1
|
Song M, Deng M, Peng Z, Dai F, Wang Y, Shu W, Zhou X, Zhang J, Hou Y, Yu B. Granulocyte colony-stimulating factor mediates bone loss via the activation of IL-1β/JNK signaling pathway in murine Staphylococcus aureus-induced osteomyelitis. Int Immunopharmacol 2024; 141:112959. [PMID: 39163688 DOI: 10.1016/j.intimp.2024.112959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024]
Abstract
Staphylococcus aureus (S. aureus)-induced bone loss is a significant challenge in the treatment of osteomyelitis. Our previous study was the first to confirm that granulocyte colony-stimulating factor (G-CSF) mediates S. aureus-induced bone loss. However, the underlying mechanism remains unknown. The objective of this study was to elucidate this. We found G-CSF mediated BMSC senescence and increased IL-1β concentration of serum and bone marrow in mice after S. aureus infection. Furthermore, we demonstrated that G-CSF promoted the expression of IL1b in murine bone marrow-derived neutrophils. Notably, we identified that IL-1β mediated BMSC (bone marrow mesenchymal stromal cell) senescence in mice after S. aureus infection. Importantly, IL-1β neutralizing antibody effectively alleviated BMSC senescence and bone loss caused by S. aureus infection in mice. In terms of molecular mechanism, we found IL-1β induced BMSC senescence by JNK/P53 and JNK/BCL2 pathways. Collectively, G-CSF promotes IL-1β production which induces BMSC senescence via JNK/P53 and JNK/BCL2 pathways, leading to S. aureus-induced bone loss. This study identified novel targets for preventing and treating S. aureus-induced bone loss in osteomyelitis.
Collapse
Affiliation(s)
- Mingrui Song
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingye Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziyue Peng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangfang Dai
- Huiqiao Medical Center, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yutian Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Shu
- Department of Trauma Orthopedics, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Xuyou Zhou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinye Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yilong Hou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Nagata Y, Tomimori J, Hagiwara T. Anti-apoptotic protein Bcl-2 contributes to the determination of reserve cells during myogenic differentiation of C2C12 cells. In Vitro Cell Dev Biol Anim 2024; 60:760-770. [PMID: 38619740 DOI: 10.1007/s11626-024-00905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 03/28/2024] [Indexed: 04/16/2024]
Abstract
Skeletal muscle's regenerative ability is vital for maintaining muscle function, but chronic diseases like Duchenne muscular dystrophy can deplete this capacity. Muscle satellite cells, quiescent in normal situations, are activated during muscle injury, expressing myogenic regulatory factors, and producing myogenic progenitor cells. It was reported that muscle stem cells in primary culture and reserve cells in C2C12 cells express anti-apoptotic protein Bcl-2. Although the role of Bcl-2 expressed in myogenic cells has been thought to be to enhance cell viability, we hypothesized that Bcl-2 may promote the formation of reserve cells. The expression pattern analysis showed the expression of Bcl-2 in undifferentiated mononucleated cells, emphasizing its usefulness as a reserve cell marker and reminding us that cells expressing Bcl-2 have low proliferative potential. Silencing of Bcl-2 by transfection with siRNA decreased cell viability and the number of reserve cells, while overexpression of Bcl-2 not only increases cell viability but also inhibits muscle differentiation and proliferation. These results emphasize dual roles of Bcl-2 in protecting cells from apoptosis and contributing to reserve cell formation by regulating myoblast proliferation and/or differentiation. Overall, the study sheds light on the multifaceted role of Bcl-2 in the maintenance of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Yosuke Nagata
- Department of Bioscience, Faculty of Life Science, Okayama University of Science, 1-1 Ridaicho, Kita-Ku, Okayama-Shi, 700-0005, Japan.
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan.
- Department of Natural Science, Graduate School of Science and Engineering, Okayama University of Science, Okayama, Japan.
| | - Jun Tomimori
- Department of Life Science, Faculty of Science, Okayama University of Science, Okayama, Japan
| | - Tomoharu Hagiwara
- Department of Natural Science, Graduate School of Science and Engineering, Okayama University of Science, Okayama, Japan
| |
Collapse
|
3
|
Zhang L, Xia J. N6-Methyladenosine Methylation of mRNA in Cell Apoptosis. Mol Neurobiol 2024; 61:3934-3948. [PMID: 38040996 DOI: 10.1007/s12035-023-03813-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
Apoptosis, a highly controlled homeostatic mechanism that eliminates single cells without destroying tissue function, occurs during growing development and senescence. N6-methyladenosine (m6A), as the most common internal modification of eukaryotic mRNA, fine-tunes gene expression by regulating many aspects of mRNA metabolism, such as splicing, nucleation, stability, translation, and degradation. Remarkably, recent reports have indicated that aberrant methylation of m6A-related RNA may directly or indirectly influence the expression of apoptosis-related genes, thus regulating the process of cell apoptosis. In this review, we summarized the relationship between m6A modification and cell apoptosis, especially its role in the nervous system, and analyzed the limitations of the current research.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Cardona-Mendoza A, Fonseca-Benitez A, Buitrago DM, Coy-Barrera E, Perdomo SJ. Down-regulation of human papillomavirus E6 oncogene and antiproliferative effect of Schisandra chinensis and Pueraria lobata natural extracts on Hela cell line. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117225. [PMID: 37797877 DOI: 10.1016/j.jep.2023.117225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cervical cancer is one of the most common malignancies in women that continues to be a public health problem worldwide. Human papillomavirus (HPV) infection is closely related as the causative agent of almost all cases of cervical cancer. Currently, there is no effective treatment for the persistence of HPV. Although vaccines have shown promising results in recent years, they are still a costly strategy for developing countries and have no therapeutic effect on existing infections, which is why the need arises to search for new strategies that can be used in treatment, suppressing oncogenic HPV and disease progression. Extracts of Schisandra Chinensis and Pueraria lobata have been used in traditional medicine, and it has been shown in recent years that some of their bioactive compounds have pharmacological, antioxidant, antitumor, apoptotic, and proliferation effects in HPV-positive cells. However, its mechanism of action has yet to be fully explored. AIM OF THE STUDY The following study aimed to determine the chemical composition, antioxidant activity, and potential antiproliferative and viral oncogene effects of natural extracts of S. chinensis and P. lobata on HPV-18 positive cervical cancer cells. MATERIALS AND METHODS The HPV-18-positive HeLa cells were treated for 24 and 48 h with the ethanolic extracts of S chinensis and P. lobata. Subsequently, cell viability was evaluated using the resazurin method, the effect on the cell cycle of the extracts (1.0, 10, and 100 μg/mL) was measured by flow cytometry, the gene of expression of the E6/E7, P53, BCL-2, and E2F-1 were determined by RT-PCR and the protein expression of p53, Ki-67, x|and Bcl-2 by immunohistochemistry. Additionally, the chemical characterization of the two extracts was carried out using LC-MS, and the total phenolics content (TPC), Total flavonoid content (TFC), and DPPH radical scavenging capacity were determined. Data were analyzed using the Mann-Whitney and Kruskal Wallis U test with GraphPad Prism 6 software. RESULTS The natural extracts of Schisandra chinensis and Pueraria lobata induced down-regulation of E6 HPV oncogene (p<0.05) and a strong up-regulation of P53 (p<0.05), E2F-1 (p<0.05), and Bcl-2 (p<0.05) gene expression. Simultaneously, the natural extracts tend to increase the p53 protein levels and arrest the cell cycle of HeLa in the G1/S phase (p<0.05). Investigated extracts were characterized by the occurrence of bioactive lignans and isoflavones in S. chinensis and P. lobata, respectively. CONCLUSION The extracts of S. chinensis and P. lobata within their chemical characterization mainly present lignan and isoflavone-type compounds, which are probably responsible for inhibiting the expression of the HPV E6 oncogene and inducing an increase in the expression of p53, Bcl -2 and E2F-1 producing cell cycle detection in S phase in HeLa cells. Therefore, these extracts are good candidates to continue studying their antiviral and antiproliferative potential in cells transformed by HPV.
Collapse
Affiliation(s)
- Andrés Cardona-Mendoza
- Cellular and Molecular Immunology Group-INMUBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Angela Fonseca-Benitez
- Cellular and Molecular Immunology Group-INMUBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Diana Marcela Buitrago
- Cellular and Molecular Immunology Group-INMUBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia; Unidad de Investigación Básica Oral-UIBO, Facultad de Odontología, Universidad El Bosque, Bogotá, Colombia
| | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Department of Chemistry, Universidad Militar Nueva Granada, Cajicá, 250247, Colombia
| | - Sandra J Perdomo
- Cellular and Molecular Immunology Group-INMUBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia.
| |
Collapse
|
5
|
Wiecek AJ, Cutty SJ, Kornai D, Parreno-Centeno M, Gourmet LE, Tagliazucchi GM, Jacobson DH, Zhang P, Xiong L, Bond GL, Barr AR, Secrier M. Genomic hallmarks and therapeutic implications of G0 cell cycle arrest in cancer. Genome Biol 2023; 24:128. [PMID: 37221612 PMCID: PMC10204193 DOI: 10.1186/s13059-023-02963-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 05/07/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Therapy resistance in cancer is often driven by a subpopulation of cells that are temporarily arrested in a non-proliferative G0 state, which is difficult to capture and whose mutational drivers remain largely unknown. RESULTS We develop methodology to robustly identify this state from transcriptomic signals and characterise its prevalence and genomic constraints in solid primary tumours. We show that G0 arrest preferentially emerges in the context of more stable, less mutated genomes which maintain TP53 integrity and lack the hallmarks of DNA damage repair deficiency, while presenting increased APOBEC mutagenesis. We employ machine learning to uncover novel genomic dependencies of this process and validate the role of the centrosomal gene CEP89 as a modulator of proliferation and G0 arrest capacity. Lastly, we demonstrate that G0 arrest underlies unfavourable responses to various therapies exploiting cell cycle, kinase signalling and epigenetic mechanisms in single-cell data. CONCLUSIONS We propose a G0 arrest transcriptional signature that is linked with therapeutic resistance and can be used to further study and clinically track this state.
Collapse
Affiliation(s)
- Anna J. Wiecek
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Stephen J. Cutty
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Daniel Kornai
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Mario Parreno-Centeno
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Lucie E. Gourmet
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | | | - Daniel H. Jacobson
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
- UCL Cancer Institute, Paul O’Gorman Building, University College London, London, UK
| | - Ping Zhang
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lingyun Xiong
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gareth L. Bond
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alexis R. Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Cell Cycle Control Team, MRC London Institute of Medical Sciences (LMS), London, UK
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
6
|
Barriuso D, Alvarez-Frutos L, Gonzalez-Gutierrez L, Motiño O, Kroemer G, Palacios-Ramirez R, Senovilla L. Involvement of Bcl-2 Family Proteins in Tetraploidization-Related Senescence. Int J Mol Sci 2023; 24:ijms24076374. [PMID: 37047342 PMCID: PMC10094710 DOI: 10.3390/ijms24076374] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
The B-cell lymphoma 2 (Bcl-2) family of proteins is the main regulator of apoptosis. However, multiple emerging evidence has revealed that Bcl-2 family proteins are also involved in cellular senescence. On the one hand, the different expression of these proteins determines the entry into senescence. On the other hand, entry into senescence modulates the expression of these proteins, generally conferring resistance to apoptosis. With some exceptions, senescent cells are characterized by the upregulation of antiapoptotic proteins and downregulation of proapoptotic proteins. Under physiological conditions, freshly formed tetraploid cells die by apoptosis due to the tetraploidy checkpoint. However, suppression of Bcl-2 associated x protein (Bax), as well as overexpression of Bcl-2, favors the appearance and survival of tetraploid cells. Furthermore, it is noteworthy that our laboratory has shown that the joint absence of Bax and Bcl-2 antagonist/killer (Bak) favors the entry into senescence of tetraploid cells. Certain microtubule inhibitory chemotherapies, such as taxanes and vinca alkaloids, induce the generation of tetraploid cells. Moreover, the combined use of inhibitors of antiapoptotic proteins of the Bcl-2 family with microtubule inhibitors increases their efficacy. In this review, we aim to shed light on the involvement of the Bcl-2 family of proteins in the senescence program activated after tetraploidization and the possibility of using this knowledge to create a new therapeutic strategy targeting cancer cells.
