hypoxic cells were indicative of a state of cell cycle arrest in hypoxia. Furthermore, a decrease in the expression of p21(WAF1/CIP1) and p16(INK4A) and fewer β-galactosidase-positive cells suggested a quiescent phenotype rather than a senescent one in hypoxic cells. Compared with normoxic cells, the differential expression patterns of keratinocyte phenotypic markers suggest that hypoxic cells that generate minimal reactive oxygen species, suppress the mammalian target of rapamycin activity and express hypoxia-inducible factor-1α favor a basal cell phenotype. Thus, regardless of the predisposition to the state of cell cycle arrest, hypoxic conditions can maintain oral keratinocytes in vitro in an undifferentiated and quiescent state.
Collapse
Affiliation(s)
- Hiroko Kato
- Department of Oral and Maxillofacial Surgery, University of Michigan, Ann Arbor, Mich., USA
| | | | | | | | | | | |
Collapse
|
25
|
Ichijo S, Furuya F, Shimura H, Hayashi Y, Takahashi K, Ohta K, Kobayashi T, Kitamura K. Activation of the RhoB signaling pathway by thyroid hormone receptor β in thyroid cancer cells. PLoS One 2014; 9:e116252. [PMID: 25548921 PMCID: PMC4280201 DOI: 10.1371/journal.pone.0116252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 12/05/2014] [Indexed: 11/19/2022] Open
Abstract
Thyroid hormone receptor (TR) mediates the crucial effects of the thyroid hormone (T3) on cellular growth, development, and differentiation. Decreased expression or inactivating somatic mutations of TRs have been found in human cancers of the liver, breast, lung, and thyroid. The mechanisms of TR-associated carcinogenesis are still not clear. To establish the function of TRβ in thyroid cancer cell proliferation, we constructed a recombinant adenovirus vector, AdTRβ, which expresses human TRβ1 cDNA. Thyroid cancer cell lines in which TRβ protein levels were significantly decreased as compared to intact thyroid tissues were infected with AdTRβ and the function of TRβ on cell proliferation and migration was analyzed. Ligand-bound TRβ induced HDAC1 and HDAC3 dissociation from, and histone acetylation associated with the RhoB promoter and enhanced the expression of RhoB mRNA and protein. In AdTRβ-infected cells, T3 and farnesyl transferase inhibitor (FTI)-treatment induced the distribution of RhoB on the cell membrane and enhanced the abundance of active GTP-bound RhoB. This RhoB protein led to p21-associated cell-cycle arrest in the G0/G1 phase, following inhibition of cell proliferation and invasion. Conversely, lowering cellular RhoB by small interfering RNA knockdown in AdTRβ-infected cells led to downregulation of p21 and inhibited cell-cycle arrest. The growth of BHP18-21v tumor xenografts invivo was significantly inhibited by AdTRβ injection with FTIs-treatment, as compared to control virus-injected tumors. This novel signaling pathway triggered by ligand-bound TRβ provides insight into possible mechanisms of proliferation and invasion of thyroid cancer and may provide new therapeutic targets for thyroid cancers.
Collapse
Affiliation(s)
- Sayaka Ichijo
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Fumihiko Furuya
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
- * E-mail:
| | - Hiroki Shimura
- Department of Laboratory Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yoshitaka Hayashi
- Department of Endocrinology and Metabolism, Division of Molecular and Cellular Adaptation, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kazuya Takahashi
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Kazuyasu Ohta
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Tetsuro Kobayashi
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Kenichiro Kitamura
- Third Department of Internal Medicine, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
26
|
Rossi S, Motta C, Studer V, Macchiarulo G, Volpe E, Barbieri F, Ruocco G, Buttari F, Finardi A, Mancino R, Weiss S, Battistini L, Martino G, Furlan R, Drulovic J, Centonze D. Interleukin-1β causes excitotoxic neurodegeneration and multiple sclerosis disease progression by activating the apoptotic protein p53. Mol Neurodegener 2014; 9:56. [PMID: 25495224 PMCID: PMC4292815 DOI: 10.1186/1750-1326-9-56] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022] Open
Abstract
Background Understanding how inflammation causes neuronal damage is of paramount importance in multiple sclerosis (MS) and in other neurodegenerative diseases. Here we addressed the role of the apoptotic cascade in the synaptic abnormalities and neuronal loss caused by the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor (TNF-α) in brain tissues, and disease progression caused by inflammation in relapsing-remitting MS (RRMS) patients. Results The effect of IL-1β, but not of TNF-α, on glutamate-mediated excitatory postsynaptic currents was blocked by pifithrin-α (PFT), inhibitor of p53. The protein kinase C (PKC)/transient receptor potential vanilloid 1 (TRPV1) pathway was involved in IL-1β-p53 interaction at glutamatergic synapses, as pharmacological modulation of this inflammation-relevant molecular pathway affected PFT effects on the synaptic action of IL-1β. IL-1β-induced neuronal swelling was also blocked by PFT, and IL-1β increased the expression of p21, a canonical downstream target of activated p53. Consistent with these in vitro results, the Pro/Pro genotype of p53, associated with low efficiency of transcription of p53-regulated genes, abrogated the association between IL-1β cerebrospinal fluid (CSF) levels and disability progression in RRMS patients. The interaction between p53 and CSF IL-1β was also evaluated at the optical coherence tomography (OCT), showing that IL-1β-driven neurodegenerative damage, causing alterations of macular volume and of retinal nerve fibre layer thickness, was modulated by the p53 genotype. Conclusions Inflammatory synaptopathy and neurodegeneration caused by IL-1β in RRMS patients involve the apoptotic cascade. Targeting IL-1β-p53 interaction might result in significant neuroprotection in MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Diego Centonze
- Clinica Neurologica, Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
