1
|
Kim G, Jang SK, Ahn SH, Kim S, Park CS, Seong MK, Kim HA, Bae S, Lee JH, Kim H, Jin HO, Park IC. Proapoptotic role of CDK1 in overcoming paclitaxel resistance in ovarian cancer cells in response to combined treatment with paclitaxel and duloxetine. Cancer Cell Int 2024; 24:409. [PMID: 39702300 DOI: 10.1186/s12935-024-03607-8] [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: 06/13/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Paclitaxel resistance and recurrence are major obstacles in ovarian cancer, which is the leading cause of death among gynecologic cancers. During cancer cell progression, cyclin-dependent kinase 1 (CDK1) drives cells through the G2 phase and into mitosis. In this study, we demonstrated that CDK1 played a crucial role in switching paclitaxel-resistant ovarian cancer cells from mitotic arrest to apoptosis following combined treatment with paclitaxel and duloxetine, an antidepressant known as a serotonin-norepinephrine reuptake inhibitor (SNRI). METHODS Cell viability was assessed by MTT assay. Apoptotic cell death and mitochondrial membrane potential (MMP) were detected by flow cytometry. Protein expression levels were explored using western blotting. Mitochondrial and cytosolic fractionation were performed to determine the mitochondrial localization of proteins. Immunofluorescence was used to detect protein expression levels and localization. RESULTS Combined treatment with paclitaxel and duloxetine induced apoptotic cell death in paclitaxel-resistant ovarian cancer cells. We suggested that combined treatment of these drugs induced CDK1 activation and increased mitochondrial localization of activated CDK1, which caused phosphorylation of the antiapoptotic Bcl-2 and Bcl-xL proteins. Selective CDK1 inhibitors blocked Bcl-2 and Bcl-xL phosphorylation induced by paclitaxel and duloxetine, and strongly suppressed apoptotic cell death. Furthermore, we demonstrated that S6K is a potential upstream mediator of the proapoptotic activation of CDK1. CONCLUSION Taken together, switching CDK1 to a proapoptotic role through the combination of paclitaxel and duloxetine could overcome paclitaxel resistance in ovarian cancer cells, providing promising therapeutic strategies for treating paclitaxel-resistant ovarian cancer.
Collapse
Affiliation(s)
- Gyeongmi Kim
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Se-Kyeong Jang
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - Se Hee Ahn
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - Selim Kim
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Chan Sub Park
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - Min-Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - Hyun-Ah Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Korea
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, Seoul, 05029, Korea
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea
| | - In-Chul Park
- Division of Fusion Radiology Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Korea.
| |
Collapse
|
2
|
Pluta AJ, Studniarek C, Murphy S, Norbury CJ. Cyclin-dependent kinases: Masters of the eukaryotic universe. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 15:e1816. [PMID: 37718413 PMCID: PMC10909489 DOI: 10.1002/wrna.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/19/2023]
Abstract
A family of structurally related cyclin-dependent protein kinases (CDKs) drives many aspects of eukaryotic cell function. Much of the literature in this area has considered individual members of this family to act primarily either as regulators of the cell cycle, the context in which CDKs were first discovered, or as regulators of transcription. Until recently, CDK7 was the only clear example of a CDK that functions in both processes. However, new data points to several "cell-cycle" CDKs having important roles in transcription and some "transcriptional" CDKs having cell cycle-related targets. For example, novel functions in transcription have been demonstrated for the archetypal cell cycle regulator CDK1. The increasing evidence of the overlap between these two CDK types suggests that they might play a critical role in coordinating the two processes. Here we review the canonical functions of cell-cycle and transcriptional CDKs, and provide an update on how these kinases collaborate to perform important cellular functions. We also provide a brief overview of how dysregulation of CDKs contributes to carcinogenesis, and possible treatment avenues. This article is categorized under: RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes RNA Processing > 3' End Processing RNA Processing > Splicing Regulation/Alternative Splicing.
Collapse
Affiliation(s)
| | | | - Shona Murphy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Chris J. Norbury
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| |
Collapse
|
3
|
Kwon NY, Sung SH, Sung HK, Park JK. Anticancer Activity of Bee Venom Components against Breast Cancer. Toxins (Basel) 2022; 14:toxins14070460. [PMID: 35878198 PMCID: PMC9318616 DOI: 10.3390/toxins14070460] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 12/10/2022] Open
Abstract
While the survival rate has increased due to treatments for breast cancer, the quality of life has decreased because of the side effects of chemotherapy. Various toxins are being developed as alternative breast cancer treatments, and bee venom is drawing attention as one of them. We analyzed the effect of bee venom and its components on breast cancer cells and reviewed the mechanism underlying the anticancer effects of bee venom. Data up to March 2022 were searched from PubMed, EMBASE, OASIS, KISS, and Science Direct online databases, and studies that met the inclusion criteria were reviewed. Among 612 studies, 11 were selected for this research. Diverse drugs were administered, including crude bee venom, melittin, phospholipase A2, and their complexes. All drugs reduced the number of breast cancer cells in proportion to the dose and time. The mechanisms of anticancer effects included cytotoxicity, apoptosis, cell targeting, gene expression regulation, and cell lysis. Summarily, bee venom and its components exert anticancer effects on human breast cancer cells. Depending on the mechanisms of anticancer effects, side effects are expected to be reduced by using various vehicles. Bee venom and its components have the potential to prevent and treat breast cancer in the future.
Collapse
Affiliation(s)
- Na-Yoen Kwon
- Department of Obstetrics and Gynecology, College of Korean Medicine, Ga-Chon University, Seongnam-si 13120, Korea;
| | - Soo-Hyun Sung
- Department of Policy Development, National Institute of Korean Medicine Development, Seoul 04554, Korea;
| | - Hyun-Kyung Sung
- Department of Korean Medicine Pediatrics, School of Korean Medicine, Semyung University, Jecheon 27136, Korea
- Correspondence: (H.-K.S.); (J.-K.P.); Tel.: +82-43-841-1739 (H.-K.S.); +82-55-360-5978 (J.-K.P.)
| | - Jang-Kyung Park
- Department of Korean Medicine Obstetrics and Gynecology, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea
- Correspondence: (H.-K.S.); (J.-K.P.); Tel.: +82-43-841-1739 (H.-K.S.); +82-55-360-5978 (J.-K.P.)
| |
Collapse
|
4
|
Coccè V, Rimoldi I, Facchetti G, Ciusani E, Alessandri G, Signorini L, Sisto F, Giannì A, Paino F, Pessina A. In Vitro Activity of Monofunctional Pt-II Complex Based on 8-Aminoquinoline against Human Glioblastoma. Pharmaceutics 2021; 13:pharmaceutics13122101. [PMID: 34959382 PMCID: PMC8704014 DOI: 10.3390/pharmaceutics13122101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/18/2022] Open
Abstract
A new cationic Pt(II) complex bearing 8-aminoquinoline as chelating ligand (called Pt-8AQ) was evaluated against two human carcinomas, one mesothelioma, and three glioblastoma cell lines. The in vitro comparison to the clinically approved CisPt showed a minor activity of Pt-8AQ against carcinoma and mesothelioma, whereas a significant activity of Pt-8AQ was observed on the proliferation of the three glioblastoma cell lines (U87-MG IC50 = 3.68 ± 0.69 µM; U373-MG IC50 = 11.53 ± 0.16 µM; U138-MG IC50 = 8.05 ± 0.23 µM) that was higher than that observed with the clinically approved CisPt (U87-MG IC50 = 7.27 + 1.80 µM; U373-MG IC50 = 22.69 ± 0.05 µM; U138-MG IC50 = 32.1 ± 4.44 µM). Cell cycle analysis proved that Pt-8AQ significantly affected the cell cycle pattern by increasing the apoptotic cells represented by the sub G0/G1 region related with a downregulation of p53 and Bcl-2. Moreover, an NMR investigation of Pt-8AQ interaction with 9-EtG, GSH, and Mets7 excluded DNA as the main target, suggesting a novel mechanism of action. Our study demonstrated the high stability of Pt-8AQ after incubation at 37 °C and a significant antineoplastic activity on glioblastomas. These features also make Pt-8AQ a good candidate for developing a new selective advanced cell chemotherapy approach in combination with MSCs.
Collapse
Affiliation(s)
- Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Isabella Rimoldi
- Department of Pharmaceutical Science, University of Milan, Via Golgi 19, 20133 Milan, Italy;
| | - Giorgio Facchetti
- Department of Pharmaceutical Science, University of Milan, Via Golgi 19, 20133 Milan, Italy;
- Correspondence: (G.F.); (A.P.)
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute Carlo Besta, 20133 Milan, Italy;
| | - Giulio Alessandri
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy;
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Aldo Giannì
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
- Maxillo-Facial and Dental Unit, Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Paino
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, CRC StaMeTec, 20133 Milan, Italy; (V.C.); (G.A.); (F.S.); (A.G.); (F.P.)
- Correspondence: (G.F.); (A.P.)
| |
Collapse
|
5
|
Boldinova EO, Yudkina AV, Shilkin ES, Gagarinskaya DI, Baranovskiy AG, Tahirov TH, Zharkov DO, Makarova AV. Translesion activity of PrimPol on DNA with cisplatin and DNA-protein cross-links. Sci Rep 2021; 11:17588. [PMID: 34475447 PMCID: PMC8413282 DOI: 10.1038/s41598-021-96692-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
Human PrimPol belongs to the archaeo-eukaryotic primase superfamily of primases and is involved in de novo DNA synthesis downstream of blocking DNA lesions and non-B DNA structures. PrimPol possesses both DNA/RNA primase and DNA polymerase activities, and also bypasses a number of DNA lesions in vitro. In this work, we have analyzed translesion synthesis activity of PrimPol in vitro on DNA with an 1,2-intrastrand cisplatin cross-link (1,2-GG CisPt CL) or a model DNA–protein cross-link (DpCL). PrimPol was capable of the 1,2-GG CisPt CL bypass in the presence of Mn2+ ions and preferentially incorporated two complementary dCMPs opposite the lesion. Nucleotide incorporation was stimulated by PolDIP2, and yeast Pol ζ efficiently extended from the nucleotides inserted opposite the 1,2-GG CisPt CL in vitro. DpCLs significantly blocked the DNA polymerase activity and strand displacement synthesis of PrimPol. However, PrimPol was able to reach the DpCL site in single strand template DNA in the presence of both Mg2+ and Mn2+ ions despite the presence of the bulky protein obstacle.