Collapse
|
7
|
Zhang Y, Dong Y, Wei Q, Zhuang Z, Liu Y, Yuan Q, He W, Jing Z, Li J, Li P, Zhang L, Hong Z, Zhang N, Wang H, Li W. miR-126 mitigates the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by targeting the ERK1/2 and Bcl-2 pathways. Acta Biochim Biophys Sin (Shanghai) 2023; 55:449-459. [PMID: 36942990 PMCID: PMC10160225 DOI: 10.3724/abbs.2023016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (hBMMSCs) are a promising cell source for bone engineering owing to their high potential to differentiate into osteoblasts. The objective of the present study is to assess microRNA-126 (miR-126) and examine its effects on the osteogenic differentiation of hBMMSCs. In this study, we investigate the role of miR-126 in the progression of osteogenic differentiation (OD) as well as the apoptosis and inflammation of hBMMSCs during OD induction. OD is induced in hBMMSCs, and matrix mineralization along with other OD-associated markers are evaluated by Alizarin Red S (AR) staining and quantitative PCR (qPCR). Gain- and loss-of-function studies are performed to demonstrate the role of miR-126 in the OD of hBMMSCs. Flow cytometry and qPCR-based cytokine expression studies are performed to investigate the effect of miR-126 on the apoptosis and inflammation of hBMMSCs. The results indicate that miR-126 expression is downregulated during the OD of hBMMSCs. Gain- and loss-of function assays reveal that miR-126 upregulation inhibits the differentiation of hBMMSCs into osteoblasts, whereas the downregulation of miR-126 promotes hBMMSC differentiation, as assessed by the determination of osteogenic genes and alkaline phosphatase activity. Furthermore, the miR-126 level is positively correlated with the production of inflammatory cytokines and apoptotic cell death. Additionally, our results suggest that miR-126 negatively regulates not only B-cell lymphoma 2 (Bcl-2) expression but also the phosphorylation of extracellular signal‑regulated protein kinase (ERK) 1/2. Moreover, restoring ERK1/2 activity and upregulating Bcl-2 expression counteract the miR-126-mediated suppression of OD in hBMMSCs by promoting inflammation and apoptosis, respectively. Overall, our findings suggest a novel molecular mechanism relevant to the differentiation of hBMMSCs into osteoblasts, which can potentially facilitate bone formation by counteracting miR-126-mediated suppression of ERK1/2 activity and Bcl-2 expression.
Collapse
Affiliation(s)
- Ying Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yiping Dong
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qiushi Wei
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Zhikun Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Youwen Liu
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Qiang Yuan
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wei He
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Zhenhao Jing
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jitian Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Peifeng Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Leilei Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Zhinan Hong
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Ning Zhang
- Hunan University of Chinese Medicine, Changsha 410208, China
| | - Haibin Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wuyin Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| |
Collapse
|
8
|
At the Crossroads of Life and Death: The Proteins That Influence Cell Fate Decisions. Cancers (Basel) 2022; 14:cancers14112745. [PMID: 35681725 PMCID: PMC9179324 DOI: 10.3390/cancers14112745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cellular senescence and apoptosis were historically thought of as two distinct cell fate pathways. However, many of the proteins involved are integral to both pathways. In particular, the ability of p53 to regulate both senescence and apoptosis meant it was seen as the decisive factor in these decisions, yet questions remain about its ability to select on its own the most appropriate cell fate according to each situation. Therefore, cell fates are no longer considered fixed endpoints but dynamic states that can be shifted given the right combination of activation and/or inhibitions of cofactors. Abstract When a cell is damaged, it must decide how to respond. As a consequence of a variety of stresses, cells can induce well-regulated programmes such as senescence, a persistent proliferative arrest that limits their replication. Alternatively, regulated programmed cell death can be induced to remove the irreversibly damaged cells in a controlled manner. These programmes are mainly triggered and controlled by the tumour suppressor protein p53 and its complex network of effectors, but how it decides between these wildly different responses is not fully understood. This review focuses on the key proteins involved both in the regulation and induction of apoptosis and senescence to examine the key events that determine cell fate following damage. Furthermore, we examine how the regulation and activity of these proteins are altered during the progression of many chronic diseases, including cancer.
Collapse
|
9
|
DU X, Xiao J, Fu X, Xu B, Han H, Wang Y, Pei X. A proteomic analysis of Bcl-2 regulation of cell cycle arrest: insight into the mechanisms. J Zhejiang Univ Sci B 2021; 22:839-855. [PMID: 34636187 PMCID: PMC8505462 DOI: 10.1631/jzus.b2000802] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/11/2021] [Indexed: 01/01/2023]
Abstract
B cell lymphoma 2 (Bcl-2) is an important antiapoptotic gene that plays a dual role in the maintenance of the dynamic balance between the survival and death of cancer cells. In our previous study, Bcl-2 was shown to delay the G0/G1 to S phase entry by regulating the mitochondrial metabolic pathways to produce lower levels of adenosine triphosphate (ATP) and reactive oxygen species (ROS). However, the detailed molecular mechanisms or pathways by which Bcl-2 regulates the cell cycle remain unknown. Here, we compared the effects of Bcl-2 overexpression with an empty vector control in the NIH3T3 cell line synchronized by serum starvation, and evaluated the effects using proteomic analysis. The effect of Bcl-2 on cell cycle regulation was detected by monitoring Bcl-2 and p27 expression. The result of subsequent proteomic analysis of Bcl-2 overexpressing cells identified 169 upregulated and 120 downregulated proteins with a 1.5-fold change. These differentially expressed proteins were enriched in a number of signaling pathways predominantly involving the ribosome and oxidative phosphorylation, according to the data of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. These results indicated that Bcl-2 potentially acts at the translation level to influence proteins or enzymes of the respiratory chain or in the ribosome, and thereby regulates the cell cycle. Additionally, differentially expressed proteins involved in oxidative phosphorylation were determined to account for most of the effects of Bcl-2 on the cell cycle mediated by the mitochondrial pathway investigated in our previous study. These results can provide assistance for additional in-depth studies on the regulation of the cell cycle by Bcl-2. The results of the proteomic analysis determined the mechanism of Bcl-2-dependent delay of the cell cycle progression. In summary, the results of this study provide a novel mechanistic basis for identifying the key proteins or pathways for designing and developing precisely targeted cancer drugs.
Collapse
Affiliation(s)
- Xing DU
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China
| | - Jingjing Xiao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan 750004, China
| | - Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
- Department of Biochemistry and Molecular Biology, Ningxia Medical University, Yinchuan 750004, China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Hang Han
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Yin Wang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China.
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan 750004, China.
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
10
|
Stress Relief Techniques: p38 MAPK Determines the Balance of Cell Cycle and Apoptosis Pathways. Biomolecules 2021; 11:biom11101444. [PMID: 34680077 PMCID: PMC8533283 DOI: 10.3390/biom11101444] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/23/2021] [Accepted: 09/30/2021] [Indexed: 12/18/2022] Open
Abstract
Protein signaling networks are formed from diverse and inter-connected cell signaling pathways converging into webs of function and regulation. These signaling pathways both receive and conduct molecular messages, often by a series of post-translation modifications such as phosphorylation or through protein-protein interactions via intrinsic motifs. The mitogen activated protein kinases (MAPKs) are components of kinase cascades that transmit signals through phosphorylation. There are several MAPK subfamilies, and one subfamily is the stress-activated protein kinases, which in mammals is the p38 family. The p38 enzymes mediate a variety of cellular outcomes including DNA repair, cell survival/cell fate decisions, and cell cycle arrest. The cell cycle is itself a signaling system that precisely controls DNA replication, chromosome segregation, and cellular division. Another indispensable cell function influenced by the p38 stress response is programmed cell death (apoptosis). As the regulators of cell survival, the BCL2 family of proteins and their dynamics are exquisitely sensitive to cell stress. The BCL2 family forms a protein-protein interaction network divided into anti-apoptotic and pro-apoptotic members, and the balance of binding between these two sides determines cell survival. Here, we discuss the intersections among the p38 MAPK, cell cycle, and apoptosis signaling pathways.
Collapse
|
11
|
Basu A. The interplay between apoptosis and cellular senescence: Bcl-2 family proteins as targets for cancer therapy. Pharmacol Ther 2021; 230:107943. [PMID: 34182005 DOI: 10.1016/j.pharmthera.2021.107943] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
Cell death by apoptosis and permanent cell cycle arrest by senescence serve as barriers to the development of cancer. Chemotherapeutic agents not only induce apoptosis, they can also induce senescence known as therapy-induced senescence (TIS). There are, however, controversies whether TIS improves or worsens therapeutic outcome. Unlike apoptosis, which permanently removes cancer cells, senescent cells are metabolically active, and can contribute to tumor progression and relapse. If senescent cells are not cleared by the immune system or if cancer cells escape senescence, they may acquire resistance to apoptotic stimuli and become highly aggressive. Thus, there have been significant efforts in developing senolytics, drugs that target these pro-survival molecules to eliminate senescent cells. The anti-apoptotic Bcl-2 family proteins not only protect against cell death by apoptosis, but they also allow senescent cells to survive. While combining senolytics with chemotherapeutic drugs is an attractive approach, there are also limitations. Moreover, members of the Bcl-2 family have distinct effects on apoptosis and senescence. The purpose of this review article is to discuss recent literatures on how members of the Bcl-2 family orchestrate the interplay between apoptosis and senescence, and the challenges and progress in targeting these Bcl-2 family proteins for cancer therapy.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
12
|
Taherzadeh-Soureshjani P, Chehelgerdi M. Algae-meditated route to cuprous oxide (Cu2O) nanoparticle: differential expression profile of MALAT1 and GAS5 LncRNAs and cytotoxic effect in human breast cancer. Cancer Nanotechnol 2020. [DOI: 10.1186/s12645-020-00066-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Abstract
Background
Breast cancer (BC), as the most widely recognized disease in women worldwide, represents about 30% of all cancers impacting women. This study was aimed to synthesize Cu2O nanoparticles from the cystoseira myrica algae (CM-Cu2O NPs) assess their antimicrobial activity against pathogenic bacteria and fungi. We evaluated the expression levels of lncRNAs (MALAT1 and GAS5) and apoptosis genes (p53, p27, bax, bcl2 and caspase3), their prognostic roles.