27
|
Marques M, Laflamme L, Benassou I, Cissokho C, Guillemette B, Gaudreau L. Low levels of 3,3'-diindolylmethane activate estrogen receptor α and induce proliferation of breast cancer cells in the absence of estradiol. BMC Cancer 2014; 14:524. [PMID: 25048790 PMCID: PMC4223525 DOI: 10.1186/1471-2407-14-524] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/08/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 3,3'-diindolylmethane (DIM) is an acid-catalyzed dimer of idole-3-carbinol (I3C), a phytochemical found in cruciferous vegetables that include broccoli, Brussels sprouts and cabbage. DIM is an aryl hydrocarbon receptor (AhR) ligand and a potential anticancer agent, namely for the treatment of breast cancer. It is also advertised as a compound that regulates sex hormone homeostasis. METHODS Here we make use of RNA expression assays coupled to Chromatin Immunoprecipitation (ChIP) in breast cancer cell lines to study the effect of DIM on estrogen signaling. We further make use of growth assays, as well as fluorescence-activated cell sorting (FACS) assays, to monitor cell growth. RESULTS In this study, we report that 'physiologically obtainable' concentrations of DIM (10 μM) activate the estrogen receptor α (ERα) signaling pathway in the human breast cancer cell lines MCF7 and T47D, in a 17β-estradiol (E2)-independent manner. Accordingly, we observe induction of ERα target genes such as GREB1 and TFF1, and an increase in cellular proliferation after treatment with 10 μM DIM in the absence of E2. By using an ERα specific inhibitor (ICI 182 780), we confirm that the transcriptional and proliferative effects of DIM treatment are mediated by ERα. We further show that the protein kinase A signaling pathway participates in DIM-mediated activation of ERα. In contrast, higher concentrations of DIM (e.g. 50 μM) have an opposite and expected effect on cells, which is to inhibit proliferation. CONCLUSIONS We document an unexpected effect of DIM on cell proliferation, which is to stimulate growth by inducing the ERα signaling pathway. Importantly, this proliferative effect of DIM happens with potentially physiological concentrations that can be provided by the diet or by taking caplet supplements.
Collapse
Affiliation(s)
| | | | | | | | | | - Luc Gaudreau
- Département de Biologie, Université de Sherbrooke, J1K 2R1 Sherbrooke, QC, Canada.
| |
Collapse
|
28
|
|
29
|
Yang HL, Kumar KJS, Kuo YT, Chang HC, Liao JW, Hsu LS, Hseu YC. Antrodia camphorata induces G1 cell-cycle arrest in human premyelocytic leukemia (HL-60) cells and suppresses tumor growth in athymic nude mice. Food Funct 2014; 5:2278-88. [DOI: 10.1039/c4fo00423j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antrodia camphorata is a well-known medicinal mushroom in Taiwan.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition
- China Medical University
- Taichung 40402, Taiwan
| | | | - Ya-Ting Kuo
- Institute of Nutrition
- China Medical University
- Taichung 40402, Taiwan
| | - Hebron C. Chang
- Institute of Biotechnology and Bioinformatics
- Asia University
- Taichung 41354, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology
- National Chung Hsing University
- Taichung 402, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry and Biotechnology
- Chung Shan Medical University
- Taichung 40401, Taiwan
| | - You-Cheng Hseu
- Department of Health and Nutrition Biotechnology
- Asia University
- Taichung 41354, Taiwan
- Department of Cosmeceutics
- College of Pharmacy
| |
Collapse
|
30
|
Chang G, Zhang H, Wang J, Zhang Y, Xu H, Wang C, Zhang H, Ma L, Li Q, Pang T. CD44 targets Wnt/β-catenin pathway to mediate the proliferation of K562 cells. Cancer Cell Int 2013; 13:117. [PMID: 24257075 PMCID: PMC4176735 DOI: 10.1186/1475-2867-13-117] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 11/19/2013] [Indexed: 11/10/2022] Open
Abstract
Background Chronic myeloid leukemia is a clonal myeloproliferative disorder disease in which BCR/ABL plays an important role as an oncoprotein and molecular target. Despite the success of targeted therapy using tyrosine kinase inhibitors, CML remains largely incurable, most likely due to the treatment resistance after firstly chemical therapy. So know well the unique molecular pathway of CML is very important. Methods The expressions of CD44 in different leukemia patients and cell lines were detected by real-time PCR and western blotting. The effects of CD44 on proliferation of K562 cells were determined using the MTT and colony formation assays, and even in a nude mouse transplantation model. Then, the cell cycle changes were detected by flow cytometric analysis and the early apoptosis of cells was detected by the annexin V/propidium iodide double-staining assay. The expressions of the cycles and apoptosis-related proteins p21, Cyclin D1 and Bcl-2 were analyzed by western blot and real-time PCR assay. Finally, the decreased nuclear accumulation of β-catenin was detected by western blotting and immunefluorescence. Results Firstly, we showed that CD44 expression was increased in several kinds of leukemia patients and K562 cells. By contrast, the down-regulation of CD44 resulted in decreased proliferation with a G0/G1 arrest of cell cycle in K562 cells according to the MTT assay and the flow cytometric analysis. And no significant induction of both the early and late phases of apoptosis was shown by the annexin V-FITC and PI staining. During this process, p21 and cyclin D1 are the major causes for cell cycle arrest. In addition, we found CD44 down-regulation decreased the expression of β-catenin and increased the expression of phosphorylated β-catenin. The instability of Wnt/β-catenin pathway induced by increased expression of p-β-catenin resulted in a decreased nuclear accumulation in CD44 silenced K562 cells. In the nude mouse transplantation model, we also found the same results. Conclusions These results show that K562 cells depend to a greater extent on CD44 for proliferation, and CD44 down-regulation may induce a cell cycle arrest through Wnt/β-catenin pathway. CD44 blockade may be beneficial in therapy of CML.