Collapse
Affiliation(s)
- Elizaveta O Boldinova
- Institute of Molecular Genetics, National Research Center «Kurchatov Institute», Kurchatov sq. 2, Moscow, Russia, 123182
| | - Anna V Yudkina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk, Russia, 630090
| | - Evgeniy S Shilkin
- Institute of Molecular Genetics, National Research Center «Kurchatov Institute», Kurchatov sq. 2, Moscow, Russia, 123182
| | - Diana I Gagarinskaya
- Institute of Molecular Genetics, National Research Center «Kurchatov Institute», Kurchatov sq. 2, Moscow, Russia, 123182
| | - Andrey G Baranovskiy
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Tahir H Tahirov
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Dmitry O Zharkov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentiev Avenue, Novosibirsk, Russia, 630090.,Novosibirsk State University, 2 Pirogova St., Novosibirsk, Russia, 630090
| | - Alena V Makarova
- Institute of Molecular Genetics, National Research Center «Kurchatov Institute», Kurchatov sq. 2, Moscow, Russia, 123182.
| |
Collapse
|
6
|
McKay LK, White JP. The AMPK/p27 Kip1 Pathway as a Novel Target to Promote Autophagy and Resilience in Aged Cells. Cells 2021; 10:cells10061430. [PMID: 34201101 PMCID: PMC8229180 DOI: 10.3390/cells10061430] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Once believed to solely function as a cyclin-dependent kinase inhibitor, p27Kip1 is now emerging as a critical mediator of autophagy, cytoskeletal dynamics, cell migration and apoptosis. During periods of metabolic stress, the subcellular location of p27Kip1 largely dictates its function. Cytoplasmic p27Kip1 has been found to be promote cellular resilience through autophagy and anti-apoptotic mechanisms. Nuclear p27Kip1, however, inhibits cell cycle progression and makes the cell susceptible to quiescence, apoptosis, and/or senescence. Cellular location of p27Kip1 is regulated, in part, by phosphorylation by various kinases, including Akt and AMPK. Aging promotes nuclear localization of p27Kip1 and a predisposition to senescence or apoptosis. Here, we will review the role of p27Kip1 in healthy and aging cells with a particular emphasis on the interplay between autophagy and apoptosis.
Collapse
Affiliation(s)
- Lauren K. McKay
- Adams School of Dentistry, UNC Chapel Hill, Chapel Hill, NC 27599, USA;
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
| | - James P. White
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Department of Medicine, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Correspondence:
| |
Collapse
|
7
|
Jan BL, Ahmad A, Khan A, Rehman MU, Alkharfy KM. Protective effect of chrysin, a flavonoid, on the genotoxic activity of carboplatin in mice. Drug Chem Toxicol 2021; 45:2146-2152. [PMID: 33829940 DOI: 10.1080/01480545.2021.1908752] [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: 10/21/2022]
Abstract
Carboplatin is amongst the most commonly used anticancer drugs for the management of several human malignancies. However, it has displayed genotoxic properties against normal cells. Evaluation of natural products for their protective effects against chemotherapeutic drug induced toxicity has been growing in recent years. A naturally occurring flavonoid, chrysin, has strong antioxidant abilities and protects against DNA impairment. This study used multiple assays to evaluate the levels of damage to DNA in normal cells and to examine any possible protective role of chrysin against such damage. Male BALB/c mice were administered chrysin orally in two doses of 20 and 40 mg/kg for 10 consecutive days and then a single injection of carboplatin [90 mg/kg body weight (b.w.)] was administered intraperitoneally to induce carboplatin toxicity. 24 h after the carboplatin injection, mice were sacrificed. DNA damage was evaluated using several genotoxicity tests (8-Hydroxydeoxy-guanosine marker, comet assay, micronucleus test, and chromosomal aberration assay) to identify diverse types of damage to the DNA. The results suggest that pretreatment with chrysin significantly decreased the level of DNA damage caused by carboplatin probably due to its potent antioxidant traits. Therefore, chrysin can be considered to be developed as a chemoprotective agent against chemotherapy associated side-effects.
Collapse
Affiliation(s)
- Basit L Jan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Altaf Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid M Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
The Affinity of Carboplatin to B-Vitamins and Nucleobases. Int J Mol Sci 2021; 22:ijms22073634. [PMID: 33807309 PMCID: PMC8037198 DOI: 10.3390/ijms22073634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
Platinum compounds have found wide application in the treatment of various types of cancer and carboplatin is one of the main platinum-based drugs used as antitumor agents. The anticancer activity of carboplatin arises from interacting with DNA and inducing programmed cell death. However, such interactions may occur with other chemical compounds, such as vitamins containing aromatic rings with lone-pair orbitals, which reduces the anti-cancer effect of carboplatin. The most important aspect of the conducted research was related to the evaluation of carboplatin affinity to vitamins from the B group and the potential impact of such interactions on the reduction of therapeutic capabilities of carboplatin in anticancer therapy. Realized computations, including estimation of Gibbs Free Energies, allowed for the identification of the most reactive molecule, namely vitamin B6 (pyridoxal phosphate). In this case, the computational estimations indicating carboplatin reactivity were confirmed by spectrophotometric measurements.
Collapse
|
9
|
Ying X, Che X, Wang J, Zou G, Yu Q, Zhang X. CDK1 serves as a novel therapeutic target for endometrioid endometrial cancer. J Cancer 2021; 12:2206-2215. [PMID: 33758599 PMCID: PMC7974891 DOI: 10.7150/jca.51139] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Endometrial cancer (EC) is one of the most common and prevalent gynecologic malignancies worldwide. The aim of this study was to identify a novel therapeutic target for endometrioid endometrial cancer. Materials and Methods: Bioinformatic analysis was performed and CDK1 was screen out as one of the hub genes in the pathogenesis of EC. Immunohistochemistry was used to verify the expression of CDK1 in endometrial cancer tissue. Cell viability and colony formation were used to study the effects of CDK1 on the proliferation and colony formation of endometrial cancer cells in vitro. Apoptosis and cell cycle assays were used to elucidate the mechanism of CDK1 affecting cell proliferation. Tumor xenograft transplantation assay was performed to show the effects of CDK1 on the growth of endometrial cancer cells in vivo. Results: CDK1 was over expressed in endometrioid endometrial cancer, and accumulation of cytoplasmic CDK1 was associated with histological grade of EC. CDK1 promoted endometrial cancer cell growth and colony formation in vitro. The inhibition of CDK1 activity induced cell apoptosis and caused G2/M phase arrest of cell cycle in endometrial cancer cells. The inhibition of CDK1 activity also inhibited endometrial cancer growth in xenograft models. Conclusion: CDK1 was involved in the pathogenesis of endometrioid endometrial cancer and provided a novel therapeutic target for endometrioid endometrial cancer.
Collapse
Affiliation(s)
- Xue Ying
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Xuan Che
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006.,Jiaxing University Affiliated Women and Children Hospital, Jiaxing, Zhejiang, P.R. China, 314000
| | - Jianzhang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Gen Zou
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Qin Yu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| | - Xinmei Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China, 310006
| |
Collapse
|
10
|
Katukam V, Rupula K, Rao Beedu S. Synthesis and characterisation of novel biopolymer stabilised organic Pt-nanocomposite: assessment of its antioxidant and antitumour properties. IET Nanobiotechnol 2020; 14:889-898. [PMID: 33399123 PMCID: PMC8676258 DOI: 10.1049/iet-nbt.2020.0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 07/30/2020] [Accepted: 08/19/2020] [Indexed: 11/19/2022] Open
Abstract
Green synthesis of organic Pt-nanocomposite was accomplished using carboplatin as a precursor and novel biopolymer - gum kondagogu (GK) as a reducing agent. The synthesised GK stabilised organic Pt-nanocomposite (GKCPt NC) was characterised by different analytical techniques such as ultraviolet-visible spectroscopy, nanoparticle analyser, scanning electron microscopy and energy dispersive X-ray analysis, X-ray diffraction (XRD), Fourier-transform infrared spectroscopy, transmission electron microscopy (TEM), X-ray photoelectron spectroscopy (XPS) and inductively coupled plasma optical emission spectrophotometer. The XRD pattern established the amorphous nature of GKCPt NC. TEM analysis revealed the homogeneous, monodisperse and spherical nature, with Pt metal size of 3.08 ± 0.62 nm. The binding energy at 71.2 and 74.6 eV show the presence of metallic platinum, Pt(0) confirmed by XPS studies. Further, in vitro radical scavenging and antitumour activity of GKCPt NC have been investigated. In comparison to GK and carboplatin, GKCPt NC showed superior 1, 1-diphenyl-2-picrylhydrazyle activity of 87.82%, whereas 2, 2-azinobis-(3-ethylbenzthinzoline-6-sulphonic acid) activity was 38.50%, respectively. In vitro studies of the antitumour property of GK, GKCPt NC and carboplatin were evaluated by potato disc tumour bioassay model. The efficacy of synthesised GKCPt NC concentration (IC50) on tumour inhibition was found to be 2.04-fold lower as compared to carboplatin. Overall, the synthesised GKCPt NC shows both antitumour and antioxidant properties when compared to the original drug - carboplatin and might have promising applications in cancer therapy.
Collapse
Affiliation(s)
- Vani Katukam
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad 500 007, Telangana State, India
| | - Karuna Rupula
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad 500 007, Telangana State, India
| | - Sashidhar Rao Beedu
- Department of Biochemistry, University College of Science, Osmania University, Hyderabad 500 007, Telangana State, India.
| |
Collapse
|
11
|
Analysis of genetically driven alternative splicing identifies FBXO38 as a novel COPD susceptibility gene. PLoS Genet 2019; 15:e1008229. [PMID: 31269066 PMCID: PMC6634423 DOI: 10.1371/journal.pgen.1008229] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 07/16/2019] [Accepted: 06/04/2019] [Indexed: 11/19/2022] Open
Abstract
While many disease-associated single nucleotide polymorphisms (SNPs) are associated with gene expression (expression quantitative trait loci, eQTLs), a large proportion of complex disease genome-wide association study (GWAS) variants are of unknown function. Some of these SNPs may contribute to disease by regulating gene splicing. Here, we investigate whether SNPs that are associated with alternative splicing (splice QTL or sQTL) can identify novel functions for existing GWAS variants or suggest new associated variants in chronic obstructive pulmonary disease (COPD). RNA sequencing was performed on whole blood from 376 subjects from the COPDGene Study. Using linear models, we identified 561,060 unique sQTL SNPs associated with 30,333 splice sites corresponding to 6,419 unique genes. Similarly, 708,928 unique eQTL SNPs involving 15,913 genes were detected at 10% FDR. While there is overlap between sQTLs and eQTLs, 55.3% of sQTLs are not eQTLs. Co-localization analysis revealed that 7 out of 21 loci associated with COPD (p<1x10-6) in a published GWAS have at least one shared causal variant between the GWAS and sQTL studies. Among the genes identified to have splice sites associated with top GWAS SNPs was FBXO38, in which a novel exon was discovered to be protective against COPD. Importantly, the sQTL in this locus was validated by qPCR in both blood and lung tissue, demonstrating that splice variants relevant to lung tissue can be identified in blood. Other identified genes included CDK11A and SULT1A2. Overall, these data indicate that analysis of alternative splicing can provide novel insights into disease mechanisms. In particular, we demonstrated that SNPs in a known COPD GWAS locus on chromosome 5q32 influence alternative splicing in the gene FBXO38.