Methods
In this study, CM-Cu2O NPs synthesized by cystoseira myrica algae extraction used to evaluate its cytotoxicity and apoptotic properties on MDA-MB-231, SKBR3 and T-47D BC cell lines compared to HDF control cell line. The CM-Cu2O NPs was characterized by UV–Vis spectroscopy, Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), Transmission electron microscopy (TEM) and Scanning electron microscopy (SEM). The antimicrobial activity of CM-Cu2O NPs was assessed against pathogenic bacteria, staphylococcus aureus (S. aureus) PTCC 1112 bacteria as a standard gram-positive bacteria and pseudomonas aeruginosa (P. aeruginosa) PTCC 1310 as a standard gram-negative bacterium. Expression profile of MALAT1 and GAS5 lncRNAs and apoptosis genes, i.e., p27, bax, bcl2 and caspase3 genes, were calculated utilizing qRT-PCR. The changes in the expression levels were determined using the DDCT method.
Results
MALAT1 was upregulated in MDA-MB-231, SKBR3 and T-47D BC (p < 0.01), while GAS5 was downregulated in SKBR3 and T-47D cell lines tested compared with HDF control cell line (p < 0.05) was found. The results revealed that, p27, bax and caspase3 were significantly upregulated in BC cell lines as compared with normal cell line. Bcl2 expression was also significantly increased in MDA-MB-231 and T47D cell lines compared with normal cell line, but bcl2 levels were downregulated in SKBR3 cell line.
Conclusions
Our results confirm the beneficial cytotoxic effects of green-synthesized CM-Cu2O NPs on BC cell lines. This nanoparticle decreased angiogenesis and induces apoptosis, so we conclude that CM-Cu2O NPs can be used as a supplemental drug in cancer treatments. Significantly, elevated circulating lncRNAs were demonstrated to be BC specific and could differentiate BC cell lines from the normal cell lines. It was demonstrated that lncRNAs used in this study and their expression profiles can be created as biomarkers for early diagnosis and prognosis of BC. Further studies utilizing patients would give recognizable identification of lncRNAs as key players in intercellular interactions.
Collapse
|
13
|
Whitaker RH, Placzek WJ. MCL1 binding to the reverse BH3 motif of P18INK4C couples cell survival to cell proliferation. Cell Death Dis 2020; 11:156. [PMID: 32111816 PMCID: PMC7048787 DOI: 10.1038/s41419-020-2351-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Commitment to cell cycle entry and cellular duplication is a tightly coordinated and regulated process. Once initiated, a series of multiple checkpoints ensure both accurate genomic replication and chromosomal separation. In the event of unsuccessful cell division, parallel pathways exist that induce the cell to undergo programmed cell death, or apoptosis. At the center of such stress-induced, intrinsic apoptotic regulation lies the BCL2 family of pro- and anti-apoptotic regulatory proteins. In a proliferative state the balance of pro- and anti-apoptotic signaling proteins would be expected to favor an excess population of anti-apoptotic members. While the anti-apoptotic BCL2 family member, MCL1, has been identified to oversee mitotic progression, direct communication between the BCL2 family and cell proliferation has not been observed. In this study, we demonstrate a direct protein–protein interaction between MCL1 and the G1/S checkpoint protein, P18INK4C. This interaction is mediated by a reverse BH3 (rBH3) motif located in P18INK4C’s C-terminal ankyrin repeat. MCL1 is further shown to decrease P18INK4C expression and thereby regulate cell cycle entry in a retinoblastoma (RB1)-dependent manner. Our findings establish a mechanism for translation independent and direct communication between the BCL2 family regulation of apoptosis and CDK4/6-RB regulation of early G1/S transition during cellular division/growth.
Collapse
Affiliation(s)
- Robert H Whitaker
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Placzek
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
14
|
Glab JA, Cao Z, Puthalakath H. Bcl-2 family proteins, beyond the veil. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:1-22. [PMID: 32247577 DOI: 10.1016/bs.ircmb.2019.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis is an important part of both health and disease and is often regulated by the BCL-2 family of proteins. These proteins are either pro- or anti-apoptotic, existing in a delicate balance during homeostasis. They are best known for their role in regulating the activation of caspases and the execution of a cell in response to a variety of stimuli. However, it is often forgotten that these BCL-2 family proteins also have important roles to play in cell maintenance that are not associated with apoptosis. These include roles in regulating processes such as cell cycle progression, mitochondrial function, autophagy, intracellular calcium concentration, glucose and lipid metabolism, and the unfolded protein response. In addition to these established alternate functions, further discoveries are being made that have potential therapeutic benefits in diseases such as cancer. BOK, a BCL-2 family protein thought comparable to multidomain pro-apoptotic proteins BAX and BAK, has recently been identified as a key player in metabolism of and resistance to the commonly used chemotherapeutic 5-FU. As a result of such findings, which could see the potential use of BOK as a biomarker for 5-FU sensitivity or mimetic molecules as a resensitization strategy, new targets and mechanisms of pathology may arise from further investigation into the realm of alternate functions of BCL-2 family proteins.
Collapse
Affiliation(s)
- Jason Andrew Glab
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC, Australia
| | - Zhipeng Cao
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC, Australia
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
15
|
Jibrim RLM, de Carvalho CV, Invitti AL, Schor E. Expression of the TFDP1 gene in the endometrium of women with deep infiltrating endometriosis. Gynecol Endocrinol 2019; 35:490-493. [PMID: 30638096 DOI: 10.1080/09513590.2018.1540569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The field of endometriosis etiopathogenesis aims to identify the origin of disease in endometrial disorders. Changes in gene and protein expression related to cell adhesion, collagenases, and, mainly, cell cycle regulators have been identified. We set out to analyze the expression of the transcription factor DP-1 (TFDP1) gene, which encodes a protein that controls the G1/S phase passage of the cell cycle, in the endometrium of women with deep infiltrating endometriosis (DIE). Samples of endometrium from both endometriosis-affected women and healthy women were collected, cultured and maintained at the Cell Bank of the Pelvic Pain and Endometriosis Unit of the Federal University of Sao Paulo. This study analyzed five samples from the endometrium cell culture of healthy patients (i.e. no pelvic disease, as determined by means of laparoscopic tubal ligation) and six samples from women diagnosed with DIE. Samples were evaluated for TFDP1 gene expression by real-time PCR. We observed a downregulation of TFDP1 in the endometrium cells of women with DIE when compared to the control (a fold-change of -2.05, p value=.011). The TFDP1 gene is part of the cell cycle pathway, but its function is not yet clear. Additional studies are necessary to clarify the function of TFDP1 in endometriosis etiopathogenesis.
Collapse
Affiliation(s)
- Rodrigo Lopes Meime Jibrim
- a Gynecology Department, Pelvic Pain and Endometriosis Unit , Universidade Federal de São Paulo - Escola Paulista de Medicina (UNIFESP-EPM) , Sao Paulo , Brazil
| | - Cristina Valletta de Carvalho
- a Gynecology Department, Pelvic Pain and Endometriosis Unit , Universidade Federal de São Paulo - Escola Paulista de Medicina (UNIFESP-EPM) , Sao Paulo , Brazil
| | - Adriana Luckow Invitti
- a Gynecology Department, Pelvic Pain and Endometriosis Unit , Universidade Federal de São Paulo - Escola Paulista de Medicina (UNIFESP-EPM) , Sao Paulo , Brazil
| | - Eduardo Schor
- a Gynecology Department, Pelvic Pain and Endometriosis Unit , Universidade Federal de São Paulo - Escola Paulista de Medicina (UNIFESP-EPM) , Sao Paulo , Brazil
| |
Collapse
|
16
|
Farahani R, Rezaei-Lotfi S, Simonian M, Hunter N. Bi-modal reprogramming of cell cycle by MiRNA-4673 amplifies human neurogenic capacity. Cell Cycle 2019; 18:848-868. [PMID: 30907228 DOI: 10.1080/15384101.2019.1595873] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Molecular mechanisms that inform heterochronic adaptations of neurogenesis in Homo sapiens remain largely unknown. Here, we uncover a signature in the cell cycle that amplifies the proliferative capacity of human neural progenitors by input from microRNA4673 encoded in Notch-1. The miRNA instructs bimodal reprogramming of the cell cycle, leading to initial synchronization of neural precursors at the G0 phase of the cell cycle followed by accelerated progression through interphase. The key event in G0 synchronization is transient inhibition by miR4673 of cyclin-dependent kinase-18, a member of an ancient family of cyclins that license M-G1 transition. In parallel, autophagic degradation of p53/p21 and transcriptional silencing of XRCC3/BRCA2 relax G1/S cell cycle checkpoint and accelerate interphase by ≈2.8-fold. The resultant reprogrammed cell cycle amplifies the proliferative capacity and delays the differentiation of human neural progenitors.
Collapse
Affiliation(s)
- Ramin Farahani
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia.,b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| | - Saba Rezaei-Lotfi
- b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| | - Mary Simonian
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia
| | - Neil Hunter
- a IDR/Westmead Institute for Medical Research , Sydney , NSW , Australia.,b Department of Life Sciences, Faculty of Medicine and Health Sciences , University of Sydney , Sydney , NSW , Australia
| |
Collapse
|
17
|
Haschka M, Karbon G, Fava LL, Villunger A. Perturbing mitosis for anti-cancer therapy: is cell death the only answer? EMBO Rep 2018; 19:e45440. [PMID: 29459486 PMCID: PMC5836099 DOI: 10.15252/embr.201745440] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 12/15/2017] [Accepted: 01/29/2018] [Indexed: 12/12/2022] Open
Abstract
Interfering with mitosis for cancer treatment is an old concept that has proven highly successful in the clinics. Microtubule poisons are used to treat patients with different types of blood or solid cancer since more than 20 years, but how these drugs achieve clinical response is still unclear. Arresting cells in mitosis can promote their demise, at least in a petri dish. Yet, at the molecular level, this type of cell death is poorly defined and cancer cells often find ways to escape. The signaling pathways activated can lead to mitotic slippage, cell death, or senescence. Therefore, any attempt to unravel the mechanistic action of microtubule poisons will have to investigate aspects of cell cycle control, cell death initiation in mitosis and after slippage, at single-cell resolution. Here, we discuss possible mechanisms and signaling pathways controlling cell death in mitosis or after escape from mitotic arrest, as well as secondary consequences of mitotic errors, particularly sterile inflammation, and finally address the question how clinical efficacy of anti-mitotic drugs may come about and could be improved.
Collapse
Affiliation(s)
- Manuel Haschka
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerlinde Karbon
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Luca L Fava
- Centre for Integrative Biology (CIBIO), University of Trento, Povo, Italy
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
18
|
De Amicis F, Guido C, Santoro M, Giordano F, Donà A, Rizza P, Pellegrino M, Perrotta I, Bonofiglio D, Sisci D, Panno ML, Tramontano D, Aquila S, Andò S. Ligand activated progesterone receptor B drives autophagy-senescence transition through a Beclin-1/Bcl-2 dependent mechanism in human breast cancer cells. Oncotarget 2018; 7:57955-57969. [PMID: 27462784 PMCID: PMC5295403 DOI: 10.18632/oncotarget.10799] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/09/2016] [Indexed: 12/21/2022] Open
Abstract
Loss of progesterone-receptors (PR) expression is associated with breast cancer progression. Herein we provide evidence that OHPg/PR-B through Beclin-1 evoke autophagy-senescence transition, in breast cancer cells. Specifically, OHPg increases Beclin-1 expression through a transcriptional mechanism due to the occupancy of Beclin-1 promoter by PR-B, together with the transcriptional coactivator SRC-2. This complex binds at a canonical half progesterone responsive element, which is fundamental for OHPg effects, as shown by site-directed mutagenesis. Beside, OHPg via non-genomic action rapidly activates JNK, which phosphorylates Bcl-2, producing the functional release from Beclin-1 interaction. This is not linked to an efficient autophagic flux, since p62 levels, marker of degradation via lysosomes, were not reduced after sustained OHPg stimulus. Instead, the cell cycle inhibitor p27 was induced, together with an irreversible G1 arrest, hallmark of cellular senescence. Specifically the increase of senescence-associated β-galactosidase activity was blocked by Bcl-2 siRNA but also by Beclin-1 siRNA. Collectively these findings support the importance of PR-B expression in breast cancer cells, thus targeting PR-B may be a useful strategy to provide additional approaches to existing therapies for breast cancer patients.