Collapse
Affiliation(s)
- Guoqiang Chang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Protumorigenic effects of mir-145 loss in malignant pleural mesothelioma. Oncogene 2013; 33:5319-31. [PMID: 24240684 DOI: 10.1038/onc.2013.476] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 12/11/2022]
Abstract
We identified a discrete number of microRNAs differentially expressed in benign or malignant mesothelial tissues. We focused on mir-145 whose levels were significantly downregulated in malignant mesothelial tissues and malignant pleural mesothelioma (MPM) cell lines as compared to benign tissues (pleura, peritoneum or cysts). We show that promoter hyper-methylation caused very low levels in MPM cell lines and specimens. Treatment of MPM cell lines with mir-145 agonists negatively modulated some protumorigenic properties of MPM cells, such as clonogenicity, cell migration and resistance to pemetrexed treatment. The main effector mechanism of the clonogenic death induced by mir-145 was that of accelerated senescence. We found that mir-145 targeted OCT4 via specific binding to its 3'-UTR. Increased intracellular levels of mir-145 decreased the levels of OCT4 and its target gene ZEB1, thereby counteracting the increase of OCT4 induced by pemetrexed treatment which is known to favor the development of chemoresistant cells. In line with this, reintroduction of OCT4 into mimic-145 treated cells counteracted the effects on clonogenicity and replicative senescence. This further supports the relevance of the mir-145-OCT4 interaction for the survival of MPM cells. The potential use of mir-145 expression levels to classify benign vs malignant mesothelial tissues and the differences between pemetrexed-induced senescence and that induced by the re-expression of mir-145 are discussed.
Collapse
|
32
|
Miuma S, Saldivar JC, Karras JR, Waters CE, Paisie CA, Wang Y, Jin V, Sun J, Druck T, Zhang J, Huebner K. Fhit deficiency-induced global genome instability promotes mutation and clonal expansion. PLoS One 2013; 8:e80730. [PMID: 24244712 PMCID: PMC3828255 DOI: 10.1371/journal.pone.0080730] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/07/2013] [Indexed: 01/25/2023] Open
Abstract
Loss of Fhit expression, encoded at chromosome fragile site FRA3B, leads to increased replication stress, genome instability and accumulation of genetic alterations. We have proposed that Fhit is a genome ‘caretaker’ whose loss initiates genome instability in preneoplastic lesions. We have characterized allele copy number alterations and expression changes observed in Fhit-deficient cells in conjunction with alterations in cellular proliferation and exome mutations, using cells from mouse embryo fibroblasts (MEFs), mouse kidney, early and late after establishment in culture, and in response to carcinogen treatment. Fhit-/- MEFs escape senescence to become immortal more rapidly than Fhit+/+ MEFs; -/- MEFs and kidney cultures show allele losses and gains, while +/+ derived cells show few genomic alterations. Striking alterations in expression of p53, p21, Mcl1 and active caspase 3 occurred in mouse kidney -/- cells during progressive tissue culture passage. To define genomic changes associated with preneoplastic changes in vivo, exome DNAs were sequenced for +/+ and -/- liver tissue after treatment of mice with the carcinogen, 7,12-dimethylbenz[a]anthracene, and for +/+ and -/- kidney cells treated in vitro with this carcinogen. The -/- exome DNAs, in comparison with +/+ DNA, showed small insertions, deletions and point mutations in more genes, some likely related to preneoplastic changes. Thus, Fhit loss provides a ‘mutator’ phenotype, a cellular environment in which mild genome instability permits clonal expansion, through proliferative advantage and escape from apoptosis, in response to pressures to survive.
Collapse
Affiliation(s)
- Satoshi Miuma
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Joshua C. Saldivar
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jenna R. Karras
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Catherine E. Waters
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Carolyn A. Paisie
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Yao Wang
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Victor Jin
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jin Sun
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Teresa Druck
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Jie Zhang
- Department of Biomedical Informatics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
33
|
Saiz-Ladera C, Lara MF, Garín M, Ruiz S, Santos M, Lorz C, García-Escudero R, Martínez-Fernández M, Bravo A, Fernández-Capetillo O, Segrelles C, Paramio JM. p21 suppresses inflammation and tumorigenesis on pRB-deficient stratified epithelia. Oncogene 2013; 33:4599-4612. [PMID: 24121270 DOI: 10.1038/onc.2013.417] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/24/2013] [Accepted: 08/20/2013] [Indexed: 12/11/2022]
Abstract
The retinoblastoma gene product (pRb) controls proliferation and differentiation processes in stratified epithelia. Importantly, and in contrast to other tissues, Rb deficiency does not lead to spontaneous skin tumor formation. As the cyclin-dependent kinase inhibitor p21 regulates proliferation and differentiation in the absence of pRb, we analyzed the consequences of deleting p21 in pRb-ablated stratified epithelia (hereafter pRb(ΔEpi);p21-/-). These mice display an enhancement of the phenotypic abnormalities observed in pRb(ΔEpi) animals, indicating that p21 partially compensates pRb absence. Remarkably, pRb(ΔEpi);p21-/- mice show an acute skin inflammatory phenotype and develop spontaneous epithelial tumors, particularly affecting tongue and oral tissues. Biochemical analyses and transcriptome studies reveal changes affecting multiple pathways, including DNA damage and p53-dependent signaling responses. Comparative metagenomic analyses, together with the histopathological profiles, indicate that these mice constitute a faithful model for human head and neck squamous cell carcinomas. Collectively, our findings demonstrate that p21, in conjunction with pRb, has a central role in regulating multiple epithelial processes and orchestrating specific tumor suppressor functions.