Collapse
|
12
|
Perla V, Nadimi M, Reddy R, Hankins GR, Nimmakayala P, Harris RT, Valluri J, Sirbu C, Reddy UK. Effect of ghost pepper on cell proliferation, apoptosis, senescence and global proteomic profile in human renal adenocarcinoma cells. PLoS One 2018; 13:e0206183. [PMID: 30379886 PMCID: PMC6209291 DOI: 10.1371/journal.pone.0206183] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/07/2018] [Indexed: 12/19/2022] Open
Abstract
Chili peppers are an important constituent of many foods and contain medicinally valuable compounds, such as capsaicin and dihydrocapsaicin. As various dietary botanicals have anticancer properties, this study was aimed to examine the effect of Ghost pepper (Bhut Jolokia), one of the hottest chili peppers in the world, on cell proliferation, apoptosis, senescence and the global proteomic profile in human renal cell adenocarcinoma in vitro. 769-P human renal adenocarcinoma cells were cultured on RPMI-1640 media supplemented with fetal bovine serum (10%) and antibiotic-antimycotic solution (1%). Treatment stock solutions were prepared in ethanol. Cell proliferation was tested with phenol red-free media with capsaicin (0-400 μM), dihydrocapsaicin (0-400 μM), capsaicin + dihydrocapsaicin (5:1), and dry Ghost peppers (0-3 g L-1) for 24, 48 and 72 h. Polycaspase and senescence associated-beta-galactosidase (SA-beta-gal) activities were tested with capsaicin (400 μM), dihydrocapsaicin (400 μM), capsaicin (400 μM) + dihydrocapsaicin (80 μM), and ghost pepper (3 g L-1) treatments. Global proteomic profile of cells in control and ghost pepper treatment (3 g L-1) was analyzed after 6 h by a shotgun proteomic approach using tandem mass spectrometry. At 24 h after treatment (24 HAT), relative to control, cell proportion with capsaicin (400 μM), dihydrocapsaicin (400 μM), capsaicin (400 μM) + dihydrocapsaicin (80 μM), and ghost pepper (3 g L-1) treatments was reduced to 36%, 18%, 33% and 20%, respectively, and further reduced at 48 and 72 HAT. All treatments triggered an early polycaspase response. SA-beta-gal activity was normal or suppressed with all treatments. About 68,220 protein isoforms were identified by shotgun proteomic approach. Among these, about 8.2% were significantly affected by ghost pepper. Ghost pepper regulated various proteins involved in intrinsic and extrinsic apoptotic pathways, Ras, Rb/E2F, p53, TGF-beta, WNT-beta catenin, and calcium induced cell death pathways. Ghost pepper also induced changes in proteins related to methylation, acetylation, genome stability, cell cycle check points, carbohydrate, protein and other metabolism and cellular mechanisms. Ghost pepper exhibited antiproliferation activity by inducing apoptosis through a complex network of proteins in human renal cell adenocarcinoma in vitro.
Collapse
Affiliation(s)
- Venu Perla
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Marjan Nadimi
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Rishi Reddy
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Gerald R. Hankins
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Padma Nimmakayala
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Robert T. Harris
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| | - Jagan Valluri
- Department of Biological Sciences, One John Marshall Drive, Marshall University, Huntington, West Virginia, United States of America
| | - Cristian Sirbu
- Center for Cancer Research, Charleston Area Medical Center, SE, Charleston, West Virginia, United States of America
| | - Umesh K. Reddy
- Gus R. Douglass Land-Grant Institute and Department of Biology, West Virginia State University, Institute, West Virginia, United States of America
| |
Collapse
|
13
|
Kalimutho M, Sinha D, Jeffery J, Nones K, Srihari S, Fernando WC, Duijf PH, Vennin C, Raninga P, Nanayakkara D, Mittal D, Saunus JM, Lakhani SR, López JA, Spring KJ, Timpson P, Gabrielli B, Waddell N, Khanna KK. CEP55 is a determinant of cell fate during perturbed mitosis in breast cancer. EMBO Mol Med 2018; 10:e8566. [PMID: 30108112 PMCID: PMC6127888 DOI: 10.15252/emmm.201708566] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 07/15/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022] Open
Abstract
The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in in vivo models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers.
Collapse
Affiliation(s)
- Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Debottam Sinha
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Jessie Jeffery
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | - Sriganesh Srihari
- Computational Systems Biology Laboratory, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | | | - Pascal Hg Duijf
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Prahlad Raninga
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | | | - Deepak Mittal
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Jodi M Saunus
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
| | - Sunil R Lakhani
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
- School of Medicine, The University of Queensland, Herston, Qld, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, Qld, Australia
| | - J Alejandro López
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Kevin J Spring
- Liverpool Clinical School, University of Western Sydney, Liverpool, NSW, Australia
- Ingham Institute, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW, Australia
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Brian Gabrielli
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| |
Collapse
|
14
|
Sun L, Moore E, Berman R, Clavijo PE, Saleh A, Chen Z, Van Waes C, Davies J, Friedman J, Allen CT. WEE1 kinase inhibition reverses G2/M cell cycle checkpoint activation to sensitize cancer cells to immunotherapy. Oncoimmunology 2018; 7:e1488359. [PMID: 30288354 DOI: 10.1080/2162402x.2018.1488359] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Indexed: 01/29/2023] Open
Abstract
Intrinsic resistance to cytotoxic T-lymphocyte (CTL) killing limits responses to immune activating anti-cancer therapies. Here, we established that activation of the G2/M cell cycle checkpoint results in tumor cell cycle pause and protection from granzyme B-induced cell death. This was reversed with WEE1 kinase inhibition, leading to enhanced CTL killing of antigen-positive tumor cells. Similarly, but at a later time point, cell cycle pause following TNFα exposure was reversed with WEE1 kinase inhibition, leading to CTL transmembrane TNFα-dependent induction of apoptosis and necroptosis in bystander antigen-negative tumor cells. Results were reproducible in models of oral cavity carcinoma, melanoma and colon adenocarcinoma harboring variable Tp53 genomic alterations. WEE1 kinase inhibition sensitized tumors to PD-1 mAb immune checkpoint blockade in vivo, resulting in CD8+-dependent rejection of established tumors harboring antigen-positive or mixed antigen-positive and negative tumor cells. Together, these data describe activation of the G2/M cell cycle checkpoint in response to early and late CTL products as a mechanism of resistance to CTL killing, and provide pre-clinical rationale for the clinical combination of agents that inhibit cell cycle checkpoints and activate anti-tumor immunity.
Collapse
Affiliation(s)
- Lillian Sun
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Ellen Moore
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Rose Berman
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Paul E Clavijo
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Anthony Saleh
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - John Davies
- Experimental Transplantation and Immunology Branch, National Cancer Institute, NIH, Bethesda, USA
| | - Jay Friedman
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| | - Clint T Allen
- Translational Tumor Immunology Program, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, NIH, Bethesda, USA
| |
Collapse
|
15
|
Tran TT, Strozynski M, Thiede B. Quantitative phosphoproteome analysis of cisplatin-induced apoptosis in Jurkat T cells. Proteomics 2017; 17. [DOI: 10.1002/pmic.201600470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/12/2017] [Accepted: 04/27/2017] [Indexed: 01/06/2023]
Affiliation(s)
- The Trung Tran
- Department of Biosciences; University of Oslo; Oslo Norway
| | | | - Bernd Thiede
- Department of Biosciences; University of Oslo; Oslo Norway
| |
Collapse
|
16
|
Biedermann S, Harashima H, Chen P, Heese M, Bouyer D, Sofroni K, Schnittger A. The retinoblastoma homolog RBR1 mediates localization of the repair protein RAD51 to DNA lesions in Arabidopsis. EMBO J 2017; 36:1279-1297. [PMID: 28320735 PMCID: PMC5412766 DOI: 10.15252/embj.201694571] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 11/13/2022] Open
Abstract
The retinoblastoma protein (Rb), which typically functions as a transcriptional repressor of E2F‐regulated genes, represents a major control hub of the cell cycle. Here, we show that loss of the Arabidopsis Rb homolog RETINOBLASTOMA‐RELATED 1 (RBR1) leads to cell death, especially upon exposure to genotoxic drugs such as the environmental toxin aluminum. While cell death can be suppressed by reduced cell‐proliferation rates, rbr1 mutant cells exhibit elevated levels of DNA lesions, indicating a direct role of RBR1 in the DNA‐damage response (DDR). Consistent with its role as a transcriptional repressor, we find that RBR1 directly binds to and represses key DDR genes such as RADIATION SENSITIVE 51 (RAD51), leaving it unclear why rbr1 mutants are hypersensitive to DNA damage. However, we find that RBR1 is also required for RAD51 localization to DNA lesions. We further show that RBR1 is itself targeted to DNA break sites in a CDKB1 activity‐dependent manner and partially co‐localizes with RAD51 at damage sites. Taken together, these results implicate RBR1 in the assembly of DNA‐bound repair complexes, in addition to its canonical function as a transcriptional regulator.