Collapse
Affiliation(s)
- Francesca De Amicis
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Carmela Guido
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Marta Santoro
- Centro Sanitario, University of Calabria, Rende, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Ada Donà
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Pietro Rizza
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | | | - Daniela Bonofiglio
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Diego Sisci
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Donatella Tramontano
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Saveria Aquila
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| | - Sebastiano Andò
- Centro Sanitario, University of Calabria, Rende, Italy.,Department of Pharmacy, Health Science and Nutrition, University of Calabria, Rende, Italy
| |
Collapse
|
19
|
Povea-Cabello S, Oropesa-Ávila M, de la Cruz-Ojeda P, Villanueva-Paz M, de la Mata M, Suárez-Rivero JM, Álvarez-Córdoba M, Villalón-García I, Cotán D, Ybot-González P, Sánchez-Alcázar JA. Dynamic Reorganization of the Cytoskeleton during Apoptosis: The Two Coffins Hypothesis. Int J Mol Sci 2017; 18:ijms18112393. [PMID: 29137119 PMCID: PMC5713361 DOI: 10.3390/ijms18112393] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/09/2017] [Accepted: 11/09/2017] [Indexed: 01/05/2023] Open
Abstract
During apoptosis, cells undergo characteristic morphological changes in which the cytoskeleton plays an active role. The cytoskeleton rearrangements have been mainly attributed to actinomyosin ring contraction, while microtubule and intermediate filaments are depolymerized at early stages of apoptosis. However, recent results have shown that microtubules are reorganized during the execution phase of apoptosis forming an apoptotic microtubule network (AMN). Evidence suggests that AMN is required to maintain plasma membrane integrity and cell morphology during the execution phase of apoptosis. The new “two coffins” hypothesis proposes that both AMN and apoptotic cells can adopt two morphological patterns, round or irregular, which result from different cytoskeleton kinetic reorganization during the execution phase of apoptosis induced by genotoxic agents. In addition, round and irregular-shaped apoptosis showed different biological properties with respect to AMN maintenance, plasma membrane integrity and phagocyte responses. These findings suggest that knowing the type of apoptosis may be important to predict how fast apoptotic cells undergo secondary necrosis and the subsequent immune response. From a pathological point of view, round-shaped apoptosis can be seen as a physiological and controlled type of apoptosis, while irregular-shaped apoptosis can be considered as a pathological type of cell death closer to necrosis.
Collapse
Affiliation(s)
- Suleva Povea-Cabello
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Manuel Oropesa-Ávila
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Patricia de la Cruz-Ojeda
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Marina Villanueva-Paz
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Mario de la Mata
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Juan Miguel Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Irene Villalón-García
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - David Cotán
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| | - Patricia Ybot-González
- Grupo de Neurodesarrollo, Unidad de Gestión de Pediatría, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain.
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD), and Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Consejo Superior de Investigaciones Científicas, Universidad Pablo de, Carretera de Utrera Km 1, 41013 Sevilla, Spain.
| |
Collapse
|
20
|
Tumor cell senescence response produces aggressive variants. Cell Death Discov 2017; 3:17049. [PMID: 28845296 PMCID: PMC5563524 DOI: 10.1038/cddiscovery.2017.49] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 06/09/2017] [Indexed: 01/17/2023] Open
Abstract
Tumors often respond favorably to initial chemotherapy but eventually relapse with drug resistance and increased metastatic potential. Cellular senescence is a major therapeutic outcome of cancer chemotherapy, which leads to tumor stasis or regression through immune clearance of senescent cells. However, senescent tumor cells have been shown to resume proliferation at low frequency. We found that subjecting arrested senescent tumor cells to cytotoxic treatments stimulates the clonogenic proliferation of remaining survivors. The senescence revertants showed a reduced rate of proliferation but increased migration and invasion potential in vitro, and increased tumorigenic potential in vivo. Gene expression profiling showed that the senescence revertants are distinct from both parental and senescent cells. A subset of senescence-activated genes remains active in the revertants. These genes are implicated in regulating cell motility, invasion, and metastasis, which may collectively contribute to the aggressiveness of the revertants. The findings suggest that although therapy-induced senescence has short-term benefits, the response also causes reprogramming of gene expression and activates invasion-related genes that accelerate tumor progression.
Collapse
|
21
|
Pedley R, Gilmore AP. Mitosis and mitochondrial priming for apoptosis. Biol Chem 2017; 397:595-605. [PMID: 27016149 DOI: 10.1515/hsz-2016-0134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/16/2016] [Indexed: 11/15/2022]
Abstract
Cell division is a period of danger for cells, as inaccurate segregation of chromosomes can lead to loss of cell viability or aneuploidy. In order to protect against these dangers, cells ultimately initiate mitochondrial apoptosis if they are unable to correctly exit mitosis. A number of important chemotherapeutics exploit this response to delayed mitotic exit, but despite this, the molecular mechanism of the apoptotic timer in mitosis has proved elusive. Some recent studies have now shed light on this, showing how passage through the cell cycle fine-tunes a cell's apoptotic sensitivity such that it can respond appropriately when errors arise.
Collapse
|
22
|
Hatok J, Racay P. Bcl-2 family proteins: master regulators of cell survival. Biomol Concepts 2017; 7:259-70. [PMID: 27505095 DOI: 10.1515/bmc-2016-0015] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
The most prominent function of proteins of the Bcl-2 family is regulation of the initiation of intrinsic (mitochondrial) pathways of apoptosis. However, recent research has revealed that in addition to regulation of mitochondrial apoptosis, proteins of the Bcl-2 family play important roles in regulating other cellular pathways with a strong impact on cell survival like autophagy, endoplasmic reticulum (ER) stress response, intracellular calcium dynamics, cell cycle progression, mitochondrial dynamics and energy metabolism. This review summarizes the recent knowledge about functions of Bcl-2 family proteins that are related to cell survival.
Collapse
|
23
|
Two coffins and a funeral: early or late caspase activation determines two types of apoptosis induced by DNA damaging agents. Apoptosis 2016; 22:421-436. [DOI: 10.1007/s10495-016-1337-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
24
|
Nair MS, Mony U, Menon D, Koyakutty M, Sidharthan N, Pavithran K, Nair SV, Menon KN. Development and molecular characterization of polymeric micro-nanofibrous scaffold of a defined 3-D niche for in vitro chemosensitivity analysis against acute myeloid leukemia cells. Int J Nanomedicine 2015; 10:3603-22. [PMID: 26028971 PMCID: PMC4440427 DOI: 10.2147/ijn.s80397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Standard in vitro drug testing employs 2-D tissue culture plate systems to test anti-leukemic drugs against cell adhesion-mediated drug-resistant leukemic cells that harbor in 3-D bone marrow microenvironments. This drawback necessitates the fabrication of 3-D scaffolds that have cell adhesion-mediated drug-resistant properties similar to in vivo niches. We therefore aimed at exploiting the known property of polyurethane (PU)/poly-l-lactic acid (PLLA) in forming a micro-nanofibrous structure to fabricate unique, not presented before, as far as we are aware, 3-D micro-nanofibrous scaffold composites using a thermally induced phase separation technique. Among the different combinations of PU/PLLA composites generated, the unique PU/PLLA 60:40 composite displayed micro-nanofibrous morphology similar to decellularized bone marrow with increased protein and fibronectin adsorption. Culturing of acute myeloid leukemia (AML) KG1a cells in FN-coated PU/PLLA 60:40 shows increased cell adhesion and cell adhesion-mediated drug resistance to the drugs cytarabine and daunorubicin without changing the original CD34+/CD38−/CD33− phenotype for 168 hours compared to fibronectin tissue culture plate systems. Molecularly, as seen in vivo, increased chemoresistance is associated with the upregulation of anti-apoptotic Bcl2 and the cell cycle regulatory protein p27Kip1 leading to cell growth arrest. Abrogation of Bcl2 activity by the Bcl2-specific inhibitor ABT 737 led to cell death in the presence of both cytarabine and daunorubicin, demonstrating that the cell adhesion-mediated drug resistance induced by Bcl2 and p27Kip1 in the scaffold was similar to that seen in vivo. These results thus show the utility of a platform technology, wherein drug testing can be performed before administering to patients without the necessity for stromal cells.
Collapse
Affiliation(s)
- Maya S Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Ullas Mony
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Deepthy Menon
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Manzoor Koyakutty
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Neeraj Sidharthan
- Department of Oncology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Keechilat Pavithran
- Department of Oncology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Shantikumar V Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| | - Krishnakumar N Menon
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham University, Kerala, India
| |
Collapse
|
25
|
Cekanova M, Fernando RI, Siriwardhana N, Sukhthankar M, Parra CDL, Woraratphoka J, Malone C, Ström A, Baek SJ, Wade PA, Saxton AM, Donnell RM, Pestell RG, Dharmawardhane S, Wimalasena J. BCL-2 family protein, BAD is down-regulated in breast cancer and inhibits cell invasion. Exp Cell Res 2014; 331:1-10. [PMID: 25499972 DOI: 10.1016/j.yexcr.2014.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 10/27/2014] [Accepted: 11/24/2014] [Indexed: 01/29/2023]
Abstract
We have previously demonstrated that the anti-apoptotic protein BAD is expressed in normal human breast tissue and shown that BAD inhibits expression of cyclin D1 to delay cell-cycle progression in breast cancer cells. Herein, expression of proteins in breast tissues was studied by immunohistochemistry and results were analyzed statistically to obtain semi-quantitative data. Biochemical and functional changes in BAD-overexpressing MCF7 breast cancer cells were evaluated using PCR, reporter assays, western blotting, ELISA and extracellular matrix invasion assays. Compared to normal tissues, Grade II breast cancers expressed low total/phosphorylated forms of BAD in both cytoplasmic and nuclear compartments. BAD overexpression decreased the expression of β-catenin, Sp1, and phosphorylation of STATs. BAD inhibited Ras/MEK/ERK and JNK signaling pathways, without affecting the p38 signaling pathway. Expression of the metastasis-related proteins, MMP10, VEGF, SNAIL, CXCR4, E-cadherin and TlMP2 was regulated by BAD with concomitant inhibition of extracellular matrix invasion. Inhibition of BAD by siRNA increased invasion and Akt/p-Akt levels. Clinical data and the results herein suggest that in addition to the effect on apoptosis, BAD conveys anti-metastatic effects and is a valuable prognostic marker in breast cancer.