Collapse
Affiliation(s)
- Cristina Saiz-Ladera
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - María Fernanda Lara
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Marina Garín
- Division of Hematopoietic Innovative Therapies (HIT). Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER). Madrid, Spain
| | - Sergio Ruiz
- Genomic Instability Group, Spanish National Cancer Research Center, Madrid, Spain
| | - Mirentxu Santos
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Corina Lorz
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Ramón García-Escudero
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Mónica Martínez-Fernández
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Ana Bravo
- Department of Veterinary Clinical Sciences Veterinary Faculty, University of Santiago de Compostela, E-27002 Lugo, Spain
| | | | - Carmen Segrelles
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| | - Jesús M Paramio
- Molecular Oncology Unit. Division of Biomedicine, CIEMAT (ed70A). Ave. Complutense 40, E-28040 Madrid, Spain
| |
Collapse
|
34
|
Gao J, Chen H, Yu Y, Song J, Song H, Su X, Li W, Tong X, Qian W, Wang H, Dai J, Guo Y. Inhibition of hepatocellular carcinoma growth using immunoliposomes for co-delivery of adriamycin and ribonucleotide reductase M2 siRNA. Biomaterials 2013; 34:10084-98. [PMID: 24060417 DOI: 10.1016/j.biomaterials.2013.08.088] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/30/2013] [Indexed: 12/18/2022]
Abstract
The chemotherapy combined with gene therapy has received great attention. We developed targeted LPD (liposome-polycation-DNA complex) conjugated with anti-EGFR (epidermal growth factor receptor) Fab' co-delivering adriamycin (ADR) and ribonucleotide reductase M2 (RRM2) siRNA (ADR-RRM2-TLPD), to achieve combined therapeutic effects in human hepatocellular carcinoma (HCC) overexpressing EGFR. The antitumor activity and mechanisms of ADR-RRM2-TLPD were investigated. The results showed that RRM2 expression was higher in HCC than in non-HCC tissue, and RRM2 siRNA inhibited HCC cell proliferation, suggesting that RRM2 is a candidate target for HCC therapy. ADR-RRM2-TLPD delivered ADR and RRM2 siRNA to EGFR overexpressing HCC cells specifically and efficiently both in vitro and in vivo, resulting in enhanced therapeutic effects (cytotoxicity, apoptosis and senescence-inducing activity) compared with single-drug loaded or non-targeted controls, including ADR-NC-TLPD (targeted LPD co-delivering ADR and negative control siRNA), RRM2-TLPD (targeted LPD delivering RRM2 siRNA) and ADR-RRM2-NTLPD (non-targeted LPD co-delivering ADR and RRM2 siRNA). Mechanism studies showed that p21 is involved in the combined therapeutic effect of ADR-RRM2-TLPD. The average weight of the orthotopic HCC in mice treated with ADR-RRM2-TLPD was significantly lighter than that of mice treated with other controls. Thus, ADR-RRM2-TLPD represents a potential strategy for combined therapy of HCC overexpressing EGFR.
