1
|
Yang CY, Tseng YC, Tu YF, Kuo BJ, Hsu LC, Lien CI, Lin YS, Wang YT, Lu YC, Su TW, Lo YC, Lin SC. Reverse hierarchical DED assembly in the cFLIP-procaspase-8 and cFLIP-procaspase-8-FADD complexes. Nat Commun 2024; 15:8974. [PMID: 39419969 PMCID: PMC11487272 DOI: 10.1038/s41467-024-53306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
cFLIP, a master anti-apoptotic regulator, targets the FADD-induced DED complexes of procaspase-8 in death receptor and ripoptosome signaling pathways. Several tumor cells maintain relatively high levels of cFLIP in achieving their immortality. However, understanding the three-dimensional regulatory mechanism initiated or mediated by elevated levels of cFLIP has been limited by the absence of the atomic coordinates for cFLIP-induced DED complexes. Here we report the crystal plus cryo-EM structures to uncover an unconventional mechanism where cFLIP and procaspase-8 autonomously form a binary tandem DED complex, independent of FADD. This complex gains the ability to recruit FADD, thereby allosterically modulating cFLIP assembly and partially activating caspase-8 for RIPK1 cleavage. Our structure-guided mutagenesis experiments provide critical insights into these regulatory mechanisms, elucidating the resistance to apoptosis and necroptosis in achieving immortality. Finally, this research offers a unified model for the intricate bidirectional hierarchy-based processes using multiprotein helical assembly to govern cell fate decisions.
Collapse
Affiliation(s)
- Chao-Yu Yang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Chun Tseng
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Fan Tu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Bai-Jiun Kuo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Li-Chung Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
- Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Chia-I Lien
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - You-Sheng Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Yin-Ting Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yen-Chen Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tsung-Wei Su
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Su-Chang Lin
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
2
|
Yang CY, Lien CI, Tseng YC, Tu YF, Kulczyk AW, Lu YC, Wang YT, Su TW, Hsu LC, Lo YC, Lin SC. Deciphering DED assembly mechanisms in FADD-procaspase-8-cFLIP complexes regulating apoptosis. Nat Commun 2024; 15:3791. [PMID: 38710704 PMCID: PMC11074299 DOI: 10.1038/s41467-024-47990-2] [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: 02/04/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Fas-associated protein with death domain (FADD), procaspase-8, and cellular FLICE-inhibitory proteins (cFLIP) assemble through death-effector domains (DEDs), directing death receptor signaling towards cell survival or apoptosis. Understanding their three-dimensional regulatory mechanism has been limited by the absence of atomic coordinates for their ternary DED complex. By employing X-ray crystallography and cryogenic electron microscopy (cryo-EM), we present the atomic coordinates of human FADD-procaspase-8-cFLIP complexes, revealing structural insights into these critical interactions. These structures illustrate how FADD and cFLIP orchestrate the assembly of caspase-8-containing complexes and offer mechanistic explanations for their role in promoting or inhibiting apoptotic and necroptotic signaling. A helical procaspase-8-cFLIP hetero-double layer in the complex appears to promote limited caspase-8 activation for cell survival. Our structure-guided mutagenesis supports the role of the triple-FADD complex in caspase-8 activation and in regulating receptor-interacting protein kinase 1 (RIPK1). These results propose a unified mechanism for DED assembly and procaspase-8 activation in the regulation of apoptotic and necroptotic signaling across various cellular pathways involved in development, innate immunity, and disease.
Collapse
Grants
- AS-TP-107-L16, AS-TP-107-L16-1, AS-102-TP-B14 and AS-102-TP-B14-2 Academia Sinica
- AS-TP-107-L16-2 and AS-102-TP-B14-1 Academia Sinica
- AS-TP-107-L16-3 Academia Sinica
- MoST 107-2320-B-001-018-, 108-2311-B-001-018-, 109-2311-B-001-016-, and 110-2311-B-001-015- Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MoST 107-2320-B-006-062-MY3, and 111-2311-B-006-005-MY3 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MoST 108-2320-B-002-020-MY3, 111-2320-B-002-048-MY3, and 112-2326-B-002-007- Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
Collapse
Affiliation(s)
- Chao-Yu Yang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Chia-I Lien
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan
| | - Yi-Chun Tseng
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Fan Tu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Arkadiusz W Kulczyk
- Institute for Quantitative Biomedicine, Rutgers University, Department of Biochemistry and Microbiology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Yen-Chen Lu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yin-Ting Wang
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Tsung-Wei Su
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan
| | - Li-Chung Hsu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan.