Collapse
Affiliation(s)
- Maria Cekanova
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA.
| | - Romaine I Fernando
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Medical Center, The University of Tennessee, Knoxville, TN, USA
| | - Nalin Siriwardhana
- Department of Animal Science, The University of Tennessee, Knoxville, TN, USA
| | - Mugdha Sukhthankar
- Department of Biomedical and Diagnostics Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| | - Columba de la Parra
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Jirayus Woraratphoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Medical Center, The University of Tennessee, Knoxville, TN, USA
| | - Christine Malone
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Anders Ström
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Seung J Baek
- Department of Biomedical and Diagnostics Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| | - Paul A Wade
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Arnold M Saxton
- Department of Animal Science, The University of Tennessee, Knoxville, TN, USA
| | - Robert M Donnell
- Department of Biomedical and Diagnostics Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, USA
| | - Richard G Pestell
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Jay Wimalasena
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Medical Center, The University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
26
|
Oricchio E, Ciriello G, Jiang M, Boice MH, Schatz JH, Heguy A, Viale A, de Stanchina E, Teruya-Feldstein J, Bouska A, McKeithan T, Sander C, Tam W, Seshan VE, Chan WC, Chaganti RSK, Wendel HG. Frequent disruption of the RB pathway in indolent follicular lymphoma suggests a new combination therapy. ACTA ACUST UNITED AC 2014; 211:1379-91. [PMID: 24913233 PMCID: PMC4076578 DOI: 10.1084/jem.20132120] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Loss of cell cycle controls is a hallmark of cancer and has a well-established role in aggressive B cell malignancies. However, the role of such lesions in indolent follicular lymphoma (FL) is unclear and individual lesions have been observed with low frequency. By analyzing genomic data from two large cohorts of indolent FLs, we identify a pattern of mutually exclusive (P = 0.003) genomic lesions that impair the retinoblastoma (RB) pathway in nearly 50% of FLs. These alterations include homozygous and heterozygous deletions of the p16/CDKN2a/b (7%) and RB1 (12%) loci, and more frequent gains of chromosome 12 that include CDK4 (29%). These aberrations are associated with high-risk disease by the FL prognostic index (FLIPI), and studies in a murine FL model confirm their pathogenic role in indolent FL. Increased CDK4 kinase activity toward RB1 is readily measured in tumor samples and indicates an opportunity for CDK4 inhibition. We find that dual CDK4 and BCL2 inhibitor treatment is safe and effective against available models of FL. In summary, frequent RB pathway lesions in indolent, high-risk FLs indicate an untapped therapeutic opportunity.
Collapse
Affiliation(s)
- Elisa Oricchio
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Giovanni Ciriello
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Man Jiang
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Michael H Boice
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Jonathan H Schatz
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Adriana Heguy
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Agnes Viale
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Elisa de Stanchina
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Julie Teruya-Feldstein
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Alyssa Bouska
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tim McKeithan
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Chris Sander
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Wayne Tam
- Department of Pathology, Weill-Cornell Medical School, New York, NY 10065
| | - Venkatraman E Seshan
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Wing-Chung Chan
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - R S K Chaganti
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Hans-Guido Wendel
- Cancer Biology & Genetics Program, Computational Biology Program, Department of Medicine, Human Oncology and Pathogenesis Program, Genomics Core Facility, Molecular Pharmacology Program, Department of Pathology, Department of Epidemiology and Biostatistics, and Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
27
|
Wang S, Jiang Y, Liu J, Zhao Y, Xiang C, Ma R, Gao H, Jin L, He F, Wang H. Revisiting the role of MCL1 in tumorigenesis of solid cancer: gene expression correlates with antiproliferative phenotype in breast cancer cells and its functional regulatory variants are associated with reduced cancer susceptibility. Tumour Biol 2014; 35:8289-99. [PMID: 24852432 DOI: 10.1007/s13277-014-2108-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023] Open
Abstract
Compared to the well-defined anti-apoptotic role of myeloid cell leukemia sequence 1 (MCL1), its antiproliferative function in tumorigenesis is less studied. We had recently reported that regulatory variants of MCL1 contribute to enhanced promoter activity but reduced risk of lung cancer. We hypothesized that MCL1 expression may manifest antiproliferative phenotype and its functional variations may have etiological relevance for breast cancer. We manipulated MCL1 expression in MCF-7 cells and MDA231 with overexpression and knockdown, analyzed the effects on cell viability and cell cycling phase, and characterized the correlation with expression profiles of key regulators of cell cycle. We further genotyped the -190 insertion polymorphism and the neighboring single nucleotide polymorphisms (SNPs) in 745 breast cancer patients and 537 controls and analyzed their association with cancer risk. We confirmed that heightened expression of MCL1 resulted in decreased proliferation ability of breast cancer cells. We further observed that MCL1 overexpression in breast cancer cells resulted in cell cycle progression arresting in S phase and concomitant enhanced expression of p27, which could be rescued by p27 knockdown with co-transfection of small interfering RNA (siRNA). Furthermore, we found a significant reduction in breast cancer risk [odds ratio (OR) = 0.74; 95 % confidence interval (CI) = 0.59-0.93] associated with -190 insertion genotype; the expression-enhancing regulatory haplotype (OR 0.79; 95 % CI 0.66-0.95) and diplotype (OR 0.71; 95 % CI 0.57-0.89) were consistently associated with decreased cancer susceptibility. The study demonstrates that the expression-enhancing regulatory variants of MCL1 are protective modifiers of breast cancer risk, and reduced cell proliferation and arrested cell cycle progression partly mediated by p27 might be the underlying mechanism.
Collapse
Affiliation(s)
- Sheng Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Rd., 200433, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wang W, Wang D, Li H. Initiation of premature senescence by Bcl-2 in hypoxic condition. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2446-2453. [PMID: 24966955 PMCID: PMC4069934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Senescence, a state of cell cycle arrest, has been regarded as an intrinsic barrier to malignance. Although being repressed in most immortal tumors, the genetic program of senescence can be reactivated by critical regulators, including the apoptosis regulator Bcl-2. We showed here that hypoxic condition resulted in an irreversible senescence-like phenotype with increased expression of Bcl-2 in mouse melanoma B16 cells. In CoCl2-simulating hypoxic condition, characteristic morphological alterations and increased activity of senescence-associated β-galactosidase (SA-β-gal) can be detected with high level of Bcl-2, which was confirmed by western blot and co-staining of SA-β-gal and Bcl-2 by immunocytochemistry. Accordingly, Bcl-2 silence by specific siRNA ahead of hypoxia treatment interrupted the senescent development. Moreover Bcl-2 overexpression led to early onset of senescence. We propose that Bcl-2 is required to initiate and maintain the senescent phenotype. In addition, p53 and p16 were not involved in hypoxia-induced senescence according to the expression levels during senescent process. These results suggest that when encountering harmful stress (hypoxia), melanoma cells overexpress Bcl-2 and turn to senescence, a permanent cell-cycle arrest, for prolonged survival.
Collapse
|
29
|
Moriishi T, Kawai Y, Komori H, Rokutanda S, Eguchi Y, Tsujimoto Y, Asahina I, Komori T. Bcl2 deficiency activates FoxO through Akt inactivation and accelerates osteoblast differentiation. PLoS One 2014; 9:e86629. [PMID: 24466179 PMCID: PMC3896485 DOI: 10.1371/journal.pone.0086629] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 12/17/2013] [Indexed: 11/19/2022] Open
Abstract
Osteoblast apoptosis plays an important role in bone development and maintenance, and is in part responsible for osteoporosis in sex steroid deficiency, glucocorticoid excess, and aging. Although Bcl2 subfamily proteins, including Bcl2 and Bcl-XL, inhibit apoptosis, the physiological significance of Bcl2 in osteoblast differentiation has not been fully elucidated. To investigate this, we examined Bcl2-deficient (Bcl2(-/-)) mice. In Bcl2(-/-) mice, bromodeoxyuridine (BrdU)-positive osteoblasts were reduced in number, while terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL)-positive osteoblasts were increased. Unexpectedly, osteoblast differentiation was accelerated in Bcl2(-/-) mice as shown by the early appearance of osteocalcin-positive osteoblasts. Osteoblast differentiation was also accelerated in vitro when primary osteoblasts were seeded at a high concentration to minimize the reduction of the cell density by apoptosis during culture. FoxO transcription factors, whose activities are negatively regulated through the phosphorylation by Akt, play important roles in multiple cell events, including proliferation, death, differentiation, longevity, and stress response. Expressions of FasL, Gadd45a, and Bim, which are regulated by FoxOs, were upregulated; the expression and activity of FoxOs were enhanced; and the phosphorylation of Akt and that of FoxO1 and FoxO3a by Akt were reduced in Bcl2(-/-) calvariae. Further, the levels of p53 mRNA and protein were increased, and the expression of p53-target genes, Pten and Igfbp3 whose proteins inhibit Akt activation, was upregulated in Bcl2(-/-) calvariae. However, Pten but not Igfbp3 was upregulated in Bcl2(-/-) primary osteoblasts, and p53 induced Pten but not Igfbp3 in vitro. Silencing of either FoxO1 or FoxO3a inhibited and constitutively-active FoxO3a enhanced osteoblast differentiation. These findings suggest that Bcl2 deficiency induces and activates FoxOs through Akt inactivation, at least in part, by upregulating Pten expression through p53 in osteoblasts, and that the enhanced expression and activities of FoxOs may be one of the causes of accelerated osteoblast differentiation in Bcl2(-/-) mice.
Collapse
Affiliation(s)
- Takeshi Moriishi
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yosuke Kawai
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hisato Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoshi Rokutanda
- Department of Oral and Maxillofacial Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yutaka Eguchi
- Department of Molecular Genetics, Osaka University Medical School, Osaka, Japan
| | - Yoshihide Tsujimoto
- Department of Molecular Genetics, Osaka University Medical School, Osaka, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
30
|
Luna-López A, González-Puertos VY, Romero-Ontiveros J, Ventura-Gallegos JL, Zentella A, Gomez-Quiroz LE, Königsberg M. A noncanonical NF-κB pathway through the p50 subunit regulates Bcl-2 overexpression during an oxidative-conditioning hormesis response. Free Radic Biol Med 2013; 63:41-50. [PMID: 23648765 DOI: 10.1016/j.freeradbiomed.2013.04.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/27/2013] [Accepted: 04/25/2013] [Indexed: 11/30/2022]
Abstract
Cells can respond to damage and stress by activating various repair and survival pathways. One of these responses can be induced by preconditioning the cells with sublethal stress to provoke a prosurvival response that will prevent damage and death, and which is known as hormesis. Bcl-2, an antiapoptotic protein recognized by its antioxidant and prosurvival functions, has been documented to play an important role during oxidative-conditioning hormesis. Using an oxidative-hormetic model, which was previously established in the L929 cell line by subjecting the cells to a mild oxidative stress of 50 μM H₂O₂ for 9 h, we identified two different transductional mechanisms that participate in the regulation of Bcl-2 expression during the hormetic response. These mechanisms converge in activating the nuclear transcription factor NF-κB. Interestingly, the noncanonical p50 subunit of the NF-κB family is apparently the subunit that participates during the oxidative-hormetic response.