Collapse
Affiliation(s)
- Jie Gao
- International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, China; National Engineering Research Center for Antibody Medicine & Shanghai Key Laboratory of Cell Engineering and Antibody, 399 Libing Road, Shanghai 201203, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Di Luccia B, Manzo N, Baccigalupi L, Calabrò V, Crescenzi E, Ricca E, Pollice A. Lactobacillus gasseri SF1183 affects intestinal epithelial cell survival and growth. PLoS One 2013; 8:e69102. [PMID: 23894414 PMCID: PMC3720908 DOI: 10.1371/journal.pone.0069102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/06/2013] [Indexed: 01/01/2023] Open
Abstract
It is now commonly accepted that the intestinal microbiota plays a crucial role in the gut physiology and homeostasis, and that both qualitative and quantitative alterations in the compositions of the gut flora exert profound effects on the host’s intestinal cells. In spite of this, the details of the interaction between commensal bacteria and intestinal cells are still largely unknown and only in few cases the molecular mechanisms have been elucidated. Here we analyze the effects of molecules produced and secreted by Lactobacillus gasseri SF1183 on human intestinal HCT116 cells. L. gasseri is a well known species of lactic acid bacteria, commonly associated to the human intestine and SF1183 is a human strain previously isolated from an ileal biopsy of an healthy volunteer. SF1183 produces and secretes, in a growth phase-dependent way, molecule(s) able to drastically interfere with HCT116 cell proliferation. Although several attempts to purify and identify the bioactive molecule(s) have been so far unsuccessful, a partial characterization has indicated that it is smaller than 3 kDa, thermostable and of proteinaceous nature. L. gasseri molecule(s) stimulate a G1-phase arrest of the cell cycle by up-regulation of p21WAF1 rendering cells protected from intrinsic and extrinsic apoptosis. A L. gasseri-mediated reduction of apoptosis and of cell proliferation could be relevant in protecting epithelial barrier integrity and helping in reconstituting tissutal homeostasis.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | - Nicola Manzo
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | - Loredana Baccigalupi
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | - Viola Calabrò
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | - Elvira Crescenzi
- Istituto di Endocrinologia ed Oncologia Sperimentale-CNR-via S. Pansini, Naples, Italy
| | - Ezio Ricca
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | - Alessandra Pollice
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
- * E-mail:
| |
Collapse
|
36
|
Di Luccia B, Manzo N, Baccigalupi L, Calabrò V, Crescenzi E, Ricca E, Pollice A. Lactobacillus gasseri SF1183 affects intestinal epithelial cell survival and growth. PLoS One 2013. [PMID: 23894414 DOI: 10.1016/j.jff.2017.12.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
It is now commonly accepted that the intestinal microbiota plays a crucial role in the gut physiology and homeostasis, and that both qualitative and quantitative alterations in the compositions of the gut flora exert profound effects on the host's intestinal cells. In spite of this, the details of the interaction between commensal bacteria and intestinal cells are still largely unknown and only in few cases the molecular mechanisms have been elucidated. Here we analyze the effects of molecules produced and secreted by Lactobacillus gasseri SF1183 on human intestinal HCT116 cells. L. gasseri is a well known species of lactic acid bacteria, commonly associated to the human intestine and SF1183 is a human strain previously isolated from an ileal biopsy of an healthy volunteer. SF1183 produces and secretes, in a growth phase-dependent way, molecule(s) able to drastically interfere with HCT116 cell proliferation. Although several attempts to purify and identify the bioactive molecule(s) have been so far unsuccessful, a partial characterization has indicated that it is smaller than 3 kDa, thermostable and of proteinaceous nature. L. gasseri molecule(s) stimulate a G1-phase arrest of the cell cycle by up-regulation of p21WAF1 rendering cells protected from intrinsic and extrinsic apoptosis. A L. gasseri-mediated reduction of apoptosis and of cell proliferation could be relevant in protecting epithelial barrier integrity and helping in reconstituting tissutal homeostasis.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Department of Biology, University of Naples Federico II-MSA-Via Cinthia, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
Drosten M, Lechuga CG, Barbacid M. Ras signaling is essential for skin development. Oncogene 2013; 33:2857-65. [PMID: 23831572 DOI: 10.1038/onc.2013.254] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/03/2013] [Accepted: 06/03/2013] [Indexed: 12/30/2022]
Abstract
Proliferation in the epidermis is a tightly controlled process. During skin development, epidermis formation and hair follicle morphogenesis crucially depend on the regulated balance between proliferation and differentiation. Here we deleted all three Ras loci (H-Ras, N-Ras and K-Ras) from keratinocytes in vitro as well as specifically from the epidermis in mice using a K5Cre strain. Upon Ras elimination, keratinocytes ceased proliferation and entered into senescence without any signs of apoptosis induction. Constitutive activation of the mitogen-activated protein kinase pathway was able to partially rescue the proliferative defects. In mice, Ras signaling was essential for proper development of the epidermis and hair follicles. Deletion of the three Ras loci during epidermis formation in mouse embryos caused a dramatic decrease in proliferation, resulting in a substantially thinner epidermis and delayed appearance of differentiation markers. We could not detect apoptotic or senescent cells in these embryos suggesting that loss of Ras protein expression only leads to severe hypoproliferation. These observations provide genetic evidence for an essential role of Ras proteins in the control of keratinocyte and epidermal proliferation.
Collapse
Affiliation(s)
- M Drosten
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Spanish National Cancer Research Center, Madrid, Spain
| | - C G Lechuga
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Spanish National Cancer Research Center, Madrid, Spain
| | - M Barbacid
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Spanish National Cancer Research Center, Madrid, Spain
| |
Collapse
|
38
|
Liu J, Wang Y, Li L, Zhou L, Wei H, Zhou Q, Liu J, Wang W, Ji L, Shan P, Wang Y, Yang Y, Jung SY, Zhang P, Wang C, Long W, Zhang B, Li X. Site-specific acetylation of the proteasome activator REGγ directs its heptameric structure and functions. J Biol Chem 2013; 288:16567-16578. [PMID: 23612972 PMCID: PMC3675592 DOI: 10.1074/jbc.m112.437129] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 04/22/2013] [Indexed: 12/22/2022] Open
Abstract
The proteasome activator REGγ has been reported to promote degradation of steroid receptor coactivator-3 and cyclin-dependent kinase inhibitors p21, p16, and p19 in a ubiquitin- and ATP-independent manner. A recent comparative analysis of REGγ expression in mouse and human tissues reveals a unique pattern of REGγ in specific cell types, suggesting undisclosed functions and biological importance of this molecule. Despite the emerging progress made in REGγ-related studies, how REGγ function is regulated remains to be explored. In this study, we report for the first time that REGγ can be acetylated mostly on its lysine 195 (Lys-195) residue by CREB binding protein (CBP), which can be reversed by sirtuin 1 (SIRT1) in mammalian cells. Site-directed mutagenesis abrogated acetylation at Lys-195 and significantly attenuated the capability of REGγ to degrade its target substrates, p21 and hepatitis C virus core protein. Mechanistically, acetylation at Lys-195 is important for the interactions between REGγ monomers and ultimately influences REGγ heptamerization. Biological analysis of cells containing REGγ-WT or REGγ-K195R mutant indicates an impact of acetylation on REGγ-mediated regulation of cell proliferation and cell cycle progression. These findings reveal a previously unknown mechanism in the regulation of REGγ assembly and activity, suggesting a potential venue for the intervention of the ubiquitin-independent REGγ proteasome activity.