Collapse
Affiliation(s)
- Sascha Biedermann
- Department of Molecular Mechanisms of Phenotypic Plasticity, Institut de Biologie Moléculaire des Plantes du Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France.,Department of Developmental Biology, Biozentrum Klein Flottbek University of Hamburg, Hamburg, Germany
| | | | - Poyu Chen
- Department of Developmental Biology, Biozentrum Klein Flottbek University of Hamburg, Hamburg, Germany
| | - Maren Heese
- Department of Developmental Biology, Biozentrum Klein Flottbek University of Hamburg, Hamburg, Germany
| | - Daniel Bouyer
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS UMR 8197-INSERM U 1024, Paris, France
| | - Kostika Sofroni
- Department of Developmental Biology, Biozentrum Klein Flottbek University of Hamburg, Hamburg, Germany
| | - Arp Schnittger
- Department of Molecular Mechanisms of Phenotypic Plasticity, Institut de Biologie Moléculaire des Plantes du Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France .,Department of Developmental Biology, Biozentrum Klein Flottbek University of Hamburg, Hamburg, Germany
| |
Collapse
|
17
|
Godin-Heymann N, Brabetz S, Murillo MM, Saponaro M, Santos CR, Lobley A, East P, Chakravarty P, Matthews N, Kelly G, Jordan S, Castellano E, Downward J. Tumour-suppression function of KLF12 through regulation of anoikis. Oncogene 2016; 35:3324-34. [PMID: 26455320 PMCID: PMC4929484 DOI: 10.1038/onc.2015.394] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 08/11/2015] [Accepted: 09/05/2015] [Indexed: 12/14/2022]
Abstract
Suppression of detachment-induced cell death, known as anoikis, is an essential step for cancer metastasis to occur. We report here that expression of KLF12, a member of the Kruppel-like family of transcription factors, is downregulated in lung cancer cell lines that have been selected to grow in the absence of cell adhesion. Knockdown of KLF12 in parental cells results in decreased apoptosis following cell detachment from matrix. KLF12 regulates anoikis by promoting the cell cycle transition through S phase and therefore cell proliferation. Reduced expression levels of KLF12 results in increased ability of lung cancer cells to form tumours in vivo and is associated with poorer survival in lung cancer patients. We therefore identify KLF12 as a novel metastasis-suppressor gene whose loss of function is associated with anoikis resistance through control of the cell cycle.
Collapse
Affiliation(s)
- N Godin-Heymann
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
| | - S Brabetz
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
| | - M M Murillo
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
- The Institute of Cancer Research, London, UK
| | - M Saponaro
- Mechanisms of Gene Transcription Laboratory, Cancer Research UK London Research Institute, Clare Hall Laboratories, Hertfordshire, UK
| | - C R Santos
- Translational Cancer Therapeutics, Cancer Research UK London Research Institute, London, UK
| | - A Lobley
- Bioinformatics and Biostatistics Laboratories, Cancer Research UK London Research Institute, London, UK
| | - P East
- Bioinformatics and Biostatistics Laboratories, Cancer Research UK London Research Institute, London, UK
| | - P Chakravarty
- Bioinformatics and Biostatistics Laboratories, Cancer Research UK London Research Institute, London, UK
| | - N Matthews
- Advanced Sequencing Facility, Cancer Research UK London Research Institute, London, UK
| | - G Kelly
- Bioinformatics and Biostatistics Laboratories, Cancer Research UK London Research Institute, London, UK
| | - S Jordan
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
| | - E Castellano
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
| | - J Downward
- Signal Transduction, Cancer Research UK London Research Institute, London, UK
- The Institute of Cancer Research, London, UK
| |
Collapse
|
18
|
WANG JIN, YIN HAILIN, PANANDIKAR ASHWINI, GANDHI VARSHA, SEN SUBRATA. Elevated cyclin A associated kinase activity promotes sensitivity of metastatic human cancer cells to DNA antimetabolite drug. Int J Oncol 2015; 47:782-790. [PMID: 26058363 PMCID: PMC4501665 DOI: 10.3892/ijo.2015.3037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/04/2015] [Indexed: 11/16/2022] Open
Abstract
Drug resistance is a major obstacle in successful systemic therapy of metastatic cancer. We analyzed the involvement of cell cycle regulatory proteins in eliciting response to N (phosphonoacetyl)-L-aspartate (PALA), an inhibitor of de novo pyrimidine synthesis, in two metastatic variants of human cancer cell line MDA-MB-435 isolated from lung (L-2) and brain (Br-1) in nude mouse, respectively. L-2 and Br-l cells markedly differed in their sensitivity to PALA. While both cell types displayed an initial S phase delay/arrest, Br-l cells proliferated but most L-2 cells underwent apoptosis. There was distinct elevation in cyclin A, and phosphorylated Rb proteins concomitant with decreased expression of bcl-2 protein in the PALA treated L-2 cells undergoing apoptosis. Markedly elevated cyclin A associated and cdk2 kinase activities together with increased E2F1-DNA binding were detected in these L-2 cells. Induced ectopic cyclin A expression sensitized Br-l cells to PALA by activating an apoptotic pathway. Our findings demonstrate that elevated expression of cyclin A and associated kinase can activate an apoptotic pathway in cells exposed to DNA antimetabolites. Abrogation of this pathway can lead to resistance against these drugs in metastatic variants of human carcinoma cells.
Collapse
Affiliation(s)
- JIN WANG
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - HAILIN YIN
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - ASHWINI PANANDIKAR
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - VARSHA GANDHI
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - SUBRATA SEN
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Program in Human and Molecular Genetics, University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
19
|
Oh YN, Jin S, Park HJ, Kim BW, Kwon HJ. Anti-oxidative and Anti-cancer Activities of Treculia africana Extract in Human Colon Adenocarcinoma HT29 Cells. ACTA ACUST UNITED AC 2015. [DOI: 10.5352/jls.2015.25.5.515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
20
|
Osman AMM, Al-Malki HS, Al-Harthi SE, El-Hanafy AA, Elashmaoui HM, Elshal MF. Modulatory role of resveratrol on cytotoxic activity of cisplatin, sensitization and modification of cisplatin resistance in colorectal cancer cells. Mol Med Rep 2015; 12:1368-74. [PMID: 25815689 DOI: 10.3892/mmr.2015.3513] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 08/29/2014] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-associated mortality worldwide. Cisplatin (CIS) is one of the most active cytotoxic agents in current use and it has proven efficacy against various human malignancies. However, its clinical usefulness has been restricted by detrimental side effects, including nephrotoxicity and myelosuppression. The aim of the present study was to attempt to decrease the required dose of CIS, in order to minimize its side effects, and increase its capability to arrest, delay or reverse carcinogenesis. In addition, the present study aimed to ameliorate CIS-resistance in CRC cells, using the natural compound resveratrol (RSVL). RSVL (3,4', 5-trihydroxy-trans-stilbene) is a naturally occurring polyphenol present in the roots of white hellebore (Veratrum grandiflorum O. Loes) and extracted from >70 other plant species. RSVL can exert antioxidant and anti-inflammatory activities, and it has been shown to be active in the regulation of numerous cellular events associated with carcinogenesis. The present study evaluated the effects of RSVL on sensitization of both parent and CIS-resistant HCT-116 CRC cells to the action of cisplatin. The CIS was administered at a dose of 5 and 20 µg/ml, and CIS cytotoxicity, apoptosis, cell cycle and cisplatin cellular uptake were examined in the presence and absence of RSVL (15 µg/ml). RSVL treatment showed anti-proliferative effects and enhanced the cytotoxic effects of cis against the growth of both parent and CIS-resistant HCT-116 CRC cells, with a half maximal inhibitory concentration of 4.20 µg/ml and 4.72 µg/ml respectively. RSVL also induced a significant increase in the early apoptosis fraction and enhanced the subsequent apoptotic effects of CIS. The cellular uptake of CIS was significantly increased in the presence of RSVL, as compared with CIS treatment alone, and RSVL treatment sensitized the CIS-resistant HCT-116 cells. In conclusion, RSVL treatment increased the cytotoxic activity of CIS against the growth of both parent and CIS-resistant HCT-116 CRC cells.
Collapse
Affiliation(s)
- Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hamdan S Al-Malki
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sameer E Al-Harthi
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amr A El-Hanafy
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hassan M Elashmaoui
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed F Elshal
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
21
|
Sung WW, Lin YM, Wu PR, Yen HH, Lai HW, Su TC, Huang RH, Wen CK, Chen CY, Chen CJ, Yeh KT. High nuclear/cytoplasmic ratio of Cdk1 expression predicts poor prognosis in colorectal cancer patients. BMC Cancer 2014; 14:951. [PMID: 25511643 PMCID: PMC4302138 DOI: 10.1186/1471-2407-14-951] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/11/2014] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Cdk1 (cyclin-dependent kinase 1) is critical regulator of the G2-M checkpoint. Cyclin-dependent kinase pathways are considered possible targets for cancer treatment; however, the prognostic role of Cdk1 in colorectal cancer is still controversial. Therefore, we attempted to determine the impact of Cdk1 on the clinical outcome of colorectal cancer patients to further identify its role in colorectal cancer. METHODS Cdk1 immunoreactivity was analyzed by immunohistochemistry (IHC) in 164 cancer specimens from primary colorectal cancer patients. The medium follow-up time after surgery was 3.7 years (range: 0.01 to 13.10 years). The prognostic value of Cdk1 on overall survival was determined by Kaplan-Meier analysis and Cox proportional hazard models. RESULTS All samples displayed detectable Cdk1 expression with predominant location in the cytoplasm and nucleus. A high Cdk1 nuclear/cytoplasmic (N/C) expression ratio was correlated with poor overall survival (5-year survival rate: 26.3% vs 46.9%, N/C ratio ≥1.5 vs N/C ratio <1.5, log-rank p = 0.027). Accordingly, a Cdk1 N/C expression ratio ≥1.5 was identified as an independent risk factor by multivariate analysis (hazard ratio = 1.712, P = 0.039). CONCLUSIONS We suggest that Cdk1 N/C expression ratio determined by IHC staining could be an independent prognostic marker for colorectal cancer.
Collapse
Affiliation(s)
- Wen-Wei Sung
- />School of Medicine, Chung Shan Medical University, Taichuang, Taiwan
| | - Yueh-Min Lin
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
- />Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Pei-Ru Wu
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu-Heng Yen
- />School of Medicine, Chung Shan Medical University, Taichuang, Taiwan
- />Department of Gastroenterology, Changhua Christian Hospital, Changhua, Taiwan
| | - Hung-Wen Lai
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
- />School of Medicine, National Yang Ming University, Taipei, Taiwan
| | - Tzu-Cheng Su
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ren-Hung Huang
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Chun-Kai Wen
- />School of Medicine, Chung Shan Medical University, Taichuang, Taiwan
| | - Chia-Yu Chen
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Jung Chen
- />School of Medicine, Chung Shan Medical University, Taichuang, Taiwan
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
- />Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Kun-Tu Yeh
- />School of Medicine, Chung Shan Medical University, Taichuang, Taiwan
- />Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
22
|
Sousa GFD, Wlodarczyk SR, Monteiro G. Carboplatin: molecular mechanisms of action associated with chemoresistance. BRAZ J PHARM SCI 2014. [DOI: 10.1590/s1984-82502014000400004] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Carboplatin is a derivative of cisplatin; it has a similar mechanism of action, but differs in terms of structure and toxicity. It was approved by the FDA in the 1980s and since then it has been widely used in the treatment of several tumor types. This agent is characterized by its ability to generate lesions in DNA through the formation of adducts with platinum, thereby inhibiting replication and transcription and leading to cell death. However, its use can lead to serious inconvenience arising from the development of resistance that some patients acquire during treatment, limiting the scope of its full potential. Currently, the biochemical mechanisms related to resistance are not precisely known. Therefore, knowledge of pathways associated with resistance caused by carboplatin exposure may provide valuable clues for more efficient rational drug design in platinum-based therapy and the development of new therapeutic strategies. In this narrative review, we discuss some of the known mechanisms of resistance to platinum-based drugs, especially carboplatin.