Collapse
|
31
|
Removal of the BH4 domain from Bcl-2 protein triggers an autophagic process that impairs tumor growth. Neoplasia 2013; 15:315-27. [PMID: 23479509 DOI: 10.1593/neo.121392] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 12/19/2022] Open
Abstract
Here, we show that forced expression of a B-cell lymphoma 2 (bcl-2) protein lacking residues 1 to 36 at the N-terminal, including the entire Bcl-2 homology 4 (BH4) domain, determines reduction of in vitro and in vivo human melanoma growth. Noteworthy, melanoma cells in vivo exhibit markedly increased autophagy, as response to expression of bcl-2 protein deleted of its BH4 domain. This observation led to the identification of a novel gain of function for bcl-2 protein lacking the BH4 domain. In particular, upon different autophagic stimuli in vitro, overexpression of bcl-2 protein deleted of BH4 domain induces autophagosome accumulation, conversion of microtubule-associated protein 1 light chain 3B-II, reduced expression of p62/SQSTM1 protein, and thereby enhanced autophagic flux. The relevance of Beclin-1 is evidenced by the fact that 1) the autophagy-promoting and growth-inhibiting properties are partially rescued by Beclin-1 knockdown in cells expressing bcl-2 protein lacking the BH4 domain, 2) Beclin-1 only interacts with wild-type but not with deleted bcl-2, and 3) BH4 domain removal from bcl-2 protein does not influence in vitro and in vivo growth of tumor cells expressing low levels of endogenous Beclin-1. These results provide new insight into molecular mechanism of bcl-2 functions and represent a rationale for the development of agents interfering with the BH4 domain of bcl-2 protein.
Collapse
|
32
|
Braun F, de Carné Trécesson S, Bertin-Ciftci J, Juin P. Protect and serve: Bcl-2 proteins as guardians and rulers of cancer cell survival. Cell Cycle 2013; 12:2937-47. [PMID: 23974114 PMCID: PMC3875667 DOI: 10.4161/cc.25972] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
It is widely accepted that anti-apoptotic Bcl-2 family members promote cancer cell survival by binding to their pro-apoptotic counterparts, thereby preventing mitochondrial outer membrane permeabilization (MOMP) and cytotoxic caspase activation. Yet, these proteins do not only function as guardians of mitochondrial permeability, preserving it, and maintaining cell survival in the face of acute or chronic stress, they also regulate non-apoptotic functions of caspases and biological processes beyond MOMP from diverse subcellular localizations and in complex with numerous binding partners outside of the Bcl-2 family. In particular, some of the non-canonical effects and functions of Bcl-2 homologs lead to an interplay with E2F-1, NFκB, and Myc transcriptional pathways, which themselves influence cancer cell growth and survival. We thus propose that, by feedback loops that we currently have only hints of, Bcl-2 proteins may act as rulers of survival signaling, predetermining the apoptotic threshold that they also directly scaffold. This underscores the robustness of the control exerted by Bcl-2 homologs over cancer cell survival, and implies that small molecules compounds currently used in the clinic to inhibit their mitochondrial activity may be not always be fully efficient to override this control.
Collapse
Affiliation(s)
- Frédérique Braun
- UMR 892 INSERM/6299 CNRS/Université de Nantes; Team 8 "Cell survival and tumor escape in breast cancer"; Institut de Recherche en Santé de l'Université de Nantes; Nantes, France
| | | | | | | |
Collapse
|
33
|
Frauenstein K, Sydlik U, Tigges J, Majora M, Wiek C, Hanenberg H, Abel J, Esser C, Fritsche E, Krutmann J, Haarmann-Stemmann T. Evidence for a novel anti-apoptotic pathway in human keratinocytes involving the aryl hydrocarbon receptor, E2F1, and checkpoint kinase 1. Cell Death Differ 2013; 20:1425-34. [PMID: 23912710 DOI: 10.1038/cdd.2013.102] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 06/14/2013] [Accepted: 07/04/2013] [Indexed: 01/22/2023] Open
Abstract
Exposure of keratinocytes (KC) to ultraviolet (UV) radiation results in the initiation of apoptosis, a protective mechanism that eliminates cells harboring irreparable DNA damage. Hence, a manipulation of UV-induced apoptosis may significantly influence photocarcinogenesis. We have discovered that the aryl hydrocarbon receptor (AHR), a key regulator of drug metabolism and an UVB-sensitive transcription factor, serves an anti-apoptotic function in UVB-irradiated human KC. Chemical and shRNA-mediated inhibition of AHR signaling sensitized KC to UVB-induced apoptosis by decreasing the expression of E2F1 and its target gene checkpoint kinase 1 (CHK1). The decreased expression of these cell-cycle regulators was due to an enhanced expression of p27(KIP1) and an associated decrease in phosphorylation of both cyclin-dependent kinase 2 and its substrate molecule retinoblastoma protein. The subsequent inhibition of E2F1 autoregulation and downstream CHK1 expression resulted in an enhanced susceptibility of damaged cells to undergo apoptosis. Accordingly, ectopic overexpression of either E2F1 or CHK1 in AHR-knockdown KC attenuated the observed sensitization to UVB-induced apoptosis. Using an AHR-knockout SKH-1 hairless mouse model, we next demonstrated the physiological relevance of the anti-apoptotic function of AHR. In contrast to their AHR-proficient littermates, the constitutive expression of E2F1 and CHK1 was significantly reduced in the skin of AHR-knockout mice. Accordingly, a single exposure of the animals to UVB resulted in an enhanced cleavage of caspase-3 in the skin of AHR-knockout mice. These results identify for the first time the AHR-E2F1-CHK1 axis as a novel anti-apoptotic pathway in KC, which may represent a suitable target for chemoprevention of non-melanoma skin cancer.
Collapse
Affiliation(s)
- K Frauenstein
- IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hasan SMM, Sheen AD, Power AM, Langevin LM, Xiong J, Furlong M, Day K, Schuurmans C, Opferman JT, Vanderluit JL. Mcl1 regulates the terminal mitosis of neural precursor cells in the mammalian brain through p27Kip1. Development 2013; 140:3118-27. [PMID: 23824576 DOI: 10.1242/dev.090910] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cortical development requires the precise timing of neural precursor cell (NPC) terminal mitosis. Although cell cycle proteins regulate terminal mitosis, the factors that influence the cell cycle machinery are incompletely understood. Here we show in mice that myeloid cell leukemia 1 (Mcl1), an anti-apoptotic Bcl-2 protein required for the survival of NPCs, also regulates their terminal differentiation through the cell cycle regulator p27(Kip1). A BrdU-Ki67 cell profiling assay revealed that in utero electroporation of Mcl1 into NPCs in the embryonic neocortex increased NPC cell cycle exit (the leaving fraction). This was further supported by a decrease in proliferating NPCs (Pax6(+) radial glial cells and Tbr2(+) neural progenitors) and an increase in differentiating cells (Dcx(+) neuroblasts and Tbr1(+) neurons). Similarly, BrdU birth dating demonstrated that Mcl1 promotes premature NPC terminal mitosis giving rise to neurons of the deeper cortical layers, confirming their earlier birthdate. Changes in Mcl1 expression within NPCs caused concomitant changes in the levels of p27(Kip1) protein, a key regulator of NPC differentiation. Furthermore, in the absence of p27(Kip1), Mcl1 failed to induce NPC cell cycle exit, demonstrating that p27(Kip1) is required for Mcl1-mediated NPC terminal mitosis. In summary, we have identified a novel physiological role for anti-apoptotic Mcl1 in regulating NPC terminal differentiation.
Collapse
Affiliation(s)
- S M Mahmudul Hasan
- Division of BioMedical Sciences, Memorial University, 300 Prince Philip Drive, St John's, NL A1B 3V6, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lam LT, Zhang H, Chyla B. Biomarkers of therapeutic response to BCL2 antagonists in cancer. Mol Diagn Ther 2013; 16:347-56. [PMID: 23023732 DOI: 10.1007/s40291-012-0003-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer cells persist by resisting programmed cell death or apoptosis. In particular, an imbalance of proteins that regulate apoptosis leads to lack of response to apoptotic stimuli. Thus, restoring the ability of cancer cells to undergo apoptosis is highly desirable. One apoptosis pathway, the intrinsic pathway, involves perturbation of the mitochondria. The major players of this pathway are the members of the B cell CLL/lymphoma 2 (BCL2) family. Currently, three BCL2 antagonists are in clinical trials for cancer treatment. While these antagonists show various specificity and potency, the development of companion diagnostics is crucial for developing these compounds into viable cancer treatments. In this review we describe predictive and pharmacodynamic biomarkers for these agents. Future directions on biomarker development for this class of antagonist are also discussed.
Collapse
Affiliation(s)
- Lloyd T Lam
- Department R4CD, Global Pharmaceutical R&D, Abbott Laboratories, Building AP-10, 100 Abbott Park Road, Abbott Park, IL 60064, USA.
| | | | | |
Collapse
|
36
|
Iglesias M, Postigo J, Santiuste I, González J, Buelta L, Tamayo E, Merino J, Merino R. p27(Kip1) inhibits systemic autoimmunity through the control of Treg cell activity and differentiation. ACTA ACUST UNITED AC 2013; 65:343-54. [PMID: 23124840 DOI: 10.1002/art.37778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 10/25/2012] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Despite the importance of Treg cells in the maintenance of immunologic tolerance, the mechanisms that control their generation and activity are unknown. Since the cell cycle inhibitor p27(Kip1) (p27) was involved in T cell anergy, we undertook this study to explore its role in both Treg cell processes. METHODS The development of type II collagen-induced arthritis (CIA) and lupus-like abnormalities was compared between transgenic mice overexpressing human Bcl-2 in T cells (BCL2-TgT mice) and nontransgenic mice that were deficient or not deficient in p27. The contribution of Treg cells to disease evolution was also explored. Finally, the in vitro activity of Treg cells and their differentiation from naive CD4+ cells was compared between these strains of mice. RESULTS BCL2-TgT mice were protected against CIA by a Treg cell-dependent mechanism. In association with this protection, the overexpression of Bcl-2 in T cells enhanced the differentiation and activity of Treg cells. Both Bcl-2 effects were independent of its antiapoptotic activity but dependent on its capacity to induce the expression of p27 that augmented the strength of transforming growth factor β (TGFβ) signaling in T cells. Accordingly, down-modulation of p27 expression in BCL2-TgT mice promoted CIA. In addition, p27 deficiency in aged C57BL/6 mice reduced the number and activity of Treg cells and induced the development of mild lupus-like abnormalities. CONCLUSION Our results point to p27 as a critical regulator of Treg cell differentiation and function through the positive modulation of TGFβ signaling strength in T cells.
Collapse
|
37
|
Beclin 1 interactome controls the crosstalk between apoptosis, autophagy and inflammasome activation: impact on the aging process. Ageing Res Rev 2013; 12:520-34. [PMID: 23220384 DOI: 10.1016/j.arr.2012.11.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 11/28/2012] [Accepted: 11/28/2012] [Indexed: 01/10/2023]
Abstract
Autophagy and apoptosis are crucial cellular housekeeping and tissue survival mechanisms. There is emerging evidence of important crosstalk between apoptosis and autophagy which can be linked to inflammasome activation. Beclin 1 is a platform protein which assembles an interactome consisting of diverse proteins which control the initiation of autophagocytosis and distinct phases in endocytosis. Recent studies have demonstrated that the anti-apoptotic Bcl-2 family members can interact with Beclin 1 and inhibit autophagy. Consequently, impaired autophagy can trigger inflammasome activation. Interestingly, the hallmarks of the ageing process include a decline in autophagy, increased resistance to apoptosis and a low-grade inflammatory phenotype. Age-related stresses, e.g. genotoxic, metabolic and environmental insults, enhance the expression of NF-κB-driven anti-apoptotic Bcl-2 proteins which repress the Beclin 1-dependent autophagy. Suppression of autophagocytosis provokes inflammation including NF-κB activation which further potentiates anti-apoptotic defence. In a context-dependent manner, this feedback defence mechanism can enhance the aging process or provoke tumorigenesis or cellular senescence. We will review the role of Beclin 1 interactome in the crosstalk between apoptosis, autophagy and inflammasomes emphasizing that disturbances in Beclin 1-dependent autophagy can have a crucial impact on the aging process.