Collapse
Affiliation(s)
- Jiang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Ying Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Lei Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Haibin Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qingxia Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Weicang Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lei Ji
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peipei Shan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yuanyuan Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Pei Zhang
- Department of Pathology, the Second Chengdu Municipal Hospital, Chengdu, Sichuan 610017, China
| | - Chuangui Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Weiwen Long
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Bianhong Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Xiaotao Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030.
| |
Collapse
|
39
|
Lauxen IS, Oliveira MG, Rados PV, Lingen MW, Nör JE, Sant'ana Filho M. Immunoprofiling of oral squamous cell carcinomas reveals high p63 and survivin expression. Oral Dis 2013; 20:e76-80. [PMID: 23738964 DOI: 10.1111/odi.12136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 04/23/2013] [Accepted: 05/09/2013] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cancer is a multifactorial disease composed of cells that show somatic mutations and epigenetic changes. The aim of this study was to investigate the expression of proteins involved in the development and maintenance of epithelia, cell cycle regulation, and apoptosis in human oral squamous cell carcinoma (OSCC) tissue samples. METHODS A tissue microarray containing 65 primary human OSCC specimens was immunolabeled for bcl-2, survivin, epidermal growth factor receptor (EGFR), p21, p53, p63, and cleaved caspase-3. RESULTS Samples were scored for percentage of positively stained tumor cells and staining intensity. A total immunostaining score was also calculated, using the product of percentage and intensity scores. All specimens showed high scores, > 75%, for p63 and survivin, and 75.4% of the specimens also presented high EGFR expression. All cases showed p53-positive cells. p21 showed a diffuse staining pattern. The percentage of cells positive for cleaved caspase-3 and bcl-2 was low. CONCLUSIONS The high frequency of tumor cells expressing p63 and survivin highlights the role of these proteins in the malignant transformation of oral epithelium. Collectively, our results suggest that p63 and survivin may constitute attractive targets for cancer therapy in patients with OSCC.
Collapse
Affiliation(s)
- I S Lauxen
- Oral Pathology, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | |
Collapse
|
40
|
Zakharchenko O, Cojoc M, Dubrovska A, Souchelnytskyi S. A role of TGFß1 dependent 14-3-3σ phosphorylation at Ser69 and Ser74 in the regulation of gene transcription, stemness and radioresistance. PLoS One 2013; 8:e65163. [PMID: 23741479 PMCID: PMC3669286 DOI: 10.1371/journal.pone.0065163] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/23/2013] [Indexed: 01/16/2023] Open
Abstract
Transforming growth factor-β (TGFβ) is a potent regulator of tumorigenesis, although mechanisms defining its tumor suppressing and tumor promoting activities are not understood. Here we describe phosphoproteome profiling of TGFβ signaling in mammary epithelial cells, and show that 60 identified TGFβ-regulated phosphoproteins form a network with scale-free characteristics. The network highlighted interactions, which may distribute signaling inputs to regulation of cell proliferation, metabolism, differentiation and cell organization. In this report, we identified two novel and TGFβ-dependent phosphorylation sites of 14-3-3σ, i.e. Ser69 and Ser74. We observed that 14-3-3σ phosphorylation is a feed-forward mechanism in TGFβ/Smad3-dependent transcription. TGFβ-dependent 14-3-3σ phosphorylation may provide a scaffold for the formation of the protein complexes which include Smad3 and p53 at the Smad3-specific CAGA element. Furthermore, breast tumor xenograft studies in mice and radiobiological assays showed that phosphorylation of 14-3-3σ at Ser69 and Ser74 is involved in regulation of cancer progenitor population and radioresistance in breast cancer MCF7 cells. Our data suggest that TGFβ-dependent phosphorylation of 14-3-3σ orchestrates a functional interaction of TGFβ/Smad3 with p53, plays a role in the maintenance of cancer stem cells and could provide a new potential target for intervention in breast cancer.