Collapse
|
23
|
Han Z, Ku L, Zhang Z, Zhang J, Guo S, Liu H, Zhao R, Ren Z, Zhang L, Su H, Dong L, Chen Y. QTLs for seed vigor-related traits identified in maize seeds germinated under artificial aging conditions. PLoS One 2014; 9:e92535. [PMID: 24651614 PMCID: PMC3961396 DOI: 10.1371/journal.pone.0092535] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/23/2014] [Indexed: 01/22/2023] Open
Abstract
High seed vigor is important for agricultural production due to the associated potential for increased growth and productivity. However, a better understanding of the underlying molecular mechanisms is required because the genetic basis for seed vigor remains unknown. We used single-nucleotide polymorphism (SNP) markers to map quantitative trait loci (QTLs) for four seed vigor traits in two connected recombinant inbred line (RIL) maize populations under four treatment conditions during seed germination. Sixty-five QTLs distributed between the two populations were identified and a meta-analysis was used to integrate genetic maps. Sixty-one initially identified QTLs were integrated into 18 meta-QTLs (mQTLs). Initial QTLs with contribution to phenotypic variation values of R2>10% were integrated into mQTLs. Twenty-three candidate genes for association with seed vigor traits coincided with 13 mQTLs. The candidate genes had functions in the glycolytic pathway and in protein metabolism. QTLs with major effects (R2>10%) were identified under at least one treatment condition for mQTL2, mQTL3-2, and mQTL3-4. Candidate genes included a calcium-dependent protein kinase gene (302810918) involved in signal transduction that mapped in the mQTL3-2 interval associated with germination energy (GE) and germination percentage (GP), and an hsp20/alpha crystallin family protein gene (At5g51440) that mapped in the mQTL3-4 interval associated with GE and GP. Two initial QTLs with a major effect under at least two treatment conditions were identified for mQTL5-2. A cucumisin-like Ser protease gene (At5g67360) mapped in the mQTL5-2 interval associated with GP. The chromosome regions for mQTL2, mQTL3-2, mQTL3-4, and mQTL5-2 may be hot spots for QTLs related to seed vigor traits. The mQTLs and candidate genes identified in this study provide valuable information for the identification of additional quantitative trait genes.
Collapse
Affiliation(s)
- Zanping Han
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
- College of Agronomy, Henan University of Science and Technology, Luoyang, China
| | - Lixia Ku
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Zhenzhen Zhang
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Jun Zhang
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - ShuLei Guo
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Haiying Liu
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Ruifang Zhao
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Zhenzhen Ren
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Liangkun Zhang
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Huihui Su
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Lei Dong
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
| | - Yanhui Chen
- College of Agronomy, Synergetic Innovation Center of Henan Grain Crops and National Key Laboratory of wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou, China
- * E-mail:
| |
Collapse
|
24
|
Zhou L, Cai X, Han X, Xu N, Chang DC. CDK1 switches mitotic arrest to apoptosis by phosphorylating Bcl-2/Bax family proteins during treatment with microtubule interfering agents. Cell Biol Int 2014; 38:737-46. [PMID: 24677263 DOI: 10.1002/cbin.10259] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 01/20/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Lingli Zhou
- Department of Endocrinology and Metabolism; Peking University People's Hospital; Beijing China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism; Peking University People's Hospital; Beijing China
| | - Xueyao Han
- Department of Endocrinology and Metabolism; Peking University People's Hospital; Beijing China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen 518055 P. R. China
| | - Donald C. Chang
- Division of Life Science; The Hong Kong University of Science and Technology; Clear Water Bay Hong Kong China
| |
Collapse
|
25
|
Zhao J, Han SX, Ma JL, Ying X, Liu P, Li J, Wang L, Zhang Y, Ma J, Zhang L, Zhu Q. The role of CDK1 in apoptin-induced apoptosis in hepatocellular carcinoma cells. Oncol Rep 2013; 30:253-9. [PMID: 23619525 DOI: 10.3892/or.2013.2426] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 03/05/2013] [Indexed: 11/05/2022] Open
Abstract
Apoptin, a small protein derived from the chicken anemia virus, specifically induces apoptosis in transformed cells or tumor cells but not in normal cells. Thus, apoptin is involved in a general, tumor-specific pathway. Apoptin-induced apoptosis presumably requires additional interaction partners that activate specific signaling pathways in cancer cells. A number of molecules interact with apoptin and play an important role in the nuclear localization of apoptin or its tumor-selective cytotoxicity. Our data indicated that apoptin selectively kills HepG2 hepatocellular carcinoma (HCC) cells but has no effect on the normal liver cell line HL-7702. Analyses of human HCC tissue samples confirmed that CDK1 (cyclin-dependent kinase 1) activity was detected in primary malignancies but not in healthy paraneoplastic tissues. shRNA knockdown of CDK1 significantly reduced the tumor-specific killing effects of apoptin, suggesting that CDK1 plays an important role in the regulation of apoptin-induced apoptosis. Furthermore, the majority of apoptin translocated to the cytoplasm from the nucleus after knockdown of CDK1. Collectively, our results revealed for the first time that apoptin interacts with CDK1 in the complex process of tumorigenesis. The link between CDK1 and apoptin may be a novel cellular signaling pathway to modulate apoptosis in cancer; therefore, apoptin may have pharmacological potential to be directly employed for cancer therapy.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical college, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chang KH, Vincent F, Shah K. Deregulated Cdk5 triggers aberrant activation of cell cycle kinases and phosphatases inducing neuronal death. J Cell Sci 2012; 125:5124-37. [PMID: 22899714 DOI: 10.1242/jcs.108183] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aberrant activation of cell cycle proteins is believed to play a critical role in Alzheimer's disease (AD) pathogenesis; although, the molecular mechanisms leading to their activation in diseased neurons remain elusive. The goal of this study was to investigate the mechanistic link between Cdk5 deregulation and cell cycle re-activation in β-amyloid(1-42) (Aβ(1-42))-induced neurotoxicity. Using a chemical genetic approach, we identified Cdc25A, Cdc25B and Cdc25C as direct Cdk5 substrates in mouse brain lysates. We show that deregulated Cdk5 directly phosphorylates Cdc25A, Cdc25B and Cdc25C at multiple sites, which not only increases their phosphatase activities but also facilitates their release from 14-3-3 inhibitory binding. Cdc25A, Cdc25B and Cdc25C in turn activate Cdk1, Cdk2 and Cdk4 kinases causing neuronal death. Selective inhibition of Cdk5 abrogates Cdc25 and Cdk activations in Aβ(1-42)-treated neurons. Similarly, phosphorylation-resistant mutants of Cdc25 isoforms at Cdk5 sites are defective in activating Cdk1, Cdk2 and Cdk4 in Aβ(1-42)-treated primary cortical neurons, emphasizing a major role of Cdk5 in the activation of Cdc25 isoforms and Cdks in AD pathogenesis. These results were further confirmed in human AD clinical samples, which had higher Cdc25A, Cdc25B and Cdc25C activities that were coincident with increased Cdk5 activity, as compared to age-matched controls. Inhibition of Cdk5 confers the highest neuroprotection against Aβ(1-42) toxicity, whereas inhibition of Cdc25 isoforms was partially neuroprotective, further emphasizing a decisive role of Cdk5 deregulation in cell-cycle-driven AD neuronal death.
Collapse
Affiliation(s)
- Kuei-Hua Chang
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | | | | |
Collapse
|
27
|
Matés JM, Segura JA, Alonso FJ, Márquez J. Oxidative stress in apoptosis and cancer: an update. Arch Toxicol 2012; 86:1649-65. [PMID: 22811024 DOI: 10.1007/s00204-012-0906-3] [Citation(s) in RCA: 252] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/03/2012] [Indexed: 02/07/2023]
Abstract
The oxygen paradox tells us that oxygen is both necessary for aerobic life and toxic to all life forms. Reactive oxygen species (ROS) touch every biological and medical discipline, especially those involving proliferative status, supporting the idea that active oxygen may be increased in tumor cells. In fact, metabolism of oxygen and the resulting toxic byproducts can cause cancer and death. Efforts to counteract the damage caused by ROS are gaining acceptance as a basis for novel therapeutic approaches, and the field of prevention of cancer is experiencing an upsurge of interest in medically useful antioxidants. Apoptosis is an important means of regulating cell numbers in the developing cell system, but it is so important that it must be controlled. Normal cell death in homeostasis of multicellular organisms is mediated through tightly regulated apoptotic pathways that involve oxidative stress regulation. Defective signaling through these pathways can contribute to both unbalance in apoptosis and development of cancer. Finally, in this review, we discuss new knowledge about recent tools that provide powerful antioxidant strategies, and designing methods to deliver to target cells, in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- José M Matés
- Department of Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, Málaga, Spain.
| | | | | | | |
Collapse
|
28
|
Mechanisms of Cisplatin-Induced Apoptosis and of Cisplatin Sensitivity: Potential of BIN1 to Act as a Potent Predictor of Cisplatin Sensitivity in Gastric Cancer Treatment. Int J Surg Oncol 2012; 2012:862879. [PMID: 22778941 PMCID: PMC3384945 DOI: 10.1155/2012/862879] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/05/2012] [Indexed: 12/12/2022] Open
Abstract
Cisplatin is the most important and efficacious chemotherapeutic agent for the treatment of advanced gastric cancer. Cisplatin forms inter- and intrastrand crosslinked DNA adducts and its cytotoxicity is mediated by propagation of DNA damage recognition signals to downstream pathways involving ATR, p53, p73, and mitogen-activated protein kinases, ultimately resulting in apoptosis. Cisplatin resistance arises through a multifactorial mechanism involving reduced drug uptake, increased drug inactivation, increased DNA damage repair, and inhibition of transmission of DNA damage recognition signals to the apoptotic pathway. In addition, a new mechanism has recently been revealed, in which the oncoprotein c-Myc suppresses bridging integrator 1 (BIN1), thereby releasing poly(ADP-ribose)polymerase 1, which results in increased DNA repair activity and allows cancer cells to acquire cisplatin resistance. The present paper focuses on the molecular mechanisms of cisplatin-induced apoptosis and of cisplatin resistance, in particular on the involvement of BIN1 in the maintenance of cisplatin sensitivity.