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10002, Taiwan.
| | - Yu-Chih Lo
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan.
| | - Su-Chang Lin
- Genomics Research Center, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
3
|
Boccellato C, Rehm M. TRAIL-induced apoptosis and proteasomal activity - Mechanisms, signalling and interplay. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119688. [PMID: 38368955 DOI: 10.1016/j.bbamcr.2024.119688] [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: 11/30/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/20/2024]
Abstract
Programmed cell death, in particular apoptosis, is essential during development and tissue homeostasis, and also is the primary strategy to induce cancer cell death by cytotoxic therapies. Precision therapeutics targeting TRAIL death receptors are being evaluated as novel anti-cancer agents, while in parallel highly specific proteasome inhibitors have gained approval as drugs. TRAIL-dependent signalling and proteasomal control of cellular proteostasis are intricate processes, and their interplay can be exploited to enhance therapeutic killing of cancer cells in combination therapies. This review provides detailed insights into the complex signalling of TRAIL-induced pathways and the activities of the proteasome. It explores their core mechanisms of action, pharmaceutical druggability, and describes how their interplay can be strategically leveraged to enhance cell death responses in cancer cells. Offering this comprehensive and timely overview will allow to navigate the complexity of the processes governing cell death mechanisms in TRAIL- and proteasome inhibitor-based treatment conditions.
Collapse
Affiliation(s)
- Chiara Boccellato
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany.
| | - Markus Rehm
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany; University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart 70569, Germany.
| |
Collapse
|
4
|
Fang X, Zhu Y, Zhang T, Li Q, Fan L, Li X, Jiang D, Lin J, Zou L, Ren J, Huang Z, Ye H, Liu Y. Fucoxanthin Inactivates the PI3K/Akt Signaling Pathway to Mediate Malignant Biological Behaviors of Non-Small Cell Lung Cancer. Nutr Cancer 2022; 74:3747-3760. [PMID: 35838029 DOI: 10.1080/01635581.2022.2091149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although lung cancer treatment strategies have improved in recent years, the 5-year overall survival of non-small cell lung cancer (NSCLC) remains less than 15%. Chemotherapy is considered the most promising option in the comprehensive treatment of NSCLC. Fucoxanthin (FX) is a natural product derived from brown algae and has extensive applications in medicine. Previous studies reported that FX effectively inhibits the growth of NSCLC cells in vitro and in vivo. However, the mechanism underlying the anti-NSCLC effect of FX remains unknown. In this study, NSCLC cell lines and a xenograft nude mouse model were used to examine the anti-NSCLC activities of FX in vitro and in vivo. Network pharmacology analysis and inhibitors or activators of the PI3K/Akt signaling pathway were used to explore the anti-NSCLC mechanisms of FX. The results indicated that FX could inhibit proliferation, migration, and invasion, arrest cell cycle at the G0/G1 phase, and induce apoptosis of NSCLC cells in vitro. Additionally, FX suppressed tumor growth in vivo. The PI3K/Akt signaling pathway was found to be involved in the anti-NSCLC activity of FX. In conclusion, FX inhibits malignant biological behaviors of NSCLC by suppressing the phosphorylation of both PI3K and AKT, and subsequently inactivating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xuehong Fang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Yuzhen Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Taomin Zhang
- Department of Pharmacology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Qian Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Lvhua Fan
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Xiaodan Li
- People's Hospital of Longhua District, Shenzhen, Guangdong, China
| | - Daishun Jiang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Jie Lin
- Department of Pharmacology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Liyi Zou
- Department of Pharmacology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jianwei Ren
- Shenzhen Ritzcon Biological Technology Co., LTD, Shenzhen, Guangdong, China
| | - Zunnan Huang
- Department of Pharmacology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Hua Ye
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| | - Yi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Biomedical Research Institute, Department of Pharmacology, Guangdong Medical University, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, China
| |
Collapse
|
5
|
Proteasome inhibition triggers the formation of TRAIL receptor 2 platforms for caspase-8 activation that accumulate in the cytosol. Cell Death Differ 2021; 29:147-155. [PMID: 34354257 PMCID: PMC8738721 DOI: 10.1038/s41418-021-00843-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 11/24/2022] Open
Abstract
Cancer cells that are resistant to Bax/Bak-dependent intrinsic apoptosis can be eliminated by proteasome inhibition. Here, we show that proteasome inhibition induces the formation of high molecular weight platforms in the cytosol that serve to activate caspase-8. The activation complexes contain Fas-associated death domain (FADD) and receptor-interacting serine/threonine-protein kinase 1 (RIPK1). Furthermore, the complexes contain TRAIL-receptor 2 (TRAIL-R2) but not TRAIL-receptor 1 (TRAIL-R1). While RIPK1 inhibition or depletion did not affect proteasome inhibitor-induced cell death, TRAIL-R2 was found essential for efficient caspase-8 activation, since the loss of TRAIL-R2 expression abrogated caspase processing, significantly reduced cell death, and promoted cell re-growth after drug washout. Overall, our study provides novel insight into the mechanisms by which proteasome inhibition eliminates otherwise apoptosis-resistant cells, and highlights the crucial role of a ligand-independent but TRAIL-R2-dependent activation mechanism for caspase-8 in this scenario.