Collapse
Affiliation(s)
- Olena Zakharchenko
- Karolinska Biomics Center, Dept. of Oncology-Pathology, Karolinska Institute, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | | | | | |
Collapse
|
41
|
King KE, Reddi DM, Ponnamperuma RM, Gerdes M, Weinberg WC. Dysregulated ΔNp63α negatively regulates the maspin promoter in keratinocytes via blocking endogenous p73 binding. Mol Carcinog 2013; 53:698-710. [DOI: 10.1002/mc.22022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 02/04/2013] [Indexed: 12/30/2022]
Affiliation(s)
- Kathryn E. King
- Office of Biotechnology Products; CDER/FDA; Bethesda Maryland
| | | | | | | | | |
Collapse
|
42
|
Homocysteine inhibits hepatocyte proliferation via endoplasmic reticulum stress. PLoS One 2013; 8:e54265. [PMID: 23349842 PMCID: PMC3551933 DOI: 10.1371/journal.pone.0054265] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/10/2012] [Indexed: 01/30/2023] Open
Abstract
Homocysteine is an independent risk factor for coronary, cerebral, and peripheral vascular diseases. Recent studies have shown that levels of homocysteine are elevated in patients with impaired hepatic function, but the precise role of homocysteine in the development of hepatic dysfunction is unclear. In this study, we examined the effect of homocysteine on hepatocyte proliferation in vitro. Our results demonstrated that homocysteine inhibited hepatocyte proliferation by up-regulating protein levels of p53 as well as mRNA and protein levels of p21Cip1 in primary cultured hepatocytes. Homocysteine induced cell growth arrest in p53-positive hepatocarcinoma cell line HepG2, but not in p53-null hepatocarcinoma cell line Hep3B. A p53 inhibitor pifithrin-α inhibited the expression of p21Cip1 and attenuated homocysteine-induced cell growth arrest. Homocysteine induced TRB3 expression via endoplasmic reticulum stress pathway, resulting in Akt dephosphorylation. Knock-down of endogenous TRB3 significantly suppressed the inhibitory effect of homocysteine on cell proliferation and the phosphorylation of Akt. LiCl reversed homocysteine-mediated cell growth arrest by inhibiting TRB3-mediated Akt dephosphorylation. These results demonstrate that both TRB3 and p21Cip1 are critical molecules in the homocysteine signaling cascade and provide a mechanistic explanation for impairment of liver regeneration in hyperhomocysteinemia.
Collapse
|
43
|
Visioli F, Lauxen IS, Sant'ana Filho M, Rados PV. Expression of the cell cycle regulation proteins p53 and p21WAF1 in different types of non-dysplastic leukoplakias. J Appl Oral Sci 2013; 20:369-75. [PMID: 22858706 PMCID: PMC3881781 DOI: 10.1590/s1678-77572012000300013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 08/11/2011] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES The aim of this study was to analyze the immunolabeling of two cell cycle protein regulators, p53 and p21WAF1, in non-dysplastic leukoplakias with different epithelial alterations: acanthosis, hyperkeratosis and acanthosis combined with hyperkeratosis, and compare them with dysplastic leukoplakias. MATERIAL AND METHODS This was a prospective cohort study involving 36 patients with oral homogeneous leukoplakias. excisional biopsies were performed and the patients remain under clinical follow-up. The leukoplakias were divided into four groups: 6 acanthosis, 9 hyperkeratosis, 10 acanthosis combined with hyperkeratosis, and 11 epithelial dysplasias. Paraffin-embebeded sections were immunostained for p53 and p21WAF1. Five hundred cells from the basal layer and 500 from the parabasal layer were counted to determine the percentage of positive cells. A qualitative analysis was also carried out to determine the presence or absence of immunohistochemical staining in the intermediate and superficial layers. Groups were compared with ANOVA (p<0.05). Pearson's correlation coefficient was used to test for associations between the two markers, p53 and p21WAF1. RESULTS No leukoplakia recurred and no malignant transformation was observed whitin a follow-up period of 3-6 years. The mean percentage of p53 staining in the basal and parabasal layers was similar in all groups. p21WAF1 staining differed between layers was as follows: in the basal, only 3 to 4% of cells were stained, while in the parabasal, between 16 and 28% of the epithelial cells were stained in the four different studied groups with no statistically significant difference (p>0.05). CONCLUSIONS Our findings failed to differentiate the non-dysplastic lesions by means of p53 and p21WAF1 immunostaining, notwithstanding similar profiles between non-dysplastic and dysplastic leukoplakias were observed.
Collapse
Affiliation(s)
- Fernanda Visioli
- Oral Pathology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | |
Collapse
|
44
|
PIR2/Rnf144B regulates epithelial homeostasis by mediating degradation of p21WAF1 and p63. Oncogene 2012; 32:4758-65. [DOI: 10.1038/onc.2012.497] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 09/06/2012] [Accepted: 09/13/2012] [Indexed: 12/28/2022]
|
45
|
Abstract
A daily moisturizing routine is a vital part of the management of patients with atopic dermatitis and other dry skin conditions. The composition of the moisturizer determines whether the treatment strengthens or deteriorates the skin barrier function, which may have consequences for the outcome of the dermatitis. One might expect that a patient's impaired skin barrier function should improve in association with a reduction in the clinical signs of dryness. Despite visible relief of the dryness symptoms, however, the abnormal transepidermal water loss has been reported to remain high, or even to increase under certain regimens, whereas other moisturizers improve skin barrier function. Differing outcomes have also been reported in healthy skin: some moisturizers produce deterioration in skin barrier function and others improve the skin. Possible targets for barrier-influencing moisturizing creams include the intercellular lipid bilayers, where the fraction of lipids forming a fluid phase might be changed due to compositional or organizational changes. Other targets are the projected size of the corneocytes or the thickness of the stratum corneum. Moisturizers with barrier-improving properties may delay relapse of dermatitis in patients with atopic dermatitis. In a worst-case scenario, treatment with moisturizing creams could increase the risks of dermatitis and asthma.