Collapse
|
29
|
Chen M, Gutierrez GJ, Ronai ZA. The anaphase-promoting complex or cyclosome supports cell survival in response to endoplasmic reticulum stress. PLoS One 2012; 7:e35520. [PMID: 22539978 PMCID: PMC3335095 DOI: 10.1371/journal.pone.0035520] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 03/18/2012] [Indexed: 11/23/2022] Open
Abstract
The anaphase-promoting complex or cyclosome (APC/C) is a multi-subunit ubiquitin ligase that regulates exit from mitosis and G1 phase of the cell cycle. Although the regulation and function of APC/CCdh1 in the unperturbed cell cycle is well studied, little is known of its role in non-genotoxic stress responses. Here, we demonstrate the role of APC/CCdh1 (APC/C activated by Cdh1 protein) in cellular protection from endoplasmic reticulum (ER) stress. Activation of APC/CCdh1 under ER stress conditions is evidenced by Cdh1-dependent degradation of its substrates. Importantly, the activity of APC/CCdh1 maintains the ER stress checkpoint, as depletion of Cdh1 by RNAi impairs cell cycle arrest and accelerates cell death following ER stress. Our findings identify APC/CCdh1 as a regulator of cell cycle checkpoint and cell survival in response to proteotoxic insults.
Collapse
Affiliation(s)
- Meifan Chen
- Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Gustavo J. Gutierrez
- Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Ze'ev A. Ronai
- Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Nagy O, Pál M, Udvardy A, Shirras CA, Boros I, Shirras AD, Deák P. lemmingA encodes the Apc11 subunit of the APC/C in Drosophila melanogaster that forms a ternary complex with the E2-C type ubiquitin conjugating enzyme, Vihar and Morula/Apc2. Cell Div 2012; 7:9. [PMID: 22417125 PMCID: PMC3372440 DOI: 10.1186/1747-1028-7-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 03/14/2012] [Indexed: 12/16/2022] Open
Abstract
Background Ubiquitin-dependent protein degradation is a critical step in key cell cycle events, such as metaphase-anaphase transition and mitotic exit. The anaphase promoting complex/cyclosome (APC/C) plays a pivotal role in these transitions by recognizing and marking regulatory proteins for proteasomal degradation. Its overall structure and function has been elucidated mostly in yeasts and mammalian cell lines. The APC/C is, however, a multisubunit assembly with at least 13 subunits and their function and interaction within the complex is still relatively uncharacterized, particularly in metazoan systems. Here, lemming (lmg) mutants were used to study the APC/C subunit, Apc11, and its interaction partners in Drosophila melanogaster. Results The lmg gene was initially identified through a pharate adult lethal P element insertion mutation expressing developmental abnormalities and widespread apoptosis in larval imaginal discs and pupal abdominal histoblasts. Larval neuroblasts were observed to arrest mitosis in a metaphase-like state with highly condensed, scattered chromosomes and frequent polyploidy. These neuroblasts contain high levels of both cyclin A and cyclin B. The lmg gene was cloned by virtue of the lmg03424 P element insertion which is located in the 5' untranslated region. The lemming locus is transcribed to give a 2.0 kb mRNA that contains two ORFs, lmgA and lmgB. The lmgA ORF codes for a putative protein with more than 80% sequence homology to the APC11 subunit of the human APC/C. The 85 amino acid protein also contains a RING-finger motif characteristic of known APC11 subunits. The lmgA ORF alone was sufficient to rescue the lethal and mitotic phenotypes of the lmg138 null allele and to complement the temperature sensitive lethal phenotype of the APC11-myc9 budding yeast mutant. The LmgA protein interacts with Mr/Apc2, and they together form a binding site for Vihar, the E2-C type ubiquitin conjugating enzyme. Despite being conserved among Drosophila species, the LmgB protein is not required for viability or fertility. Conclusions Our work provides insight into the subunit structure of the Drosophila APC/C with implications for its function. Based on the presented data, we suggest that the Lmg/Apc11 subunit recruits the E2-C type ubiquitin conjugating enzyme, Vihar, to the APC/C together with Mr/Apc2 by forming a ternary complex.
Collapse
Affiliation(s)
- Olga Nagy
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| | | | | | | | | | | | | |
Collapse
|
31
|
Xin X, Lin XH, Zhou YC, Chen XL, Liu X, Lu XX. Proteome analysis of maize seeds: the effect of artificial ageing. PHYSIOLOGIA PLANTARUM 2011; 143:126-38. [PMID: 21707636 DOI: 10.1111/j.1399-3054.2011.01497.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Previous understanding of the mechanism of seed ageing is largely based on observations on imbibed seeds rather than dry seeds. The present research was conducted to investigate whether seed ageing has effects on the dry seeds through proteome analysis. Maize (Zea mays cv. Dabaitou) seeds were artificially aged at 50°C (13.58% moisture content) for 5 or 13 days, and the total protein was extracted from embryos of the dry seeds. Two-dimensional electrophoresis was performed and the differentially expressed proteins were identified by matrix-assisted laser desorption ionization-time of flight mass spectrometry. A total of 40 proteins were identified, in which 16 proteins were upregulated, indicating that artificial ageing affected the proteome of the dry seeds. Proteomic studies revealed that the signal transduction and transcription were disturbed by artificial ageing, which might lead to reduced protection against ageing. Artificial ageing also increased proteases and broke down stored proteins, impaired metabolism and energy supply, and ultimately resulted in seed deterioration. Proteins involved in metabolism and energy were the largest downregulated protein group, with regard to glycolysis, tricarboxylic acid cycle, the electron transport chain and oxidative phosphorylation. The downregulation of these proteins, together with reduction in the specific activity of glucose-6-phosphate dehydrogenase, and the content of glucose 6-phosphate, pyruvic acid and ATP in aged seeds, suggested the important roles of the mobilization of stored carbohydrates and energy supply in seed ageing and seed vigor. The present work provides new information about the proteomic changes during seed ageing and provides a possible mechanism for seed deterioration.
Collapse
Affiliation(s)
- Xia Xin
- National Genebank, Institute of Crop Science, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | | | | | | | | | | |
Collapse
|
32
|
Zhang C, Elkahloun AG, Robertson M, Gills JJ, Tsurutani J, Shih JH, Fukuoka J, Hollander MC, Harris CC, Travis WD, Jen J, Dennis PA. Loss of cytoplasmic CDK1 predicts poor survival in human lung cancer and confers chemotherapeutic resistance. PLoS One 2011; 6:e23849. [PMID: 21887332 PMCID: PMC3161069 DOI: 10.1371/journal.pone.0023849] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/26/2011] [Indexed: 01/15/2023] Open
Abstract
The dismal lethality of lung cancer is due to late stage at diagnosis and inherent therapeutic resistance. The incorporation of targeted therapies has modestly improved clinical outcomes, but the identification of new targets could further improve clinical outcomes by guiding stratification of poor-risk early stage patients and individualizing therapeutic choices. We hypothesized that a sequential, combined microarray approach would be valuable to identify and validate new targets in lung cancer. We profiled gene expression signatures during lung epithelial cell immortalization and transformation, and showed that genes involved in mitosis were progressively enhanced in carcinogenesis. 28 genes were validated by immunoblotting and 4 genes were further evaluated in non-small cell lung cancer tissue microarrays. Although CDK1 was highly expressed in tumor tissues, its loss from the cytoplasm unexpectedly predicted poor survival and conferred resistance to chemotherapy in multiple cell lines, especially microtubule-directed agents. An analysis of expression of CDK1 and CDK1-associated genes in the NCI60 cell line database confirmed the broad association of these genes with chemotherapeutic responsiveness. These results have implications for personalizing lung cancer therapy and highlight the potential of combined approaches for biomarker discovery.
Collapse
Affiliation(s)
- Chunyu Zhang
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Abdel G. Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew Robertson
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joell J. Gills
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junji Tsurutani
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Medical Oncology Department, Kinki University School of Medicine, Osaka-Sayama, Osaka, Japan
| | - Joanna H. Shih
- Biometric Research Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junya Fukuoka
- Laboratory of Population Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Surgical Pathology, Toyama University Hospital, Toyama, Japan
| | - M. Christine Hollander
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - William D. Travis
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jin Jen
- Laboratory of Population Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Phillip A. Dennis
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Apoptosis-regulated survival of primarily extravascular cells in proliferative active poststent neointima. Cardiovasc Pathol 2010; 19:353-60. [DOI: 10.1016/j.carpath.2009.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 06/24/2009] [Accepted: 07/22/2009] [Indexed: 11/23/2022] Open
|
34
|
Kuga T, Nozaki N, Matsushita K, Nomura F, Tomonaga T. Phosphorylation statuses at different residues of lamin B2, B1, and A/C dynamically and independently change throughout the cell cycle. Exp Cell Res 2010; 316:2301-12. [PMID: 20580708 DOI: 10.1016/j.yexcr.2010.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 04/23/2010] [Accepted: 05/18/2010] [Indexed: 11/17/2022]
Abstract
Lamins, major components of the nuclear lamina, undergo phosphorylation at multiple residues during cell cycle progression, but their detailed phosphorylation kinetics remain largely undetermined. Here, we examined changes in the phosphorylation of major phosphorylation residues (Thr14, Ser17, Ser385, Ser387, and Ser401) of lamin B2 and the homologous residues of lamin B1, A/C during the cell cycle using novel antibodies to the site-specific phosphorylation. The phosphorylation levels of these residues independently changed during the cell cycle. Thr14 and Ser17 were phosphorylated during G(2)/M phase to anaphase/telophase. Ser385 was persistently phosphorylated during mitosis to G(1) phase, whereas Ser387 was phosphorylated discontinuously in prophase and G(1) phase. Ser401 phosphorylation was enhanced in the G(1)/S boundary. Immunoprecipitation using the phospho-antibodies suggested that metaphase-phosphorylation at Thr14, Ser17, and Ser385 of lamins occurred simultaneously, whereas G(1)-phase phosphorylation at Ser385 and Ser387 occurred in distinct pools or with different timings. Additionally, we showed that lamin B2 phosphorylated at Ser17, but not Ser385, Ser387 and Ser401, was exclusively non-ionic detergent soluble, depolymerized forms in growing cells, implicating specific involvement of Ser17 phosphorylation in lamin depolymerization and nuclear envelope breakdown. These results suggest that the phosphorylations at different residues of lamins might play specific roles throughout the cell cycle.