Collapse
|
38
|
Sionov RV. MicroRNAs and Glucocorticoid-Induced Apoptosis in Lymphoid Malignancies. ISRN HEMATOLOGY 2013; 2013:348212. [PMID: 23431463 PMCID: PMC3569899 DOI: 10.1155/2013/348212] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 11/14/2012] [Indexed: 12/20/2022]
Abstract
The initial response of lymphoid malignancies to glucocorticoids (GCs) is a critical parameter predicting successful treatment. Although being known as a strong inducer of apoptosis in lymphoid cells for almost a century, the signaling pathways regulating the susceptibility of the cells to GCs are only partly revealed. There is still a need to develop clinical tests that can predict the outcome of GC therapy. In this paper, I discuss important parameters modulating the pro-apoptotic effects of GCs, with a specific emphasis on the microRNA world comprised of small players with big impacts. The journey through the multifaceted complexity of GC-induced apoptosis brings forth explanations for the differential treatment response and raises potential strategies for overcoming drug resistance.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Department of Biochemistry and Molecular Biology, The Institute for Medical Research-Israel-Canada, Hadassah Medical School, The Hebrew University of Jerusalem, Ein-Kerem, 91120 Jerusalem, Israel
| |
Collapse
|
39
|
Abo-Zeid MAM, Liehr T, El-Daly SM, Gamal-Eldeen AM, Glei M, Shabaka A, Bhatt S, Hamid A. Molecular cytogenetic evaluation of the efficacy of photodynamic therapy by indocyanine green in breast adenocarcinoma MCF-7 cells. Photodiagnosis Photodyn Ther 2012; 10:194-202. [PMID: 23769286 DOI: 10.1016/j.pdpdt.2012.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 09/22/2012] [Accepted: 11/05/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is used for the treatment of many types of predominantly epithelial cancers. Photosensitizer is taken up by fast growing tumor cells more actively than by other body cells and is activated by light, generating reactive oxygen species that cause cell death by necrosis or apoptosis. This study aimed to evaluate the efficacy of PDT with indocyanine green (ICG) through the investigation of TP53, HER-2 and TOP2A genes signals as breast cancer gene markers by interphase fluorescence in situ hybridization (nuc-FISH). METHODS The photosynthetizer ICG (200 μM) was applied to breast cancer cell line MCF-7 cells (adenocarcinoma) in combination with laser irradiation (807 nm) exposure for 20 min and then incubated for 12, 24 and 48 h. Cell viability was evaluated using trypan blue. The signals for nuc-FISH was investigated and counted for probes specific for the genes TP53 (17p13), HER-2 (17q11.2-q12), and TOP2A (17q21-q22), and BAC-probes RP11-746M1 in 17p11.2 and RP11-403E9 in 17q11.2. RESULTS The cell viability of MCF-7 did not reduced significantly when the cells were treated with ICG (200 μM) or exposed to laser irradiation for 20 min followed by incubation for 24 h. ICG/PDT treatment with laser irradiation exposure for 20 min reduced the cell viability after incubating cells for 12, 24 and 48 h highly significantly in a time dependent manner. For nuc-FISH analysis, TP53, HER-2, TOP2A, RP11-746M1 and RP11-403E9 signals did not reduce or increase in a significant manner when the cells were treated with ICG or exposed to laser irradiation for 20 min then incubated for 24h. PDT enhanced amplification of TP53 signals from nuc ish 17p13(TP53×2) to nuc ish 17p13(TP53×3) or nuc ish 17p13(TP53×4). However, the signals of HER-2 gene, TOP2A gene and BAC probes were reduced highly significantly when MCF-7 cells were treated with PDT with all time intervals. CONCLUSION ICG/PDT and laser induced cytotoxic effect in MCF-7 cells. Also, PDT enhanced TP53 gene amplification, and reduced HER-2, TOP2A, and BAC probes RP11-746M1 and RP11-403E9 signals. Therefore ICG/PDT can be used for breast cancer treatment. It has the potential to induce apoptotic effect and reduce HER-2 and TOP2A genes propagation. Further in vivo studies are needed to evaluate ICG/PDT as a promising therapeutic approach for breast cancer.
Collapse
Affiliation(s)
- Mona A M Abo-Zeid
- Genetics and Cytology Department, Genetic Engineering and Biotechnology Division, National Research Center, Dokki 12622, Cairo, Egypt.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
The secret life of Bcl-2: Apoptosis-independent inhibition of DNA repair by Bcl-2 family members. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2012; 751:247-257. [DOI: 10.1016/j.mrrev.2012.05.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/25/2012] [Accepted: 05/26/2012] [Indexed: 11/22/2022]
|
41
|
Non-apoptotic functions of apoptosis-regulatory proteins. EMBO Rep 2012; 13:322-30. [PMID: 22402666 DOI: 10.1038/embor.2012.19] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Accepted: 01/23/2012] [Indexed: 01/24/2023] Open
Abstract
During the past two decades, apoptotic cell death has been the subject of an intense wave of investigation, leading to the discovery of multiple gene products that govern both its induction and execution. In parallel, it has progressively become evident that most, if not all, proteins that had initially been discovered for their essential role in apoptosis also mediate a wide range of non-apoptotic functions. On the one hand, apoptotic regulators and executioners are involved in non-lethal physiological processes as diverse as cell cycle progression, differentiation, metabolism, autophagy and inflammation. On the other hand, pro-apoptotic proteins can control other modalities of programmed cell death, in particular regulated necrosis. In this review, we summarize the unconventional roles of the apoptotic core machinery from a functional perspective and discuss their pathophysiological implications.
Collapse
|
42
|
Moriishi T, Maruyama Z, Fukuyama R, Ito M, Miyazaki T, Kitaura H, Ohnishi H, Furuichi T, Kawai Y, Masuyama R, Komori H, Takada K, Kawaguchi H, Komori T. Overexpression of Bcl2 in osteoblasts inhibits osteoblast differentiation and induces osteocyte apoptosis. PLoS One 2011; 6:e27487. [PMID: 22114675 PMCID: PMC3219663 DOI: 10.1371/journal.pone.0027487] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 10/18/2011] [Indexed: 12/03/2022] Open
Abstract
Bcl2 subfamily proteins, including Bcl2 and Bcl-XL, inhibit apoptosis. As osteoblast apoptosis is in part responsible for osteoporosis in sex steroid deficiency, glucocorticoid excess, and aging, bone loss might be inhibited by the upregulation of Bcl2; however, the effects of Bcl2 overexpression on osteoblast differentiation and bone development and maintenance have not been fully investigated. To investigate these issues, we established two lines of osteoblast-specific BCL2 transgenic mice. In BCL2 transgenic mice, bone volume was increased at 6 weeks of age but not at 10 weeks of age compared with wild-type mice. The numbers of osteoblasts and osteocytes increased, but osteoid thickness and the bone formation rate were reduced in BCL2 transgenic mice with high expression at 10 weeks of age. The number of BrdU-positive cells was increased but that of TUNEL-positive cells was unaltered at 2 and 6 weeks of age. Osteoblast differentiation was inhibited, as shown by reduced Col1a1 and osteocalcin expression. Osteoblast differentiation of calvarial cells from BCL2 transgenic mice also fell in vitro. Overexpression of BCL2 in primary osteoblasts had no effect on osteoclastogenesis in co-culture with bone marrow cells. Unexpectedly, overexpression of BCL2 in osteoblasts eventually caused osteocyte apoptosis. Osteocytes, which had a reduced number of processes, gradually died with apoptotic structural alterations and the expression of apoptosis-related molecules, and dead osteocytes accumulated in cortical bone. These findings indicate that overexpression of BCL2 in osteoblasts inhibits osteoblast differentiation, reduces osteocyte processes, and causes osteocyte apoptosis.
Collapse
Affiliation(s)
- Takeshi Moriishi
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Zenjiro Maruyama
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Sensory and Motor System Medicine, University of Tokyo, Tokyo, Japan
| | - Ryo Fukuyama
- Laboratory of Pharmacology, Hiroshima International University, Kure, Japan
| | - Masako Ito
- Department of Radiology and Radiation Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Toshihiro Miyazaki
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hideki Kitaura
- Division of Orthodontic and Biomedical Engineering, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Hidetake Ohnishi
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Orthodontics and Dentofacial Orthopedics, Faculty of Dentistry, Osaka University, Osaka, Japan
| | - Tatsuya Furuichi
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Laboratory Animal Facility, Research Center for Medical Sciences, School of Medicine, Jikei University, Tokyo, Japan
| | - Yosuke Kawai
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ritsuko Masuyama
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hisato Komori
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kenji Takada
- Department of Orthodontics and Dentofacial Orthopedics, Faculty of Dentistry, Osaka University, Osaka, Japan
| | - Hiroshi Kawaguchi
- Department of Sensory and Motor System Medicine, University of Tokyo, Tokyo, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
43
|
Abstract
Hematopoietic stem cells (HSC) are a relatively quiescent pool of cells that perform the arduous task of replacing the short-lived mature cells of the peripheral blood. While a rapid expansion of HSCs under periods of hematological stress is warranted, their enhanced proliferation during homeostasis leads to loss of function. We recently reported that in HSCs, the evolutionarily conserved growth factor erv1-like (Gfer) acts to counter jun activation domain-binding protein 1 (Jab1)-mediated nuclear export and destabilization of the cell cycle inhibitor, p27kip1, by directly binding to and sequestering the COP9 signalosome (CSN) subunit. Through this mechanism, Gfer promotes quiescence and maintains the functional integrity of HSCs. Here, we extend our study to demonstrate an association between Gfer and Ca2+/calmodulin-dependent protein kinase IV (CaMKIV) in the regulation of HSC proliferation. Highly proliferative and functionally deficient Camk4-/- HSCs possess significantly lower levels of Gfer and p27kip1. Ectopic expression of Gfer restores quiescence and elevates p27kip1 expression in Camk4-/- HSCs. These results further substantiate a critical role for Gfer in the restriction of unwarranted proliferation in HSCs through the inhibition of Jab1 and subsequent stabilization and nuclear retention of p27kip1. This Gfer-mediated pro-quiescence mechanism could be therapeutically exploited in the treatment of hematological malignancies associated with elevated Jab1 and reduced p27kip1.