Collapse
Affiliation(s)
- Marie Lodén
- Eviderm Institute AB, Bergshamra Allé 9, SE-170 77 Solna, Sweden.
| |
Collapse
|
46
|
Li LP, Cheng WB, Li H, Li W, Yang H, Wen DH, Tang YD. Expression of Proteasome Activator REGγ in Human Laryngeal Carcinoma and Associations with Tumor Suppressor Proteins. Asian Pac J Cancer Prev 2012; 13:2699-703. [DOI: 10.7314/apjcp.2012.13.6.2699] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
47
|
Hönnemann J, Sanz-Moreno A, Wolf E, Eilers M, Elsässer HP. Miz1 is a critical repressor of cdkn1a during skin tumorigenesis. PLoS One 2012; 7:e34885. [PMID: 22509363 PMCID: PMC3324535 DOI: 10.1371/journal.pone.0034885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 03/08/2012] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Miz1 forms repressive DNA-binding complexes with the Myc, Gfi-1 and Bcl-6 oncoproteins. Known target genes of these complexes encode the cyclin-dependent kinase inhibitors (CKIs) cdkn2b (p15Ink4), cdkn1a (p21Cip1), and cdkn1c (p57Kip2). Whether Miz1-mediated repression is important for control of cell proliferation in vivo and for tumor formation is unknown. Here we show that deletion of the Miz1 POZ domain, which is critical for Miz1 function, restrains the development of skin tumors in a model of chemically-induced, Ras-dependent tumorigenesis. While the stem cell compartment appears unaffected, interfollicular keratinocytes lacking functional Miz1 exhibit a reduced proliferation and an accelerated differentiation of the epidermis in response to the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Tumorigenesis, proliferation and normal differentiation are restored in animals lacking cdkn1a, but not in those lacking cdkn2b. Our data demonstrate that Miz1-mediated attenuation of cell cycle arrest pathways via repression of cdkn1a has a critical role during tumorigenesis in the skin.
Collapse
Affiliation(s)
- Jan Hönnemann
- Department of Cytobiology, Philipps-University Marburg, Germany
| | | | - Elmar Wolf
- Theodor-Boveri-Institute, Biocentre, University of Würzburg, Germany
| | - Martin Eilers
- Theodor-Boveri-Institute, Biocentre, University of Würzburg, Germany
| | | |
Collapse
|
48
|
Pathogenetic and Prognostic Significance of Inactivation of RASSF Proteins in Human Hepatocellular Carcinoma. Mol Biol Int 2012; 2012:849874. [PMID: 22548173 PMCID: PMC3323848 DOI: 10.1155/2012/849874] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/26/2012] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent solid tumors worldwide, with limited treatment options and a dismal prognosis. Thus, there is a strong need to expand the basic and translational research on this deadly disease in order to improve the prognosis of HCC patients. Although the etiologic factors responsible for HCC development have been identified, the molecular pathogenesis of liver cancer remains poorly understood. Recent evidence has shown the frequent downregulation of Ras association domain family (RASSF) proteins both in the early and late stages of hepatocarcinogenesis. Here, we summarize the data available on the pathogenetic role of inactivation of RASSF proteins in liver cancer, the molecular mechanisms responsible for suppression of RASSF proteins in HCC, and the possible clinical implications arising from these discoveries. Altogether, the data indicate that inactivation of the RASSF1A tumor suppressor is ubiquitous in human liver cancer, while downregulation of RASSF2 and RASSF5 proteins is limited to specific HCC subsets. Also, the present findings speak in favour of therapeutic strategies aimed at reexpressing RASSF1A, RASSF2, and RASSF5 genes and/or inactivating the RASSF cellular inhibitors for the treatment of human liver cancer.
Collapse
|
49
|
Zhang M, Gan L, Ren GS. REGγ is a strong candidate for the regulation of cell cycle, proliferation and the invasion by poorly differentiated thyroid carcinoma cells. Braz J Med Biol Res 2012; 45:459-65. [PMID: 22415115 PMCID: PMC3854288 DOI: 10.1590/s0100-879x2012007500035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 02/28/2012] [Indexed: 11/22/2022] Open
Abstract
REGγ is a proteasome activator that facilitates the degradation of small peptides. Abnormally high expression of REGγ has been observed in thyroid carcinomas. The purpose of the present study was to explore the role of REGγ in poorly differentiated thyroid carcinoma (PDTC). For this purpose, small interfering RNA (siRNA) was introduced to down-regulate the level of REGγ in the PDTC cell line SW579. Down-regulation of REGγ at the mRNA and protein levels was confirmed by RT-PCR and Western blot analyses. FACS analysis revealed cell cycle arrest at the G1/S transition, the MTT assay showed inhibition of cell proliferation, and the Transwell assay showed restricted cell invasion. Furthermore, the expression of the p21 protein was increased, the expression of proliferating cell nuclear antigen (PCNA) protein decreased, and the expression of the p27 protein was unchanged as shown by Western blot analyses. REGγ plays a critical role in the cell cycle, proliferation and invasion of SW579 cells. The alteration of p21 and PCNA proteins related to the down-regulation of REGγ suggests that p21 and PCNA participate in the process of REGγ regulation of cell cycle progression and cell proliferation. Thus, targeting REGγ has a therapeutic potential in the management of PDTC patients.
Collapse
Affiliation(s)
- M Zhang
- Department of General Surgery, The First Affiliated Hospital, Chongqing Medical University, China
| | | | | |
Collapse
|
50
|
Wang WM, Liu ST, Huang SM, Lin WS, Chen SG, Chang YL. Zac1 functional interactions mediate AP-1 transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:2050-60. [DOI: 10.1016/j.bbamcr.2011.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 08/05/2011] [Accepted: 08/05/2011] [Indexed: 02/06/2023]
|