Collapse
Affiliation(s)
- Takahisa Kuga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Ibaraki, Osaka 567-0085, Japan.
| | | | | | | | | |
Collapse
|
35
|
Chou ST, Yen YC, Lee CM, Chen MS. Pro-apoptotic role of Cdc25A: activation of cyclin B1/Cdc2 by the Cdc25A C-terminal domain. J Biol Chem 2010; 285:17833-45. [PMID: 20368335 DOI: 10.1074/jbc.m109.078386] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Cdc25A is a dual specificity protein phosphatase that activates cyclin/cyclin-dependent protein kinase (Cdk) complexes by removing inhibitory phosphates from conserved threonine and tyrosine in Cdks. To address how Cdc25A promotes apoptosis, Jurkat cells were treated with staurosporine, an apoptosis inducer. Upon staurosporine treatment, a Cdc25A C-terminal 37-kDa fragment, designated C37, was generated by caspase cleavage at Asp-223. Thr-507 in C37 became dephosphorylated, which prevented 14-3-3 binding, as shown previously. C37 exhibited higher phosphatase activity than full-length Cdc25A. C37 with alanine substitution for Thr-507 (C37/T507A) that imitated the cleavage product during staurosporine treatment interacted with Cdc2, Cdk2, cyclin A, and cyclin B1 and markedly activated cyclin B1/Cdc2. The dephosphorylation of Thr-507 might expose the Cdc2/Cdk2-docking site in C37. C37/T507A also induced apoptosis in Jurkat and K562 cells, resulting from activating cyclin B1/Cdc2 but not Cdk2. Thus, this study reveals that Cdc25A is a pro-apoptotic protein that amplifies staurosporine-induced apoptosis through the activation of cyclin B1/Cdc2 by its C-terminal domain.
Collapse
Affiliation(s)
- Sung-Tau Chou
- National Institute of Cancer Research, National Health Research Institutes, Number 35, Keyan Road, Zhunan Town, Miaoli County 35053, Taiwan
| | | | | | | |
Collapse
|
36
|
Valente ST, Gilmartin GM, Venkatarama K, Arriagada G, Goff SP. HIV-1 mRNA 3' end processing is distinctively regulated by eIF3f, CDK11, and splice factor 9G8. Mol Cell 2009; 36:279-89. [PMID: 19854136 DOI: 10.1016/j.molcel.2009.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 06/01/2009] [Accepted: 10/06/2009] [Indexed: 01/30/2023]
Abstract
A genetic screen previously identified the N-terminal 91 amino acids of the eukaryotic initiation factor 3 subunit f (N91-eIF3f) as a potent inhibitor of HIV-1 replication. Overexpression of N91-eIF3f or full-length eIF3f reduced the level of HIV-1 mRNAs in the infected cell. Here we show that N91-eIF3f and eIF3f act by specifically blocking the 3' end processing of the HIV-1 pre-mRNA both in vivo and in vitro. Furthermore, the results suggest that eIF3f mediates this restriction of HIV-1 expression through the previously unsuspected involvement of a set of factors that includes eIF3f, the SR protein 9G8, and the cyclin-dependent kinase 11 (CDK11). eIF3f affects HIV-1 3' end processing by modulating the sequence-specific recognition of the HIV-1 pre-mRNA by 9G8.
Collapse
Affiliation(s)
- Susana T Valente
- Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, HHSC 1310c, 701 West 168th Street, New York, NY 10032, USA.
| | | | | | | | | |
Collapse
|
37
|
Jeon TW, Kim CH, Lee SK, Ko GS, Yoo JW, Ha HW, Kang WK, Jeong HG, Kang MJ, Lee ES, Jeong TC. Immunotoxicological Investigation of 1-furan-2-yl-3-pyridin-2-yl-propenone in Female BALB/c Mice. Biomol Ther (Seoul) 2009. [DOI: 10.4062/biomolther.2009.17.4.446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
38
|
Identification and characterization of a novel anticancer agent with selectivity against deleted in pancreatic cancer locus 4 (DPC4)-deficient pancreatic and colon cancer cells. Pancreas 2009; 38:551-7. [PMID: 19276868 DOI: 10.1097/mpa.0b013e31819d7415] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVES The deleted in pancreatic cancer locus 4 (DPC4)/SMAD4 tumor suppressor gene is frequently inactivated in pancreatic (approximately 55%) and colorectal cancers (approximately 30%). Like other tumor suppressor genes, the loss-of-function mutations found in the DPC4 gene are specific to cancer cells. This provides an attractive and unique opportunity for therapeutic intervention. The aim of this study was to identify and characterize small molecules that selectively kill DPC4-deficient cancer cells. METHODS An unbiased chemical screening using isogenic cell lines that only differ in the DPC4 gene was carried out to identify positive hits. Selected hits were further verified in additional isogenic cell lines and characterized in cancer cells using several different cellular assays. RESULTS A lead molecule, UA62784, was identified to be selectively cytotoxic against cancer cells with deficient DPC4. UA62784 preferentially induces cell cycle arrest and apoptosis in cells with deficient DPC4. It also selectively reduces the clonogenicity of DPC4-deficient cells on soft agar when compared with cells with wild type DPC4. We further demonstrate that UA62784 induces CDC2 kinase activity preferentially in DPC4-negative cells. CONCLUSIONS UA62784 is a small molecule that selectively kills DPC4-deficient cancer cells. Its potent activity and relatively low molecular weight make it a decent candidate for further lead optimization.
Collapse
|
39
|
Alvarez-Breckenridge C, Kaur B, Chiocca EA. Pharmacologic and chemical adjuvants in tumor virotherapy. Chem Rev 2009; 109:3125-40. [PMID: 19462957 PMCID: PMC2790404 DOI: 10.1021/cr900048k] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Christopher Alvarez-Breckenridge
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, Columbus, Ohio
| | - Balveen Kaur
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, Columbus, Ohio
| | - E. Antonio Chiocca
- Dardinger Laboratory for Neuro-oncology and Neurosciences, Department of Neurological Surgery, James Comprehensive Cancer Center and The Ohio State University Medical Center, Columbus, Ohio
| |
Collapse
|
40
|
Abstract
In summary, apoptosis is an important concept in understanding many facets of human reproduction. Recent advances in the understanding of molecular mechanisms of apoptosis will allow us to understand this physiologically important process. How can the modulation of this process be applied to human reproduction? Studies to further understand the abnormalities of apoptosis, either too much or too little, may lead to a better understanding of the clinical problems in human reproduction.We summarize future directions towards further understanding the roles of apoptotic processes in human reproduction in Table 3. The diseases listed in Table 3 are problems which could be approached from the apoptosis point of view. With further study using this concept as the lens, new diagnostic tools or therapies may be developed for these problems.
Collapse
|
41
|
Shi J, Hershey JWB, Nelson MA. Phosphorylation of the eukaryotic initiation factor 3f by cyclin-dependent kinase 11 during apoptosis. FEBS Lett 2009; 583:971-7. [PMID: 19245811 PMCID: PMC2666973 DOI: 10.1016/j.febslet.2009.02.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 02/13/2009] [Accepted: 02/17/2009] [Indexed: 11/25/2022]
Abstract
eIF3f is a subunit of eukaryotic initiation factor 3 (eIF3). We previously showed that eIF3f is phosphorylated by cyclin dependent kinase 11 (CDK11(p46)) which is an important effector in apoptosis. Here, we identified a second eIF3f phosphorylation site (Thr119) by CDK11(p46) during apoptosis. We demonstrated that eIF3f is directly phosphorylated by CDK11(p46) in vivo. Phosphorylation of eIF3f plays an important role in regulating its function in translation and apoptosis. Phosphorylation of eIF3f enhances the association of eIF3f with the core eIF3 subunits during apoptosis. Our data suggested that eIF3f may inhibit translation by increasing the binding to the eIF3 complex during apoptosis.
Collapse
Affiliation(s)
- Jiaqi Shi
- Department of Surgery, Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA
| | - John W. B. Hershey
- Department of Biological Chemistry, School of Medicine, University of California, Davis, CA 95616, USA
| | - Mark A. Nelson
- Department of Pathology, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
42
|
Skirrow RC, Veldhoen N, Domanski D, Helbing CC. Roscovitine inhibits thyroid hormone-induced tail regression of the frog tadpole and reveals a role for cyclin C/Cdk8 in the establishment of the metamorphic gene expression program. Dev Dyn 2008; 237:3787-97. [DOI: 10.1002/dvdy.21800] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
43
|
Van Zijl C, Lottering ML, Steffens F, Joubert A. In vitro effects of 2-methoxyestradiol on MCF-12A and MCF-7 cell growth, morphology and mitotic spindle formation. Cell Biochem Funct 2008; 26:632-42. [PMID: 18508385 DOI: 10.1002/cbf.1489] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The influence of 2-methoxyestradiol (2ME) was investigated on cell growth, morphology and spindle formation in a tumorigenic (MCF-7) and non-tumorigenic (MCF-12A) epithelial breast cell line. Inhibition of cell growth was more pronounced in the MCF-7 cells compared to the MCF-12A cells following 2ME treatment. Dose-dependent studies (10(-5)-10(-9) M) revealed that 10(-6) M 2ME inhibited cell growth by 44% in MCF-12A cells and by 84% in MCF-7 cells (p-value < 0.05). 2ME-treated MCF-7 cells showed abnormal metaphase cells, membrane blebbing, apoptotic cells and disrupted spindle formation. These observations were either absent or less prominent in MCF-12A cells. 2ME had no effect on the length of the cell cycle between S-phase and the time a mitotic peak was reached in either cell line but MCF-7 cells were blocked in mitosis with no statistically significant alterations in the phosphorylation status of Cdc25C. Nevertheless, Cdc2 activity was significantly increased in MCF-7 cells compared to MCF-12A cells (p-value < 0.05). The results indicate that 2ME disrupts mitotic spindle formation and enhances Cdc2 kinase activity, leading to persistence of the spindle checkpoint and thus prolonged metaphase arrest that may result in the induction of apoptosis. The tumorigenic MCF-7 cells were especially sensitive to 2ME treatment compared to the normal MCF-12A cells. Therefore, differential mechanism(s) of growth inhibition are evident between the normal and tumorigenic cells.