Collapse
Affiliation(s)
- Uma Sankar
- James Graham Brown Cancer Center and Owensboro Cancer Research Program, Department of Pharmacology and Toxicology, University of Louisville, Lousville, KY, USA.
| | | |
Collapse
|
44
|
Bhattacharyya S, Deb J, Patra AK, Thuy Pham DA, Chen W, Vaeth M, Berberich-Siebelt F, Klein-Hessling S, Lamperti ED, Reifenberg K, Jellusova J, Schweizer A, Nitschke L, Leich E, Rosenwald A, Brunner C, Engelmann S, Bommhardt U, Avots A, Müller MR, Kondo E, Serfling E. NFATc1 affects mouse splenic B cell function by controlling the calcineurin--NFAT signaling network. ACTA ACUST UNITED AC 2011; 208:823-39. [PMID: 21464221 PMCID: PMC3135343 DOI: 10.1084/jem.20100945] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mouse B cells lacking NFATc1 exhibit defective proliferation, survival, isotype class switching, cytokine production, and T cell help. By studying mice in which the Nfatc1 gene was inactivated in bone marrow, spleen, or germinal center B cells, we show that NFATc1 supports the proliferation and suppresses the activation-induced cell death of splenic B cells upon B cell receptor (BCR) stimulation. BCR triggering leads to expression of NFATc1/αA, a short isoform of NFATc1, in splenic B cells. NFATc1 ablation impaired Ig class switch to IgG3 induced by T cell–independent type II antigens, as well as IgG3+ plasmablast formation. Mice bearing NFATc1−/− B cells harbor twofold more interleukin 10–producing B cells. NFATc1−/− B cells suppress the synthesis of interferon-γ by T cells in vitro, and these mice exhibit a mild clinical course of experimental autoimmune encephalomyelitis. In large part, the defective functions of NFATc1−/− B cells are caused by decreased BCR-induced Ca2+ flux and calcineurin (Cn) activation. By affecting CD22, Rcan1, CnA, and NFATc1/αA expression, NFATc1 controls the Ca2+-dependent Cn–NFAT signaling network and, thereby, the fate of splenic B cells upon BCR stimulation.
Collapse
Affiliation(s)
- Sankar Bhattacharyya
- Department of Molecular Pathology, University of Würzburg, D-97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wu YCM, O'Reilly MA. Bcl-X(L) is the primary mediator of p21 protection against hyperoxia-induced cell death. Exp Lung Res 2010; 37:82-91. [PMID: 21128858 DOI: 10.3109/01902148.2010.521617] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A tight balance between anti- and proapoptotic members of the Bcl-2 family controls cell survival and death. Exposure to hyperoxia shifts this balance towards a prodeath state that ultimately activates Bak- and Bax-dependent cell death. Mechanisms underlying this shift are undefined; however, the cell cycle inhibitor p21 delays the loss of antiapoptotic Mcl-1 and Bcl-X(L), and protects against hyperoxia. Here, H1299 human lung adenocarcinoma cells are used to investigate how these and other members of the Bcl-2 family cooperate with p21 to protect against hyperoxia. Expression of antiapoptotic Mcl-1 and Bcl-X(L), but not Bcl-2 or A1, declined during hyperoxia, whereas proapoptotic Bak, but not Bax, increased. Conditional overexpression of p21 selectively delayed the loss of Mcl-1 and Bcl-X(L), without affecting expression of the other members. siRNA knockdown of Mcl-1 and Bcl-X(L) sensitized cells to hyperoxia, but only the loss of Bcl-X(L) ablated the protective effects of p21. Conversely, overexpression of Mcl-1 and Bcl-X(L) protected against hyperoxia, but only Bcl-X(L) bound Bak and Bax. Altogether, these data suggest that Bcl-X(L) is the primary mediator by which p21 protects against hyperoxia-induced Bak/Bax-dependent cell death.
Collapse
Affiliation(s)
- Yu-Chieh M Wu
- Department of Biomedical Genetics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York 14642, USA
| | | |
Collapse
|
46
|
Santiuste I, Buelta L, Iglesias M, Genre F, Mazorra F, Izui S, Merino J, Merino R. B-cell overexpression of Bcl-2 cooperates with p21 deficiency for the induction of autoimmunity and lymphomas. J Autoimmun 2010; 35:316-24. [PMID: 20691570 DOI: 10.1016/j.jaut.2010.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/08/2010] [Accepted: 07/09/2010] [Indexed: 11/13/2022]
Abstract
Genetic abnormalities predisposing to autoimmunity generally act in a cooperative manner affecting one or several mechanisms regulating immunological tolerance. In addition, many of these genetic abnormalities are also involved in the development of lymphoproliferative diseases. In the present study, we have determined the possible cooperation between deficiencies in members of the Cip/Kip family of cell cycle regulators (p21(WAF1/Cip1) or p27(kip1)) and the overexpression of human Bcl-2 in B lymphocytes in the induction of autoimmune and lymphoproliferative diseases in non-autoimmune C57BL/6 (B6) mice. Unlike single mutant mice, B6.p21(-/-) mice transgenic for human Bcl-2 in B cells developed a lethal autoimmune syndrome characterized by the production of autoantibodies, the prominent expansion of memory B and CD4(+) T cells and the development of severe glomerular lesions resembling IgA nephropathy. Furthermore, these mice presented a high incidence of B-cell lymphoproliferative disorders. Such genetic cooperation in the induction of autoimmunity was not observed in B6.p27(-/-) mice transgenic for human Bcl-2 in B cells. Altogether, what we have demonstrated here is the existence of preferential interactions among particular regulators of the G(1)/S transition of the cell cycle and B-cell survival in the induction of systemic autoimmune and lymphoproliferative diseases.
Collapse
Affiliation(s)
- Inés Santiuste
- Departmento de Biología Molecular, Universidad de Cantabria-Instituto de Formación e Investigación Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | | | | |
Collapse
|
47
|
González-Puertos VY, Hernández-Pérez E, Nuño-Lámbarri N, Ventura-Gallegos JL, López-Diázguerrero NE, Robles-Díaz G, Gutiérrez-Ruiz MC, Konigsberg M. Bcl-2 overexpression in hepatic stellate cell line CFSC-2G, induces a pro-fibrotic state. J Gastroenterol Hepatol 2010; 25:1306-14. [PMID: 20594261 DOI: 10.1111/j.1440-1746.2009.06175.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIM Development of hepatic fibrosis is a complex process that involves oxidative stress (OS) and an altered balance between pro- and anti-apoptotic molecules. Since Bcl-2 overexpression preserves viability against OS, our objective was to address the effect of Bcl-2 overexpression in the hepatic stellate cells (HSC) cell-line CFSC-2G under acetaldehyde and H(2)O(2) challenge, and explore if it protects these cells against OS, induces replicative senescence and/or modify extracellular matrix (ECM) remodeling potential. METHODS To induce Bcl-2 overexpression, HSC cell line CFSC-2G was transfected by lipofection technique. Green fluorescent protein-only CFSC-2G cells were used as a control. Cell survival after H(2)O(2) treatment and total protein oxidation were assessed. To determine cell cycle arrest, proliferation-rate, DNA synthesis and senescence were assessed. Matrix metalloproteinases (MMP), tissue-inhibitor of MMP (TIMP), transglutaminases (TG) and smooth muscle a-actin (alpha-SMA) were evaluated by western blot in response to acetaldehyde treatment as markers of ECM remodeling capacity in addition to transforming growth factor-beta (TGF-beta) mRNA. RESULTS Cells overexpressing Bcl-2 survived approximately 20% more than control cells when exposed to H(2)O(2) and approximately 35% proteins were protected from oxidation, but Bcl-2 did not slow proliferation or induced senescence. Bcl-2 overexpression did not change alpha-SMA levels, but it increased TIMP-1 (55%), tissue transglutaminases (tTG) (25%) and TGF-beta mRNA (49%), when exposed to acetaldehyde, while MMP-13 content decreased (47%). CONCLUSIONS Bcl-2 overexpression protected HSC against oxidative stress but it did not induce replicative senescence. It increased TIMP-1, tTG and TGF-beta mRNA levels and decreased MMP-13 content, suggesting that Bcl-2 overexpression may play a key role in the progression of fibrosis in chronic liver diseases.
Collapse
Affiliation(s)
- Viridiana Y González-Puertos
- Health and Sciences Department, Biological and Health Sciences Division, Metropolitan Independent University, México, DF, México
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Cova E, Ghiroldi A, Guareschi S, Mazzini G, Gagliardi S, Davin A, Bianchi M, Ceroni M, Cereda C. G93A SOD1 alters cell cycle in a cellular model of Amyotrophic Lateral Sclerosis. Cell Signal 2010; 22:1477-84. [PMID: 20561900 DOI: 10.1016/j.cellsig.2010.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 05/26/2010] [Indexed: 12/14/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative multifactorial disease characterized, like other diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) or frontotemporal dementia (FTD), by the degeneration of specific neuronal cell populations. Motor neuron loss is distinctive of ALS. However, the causes of onset and progression of motor neuron death are still largely unknown. In about 2% of all cases, mutations in the gene encoding for the Cu/Zn superoxide dismutase (SOD1) are implicated in the disease. Several alterations in the expression or activation of cell cycle proteins have been described in the neurodegenerative diseases and related to cell death. In this work we show that mutant SOD1 can alter cell cycle in a cellular model of ALS. Our findings suggest that modifications in the cell cycle progression could be due to an increased interaction between mutant G93A SOD1 and Bcl-2 through the cyclins regulator p27. As previously described in post mitotic neurons, cell cycle alterations could fatally lead to cell death.
Collapse
Affiliation(s)
- Emanuela Cova
- Laboratory of Experimental Neurobiology, IRCCS, National Neurological Institute C. Mondino, Via Mondino, 2, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Krampe B, Al-Rubeai M. Cell death in mammalian cell culture: molecular mechanisms and cell line engineering strategies. Cytotechnology 2010; 62:175-88. [PMID: 20502964 DOI: 10.1007/s10616-010-9274-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 04/12/2010] [Indexed: 12/15/2022] Open
Abstract
Cell death is a fundamentally important problem in cell lines used by the biopharmaceutical industry. Environmental stress, which can result from nutrient depletion, by-product accumulation and chemical agents, activates through signalling cascades regulators that promote death. The best known key regulators of death process are the Bcl-2 family proteins which constitute a critical intracellular checkpoint of apoptosis cell death within a common death pathway. Engineering of several members of the anti-apoptosis Bcl-2 family genes in several cell types has extended the knowledge of their molecular function and interaction with other proteins, and their regulation of cell death. In this review, we describe the various modes of cell death and their death pathways at molecular and organelle level and discuss the relevance of the growing knowledge of anti-apoptotic engineering strategies to inhibit cell death and increase productivity in mammalian cell culture.
Collapse
Affiliation(s)
- Britta Krampe
- School of Chemical and Bioprocess Engineering, and Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Republic of Ireland
| | | |
Collapse
|
50
|
Marín-Vidalled MJ, Bolívar A, Zubiaga A, López-Hoyos M. The combined effect of BCL-2 over-expression and E2F2 deficiency induces an autoimmune syndrome in non-susceptible mouse strain C57BL/6. Autoimmunity 2010; 43:111-20. [PMID: 20187704 DOI: 10.3109/08916930903214033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Multiple evidences support the notion that cell-cycle deregulation or apoptosis alterations can lead to autoimmune syndrome (AIS). Inactivation of the cell-cycle regulator E2F2 or over-expression of the anti-apoptotic Bcl-2 protein induces spontaneously an AIS in certain mouse strains. In the present study, we have examined the contribution of the genetic background on the development of autoimmunity after E2F2 gene inactivation, and the effect that a simultaneous inactivation of the E2F2 gene and over-expression of the Bcl-2 gene in B cells has on lymphoid homeostasis and autoimmunity. We show that E2F2(- / - ) mice carrying wild-type levels of Bcl-2 do not develop AIS when they are in a non-pro-autoimmune background (C57BL/6). However, mice harboring both genetic alterations concomitantly develop late AIS characterized by the presence of serum anti-nuclear antibodies, double and single strand anti-DNA antibodies, and the development of a mild glomerulonephritis with mesangial immunoglobulins, mainly IgA, deposits. These results suggest that alterations in cell-cycle and cell survival are critical contributing factors for the development of autoimmunity.
Collapse
|