Collapse
Affiliation(s)
- Catherina Van Zijl
- Department of Physiology, University of Pretoria, Pretoria, South Africa
| | | | | | | |
Collapse
|
44
|
Conejo-García A, Núñez MDC, Díaz-Gavilán M, Cruz-López O, Gallo MÁ, Espinosa A, Campos JM. From 5-fluorouracil acyclonucleosides to benzo-fused six- and seven-membered rings linked to pyrimidine and purine bases: the shift from differentiating anticancer agents to apoptotic inducers. Expert Opin Drug Discov 2008; 3:1223-35. [DOI: 10.1517/17460441.3.10.1223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Liu W, Li W, Fujita T, Yang Q, Wan Y. Proteolysis of CDH1 enhances susceptibility to UV radiation-induced apoptosis. Carcinogenesis 2008; 29:263-72. [PMID: 18174259 DOI: 10.1093/carcin/bgm251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
As a critical ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C) governs cell cycle progression, signaling modulation and the pathogenesis of some human diseases. Recent studies implicate APC in maintaining genomic integrity, but the mechanism by which it plays such a role remains largely unknown. We report here that acute UV radiation triggers proteolysis of CDH1, an activator of APC, which is involved in regulation of apoptosis induced by UV radiation. Depletion of CDH1 by RNA interference enhances the cellular susceptibility to apoptosis in response to UV radiation, whereas overexpression of non-degradable CDH1 delays UV radiation-induced apoptosis. In addition, UV-induced degradation of CDH1 results in the accumulation of cyclin B1 and therefore to increased CDK1 activity, which is believed to enhance UV-induced apoptosis. The present results unveil a novel role for the APC in UV-induced cell death and demonstrate a new regulatory mechanism for APC/CDH1 through proteolysis.
Collapse
Affiliation(s)
- Weijun Liu
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-1863, USA
| | | | | | | | | |
Collapse
|
46
|
Hattori T, Isobe T, Abe K, Kikuchi H, Kitagawa K, Oda T, Uchida C, Kitagawa M. Pirh2 promotes ubiquitin-dependent degradation of the cyclin-dependent kinase inhibitor p27Kip1. Cancer Res 2007; 67:10789-95. [PMID: 18006823 DOI: 10.1158/0008-5472.can-07-2033] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cyclin-dependent kinase inhibitor p27(Kip1) is degraded in late G(1) phase by the ubiquitin-proteasome pathway, allowing cells to enter S phase. Due to accelerated degradation of p27(Kip1), various human cancers express low levels of p27(Kip1) associated with poor prognosis. S-phase kinase-associated protein 2, the F-box protein component of an SCF ubiquitin ligase complex, is implicated in degradation of p27(Kip1) during S-G(2) phases. Recently, Kip1 ubiquitination-promoting complex has been reported as another ubiquitin ligase that targets cytoplasmic p27(Kip1) exported from the nucleus in G(0)-G(1) phases. Here, we identified a RING-H2-type ubiquitin ligase, Pirh2, as a p27(Kip1)-interacting protein. Endogenous Pirh2 physically interacted with endogenous p27(Kip1) in mammalian cells. Pirh2 directly ubiquitinated p27(Kip1) in an intact RING finger domain-dependent manner in vivo, as well as in vitro. Ablation of endogenous Pirh2 by small interfering RNA increased the steady-state level of p27(Kip1) and decelerated p27(Kip1) turnover. Depletion of Pirh2 induced accumulation of p27(Kip1) in both the nucleus and cytoplasm. Pirh2 expression was induced from late G(1)-S phase, whereas p27(Kip1) was decreased in synchronization with accumulation of Pirh2. Furthermore, reduction of Pirh2 resulted in an impairment of p27(Kip1) degradation and an inhibition of cell cycle progression at G(1)-S transition in a p53-independent manner. Overall, the results indicate that Pirh2 acts as a negative regulator of p27(Kip1) function by promoting ubiquitin-dependent proteasomal degradation.
Collapse
Affiliation(s)
- Takayuki Hattori
- Department of Biochemistry 1, Hamamatsu University School of Medicine, Higashi-ku, Hamamatsu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Murata H, Yagi T, Iwagaki H, Ogino T, Sadamori H, Matsukawa H, Umeda Y, Haga S, Takaka N, Ozaki M. Mechanism of impaired regeneration of fatty liver in mouse partial hepatectomy model. J Gastroenterol Hepatol 2007; 22:2173-80. [PMID: 18031377 DOI: 10.1111/j.1440-1746.2006.04798.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM The mechanism of injury in steatotic liver under pathological conditions been extensively examined. However, the mechanism of an impaired regeneration is still not well understood. The aim of this study was to analyze the mechanism of impaired regeneration of steatotic liver after partial hepatectomy (PH). METHODS db/db fatty mice and lean littermates were used for the experiments. Following 70% PH, the survival rate and recovery of liver mass were examined. Liver tissue was histologically examined and analyzed by western blotting and RT-PCR. RESULTS Of 35 db/db mice, 25 died within 48 h of PH, while all of the control mice survived. Liver regeneration of surviving db/db mice was largely impaired. In db/db mice, mitosis of hepatocytes after PH was disturbed, even though proliferating cell nuclear antigen (PCNA) expression (G1 to S phase marker) in hepatocytes was equally observed in both mice groups. Interestingly, phosphorylation of Cdc2 in db/db mice was suppressed by reduced expression of Wee1 and Myt1, which phosphorylate Cdc2 in S to G2 phase. CONCLUSIONS In steatotic liver, cell-cycle-related proliferative disorders occurred at mid-S phase after PCNA expression. Reduced expression of Wee1 and Myt1 kinases may therefore maintain Cdc2 in an unphosphorylated state and block cell cycle progression in mid-S phase. These kinases may be critical factors involved in the impaired liver regeneration in fatty liver.
Collapse
Affiliation(s)
- Hiroshi Murata
- Department of Gastroenterological Surgery, Transplant and Surgical Oncology, Okayama University Graduate School of Medicine and Dentistry, Shikata, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu ZG, Zhao LN, Liu YW, Li TT, Fan DM, Chen JJ. Activation of Cdc2 contributes to apoptosis in HPV E6 expressing human keratinocytes in response to therapeutic agents. J Mol Biol 2007; 374:334-45. [PMID: 17936297 PMCID: PMC2099577 DOI: 10.1016/j.jmb.2007.09.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2007] [Revised: 09/01/2007] [Accepted: 09/09/2007] [Indexed: 12/18/2022]
Abstract
Infection with human papillomaviruses (HPV) is strongly associated with the development of cervical cancer. The HPV E6 oncogene induces apoptosis in cervical cancer precursor lesions but the mechanism is poorly understood. While it is expected that inactivation of p53 by E6 should lead to a reduction in apoptosis, E6 also sensitizes cells to apoptosis under some experimental conditions. Here, we demonstrate that expression of E6 in human keratinocytes rendered sensitization to chemotherapeutic agents. The cell death was shown to be by apoptosis involving caspase activation and the mitochondria pathway. To explore mechanisms involved in sensitization of E6 expressing cells to apoptosis, we used a proteomic approach to identify proteins differentially expressed in E6 expressing and control keratinocytes. Among nearly a thousand proteins examined, Cdc2 was demonstrated to be the most dramatically up-regulated protein in E6 expressing cells. p53 degradation appears to be important for the up-regulation of Cdc2 by E6. Using genetic, pharmacologic, and siRNA strategies, a role for Cdc2 in E6 expression-conferred apoptosis was demonstrated. Thus, these results have important therapeutic implications in enhancing the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Zhi-Guo Liu
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605-2324, USA
- The State Key Laboratory of Cancer Biology, Institute of Gastroenterology, Xijing Hospital, the Fourth Military Medical University, Xi’an 710032, China
| | - Li-Na Zhao
- The State Key Laboratory of Cancer Biology, Institute of Gastroenterology, Xijing Hospital, the Fourth Military Medical University, Xi’an 710032, China
| | - Ying-Wang Liu
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605-2324, USA
| | - Ting-Ting Li
- The State Key Laboratory of Cancer Biology, Institute of Gastroenterology, Xijing Hospital, the Fourth Military Medical University, Xi’an 710032, China
| | - Dai-Ming Fan
- The State Key Laboratory of Cancer Biology, Institute of Gastroenterology, Xijing Hospital, the Fourth Military Medical University, Xi’an 710032, China
| | - Jason J Chen
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605-2324, USA
| |
Collapse
|
49
|
M. Noteborn MH, Koch G. Chicken anaemia virus infection: Molecular basis of pathogenicity. Avian Pathol 2007; 24:11-31. [DOI: 10.1080/03079459508419046] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Balakrishnan A, Wang L, Li X, Ohman-Strickland P, Malatesta P, Fan H. Inhibition of chlamydial infection in the genital tract of female mice by topical application of a peptide deformylase inhibitor. Microbiol Res 2007; 164:338-46. [PMID: 17936604 PMCID: PMC2735082 DOI: 10.1016/j.micres.2007.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Revised: 03/28/2007] [Accepted: 05/08/2007] [Indexed: 11/15/2022]
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium responsible for a number of health problems, including sexually transmitted infection in humans. We recently discovered that C. trachomatis infection in cell culture is highly susceptible to inhibitors of peptide deformylase, an enzyme that removes the N-formyl group from newly synthesized polypeptides. In this study, one of the deformylase inhibitors, GM6001, was tested for potential antichlamydial activity using a murine genital C. muridarum infection model. Topical application of GM6001 significantly reduced C. muridarum loading in BALB/c mice that were vaginally infected with the pathogen. In striking contrast, growth of the probiotic Lactobacillus plantarum is strongly resistant to the PDF inhibitor. GM6001 demonstrated no detectable toxicity against host cells. On the basis of these data and our previous observations, we conclude that further evaluation of PDF inhibitors for prevention and treatment of sexually transmitted chlamydial infection is warranted.
Collapse
Affiliation(s)
- Amit Balakrishnan
- University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
- University of Medicine and Dentistry of New Jersey Graduate School of Biomedical Sciences, Piscataway, NJ 08854, USA
- Rutgers the State University of New Jersey Graduate School, Piscataway, NJ 08854
| | - Lingling Wang
- University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Xiaojin Li
- University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Pamela Ohman-Strickland
- University of Medicine and Dentistry of New Jersey, School of Public Health, Piscataway, NJ 08854, USA
| | - Paul Malatesta
- University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Huizhou Fan
- University of Medicine and Dentistry of New Jersey Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| |
Collapse
|