Collapse
|
6
|
Xiao H, Zhao J, Fang C, Cao Q, Xing M, Li X, Hou J, Ji A, Song S. Advances in Studies on the Pharmacological Activities of Fucoxanthin. Mar Drugs 2020; 18:E634. [PMID: 33322296 PMCID: PMC7763821 DOI: 10.3390/md18120634] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
Fucoxanthin is a natural carotenoid derived mostly from many species of marine brown algae. It is characterized by small molecular weight, is chemically active, can be easily oxidized, and has diverse biological activities, thus protecting cell components from ROS. Fucoxanthin inhibits the proliferation of a variety of cancer cells, promotes weight loss, acts as an antioxidant and anti-inflammatory agent, interacts with the intestinal flora to protect intestinal health, prevents organ fibrosis, and exerts a multitude of other beneficial effects. Thus, fucoxanthin has a wide range of applications and broad prospects. This review focuses primarily on the latest progress in research on its pharmacological activity and underlying mechanisms.
Collapse
Affiliation(s)
- Han Xiao
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Jiarui Zhao
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Chang Fang
- Test Center for Agri‐Products Quality of Jinan, Jinan 250316, China;
| | - Qi Cao
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Maochen Xing
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Xia Li
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Junfeng Hou
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| | - Aiguo Ji
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shuliang Song
- Marine College, Shandong University, Weihai 264209, China; (H.X.); (J.Z.); (Q.C.); (M.X.); (X.L.); (J.H.)
| |
Collapse
|
7
|
Datta A, Sarmah D, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Advances in Studies on Stroke-Induced Secondary Neurodegeneration (SND) and Its Treatment. Curr Top Med Chem 2020; 20:1154-1168. [DOI: 10.2174/1568026620666200416090820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/23/2022]
Abstract
Background:
The occurrence of secondary neurodegeneration has exclusively been observed
after the first incidence of stroke. In humans and rodents, post-stroke secondary neurodegeneration
(SND) is an inevitable event that can lead to progressive neuronal loss at a region distant to initial infarct.
SND can lead to cognitive and motor function impairment, finally causing dementia. The exact
pathophysiology of the event is yet to be explored. It is seen that the thalami, in particular, are susceptible
to cause SND. The reason behind this is because the thalamus functioning as the relay center and is
positioned as an interlocked structure with direct synaptic signaling connection with the cortex. As SND
proceeds, accumulation of misfolded proteins and microglial activation are seen in the thalamus. This
leads to increased neuronal loss and worsening of functional and cognitive impairment.
Objective:
There is a necessity of specific interventions to prevent post-stroke SND, which are not properly
investigated to date owing to sparsely reproducible pre-clinical and clinical data. The basis of this
review is to investigate about post-stroke SND and its updated treatment approaches carefully.
Methods:
Our article presents a detailed survey of advances in studies on stroke-induced secondary neurodegeneration
(SND) and its treatment.
Results:
This article aims to put forward the pathophysiology of SND. We have also tabulated the latest
treatment approaches along with different neuroimaging systems that will be helpful for future reference
to explore.
Conclusion:
In this article, we have reviewed the available reports on SND pathophysiology, detection
techniques, and possible treatment modalities that have not been attempted to date.
Collapse
Affiliation(s)
- Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kunjan R. Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Dileep R. Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
8
|
Xiao Z, Chen S, Feng S, Li Y, Zou J, Ling H, Zeng Y, Zeng X. Function and mechanisms of microRNA-20a in colorectal cancer. Exp Ther Med 2020; 19:1605-1616. [PMID: 32104211 PMCID: PMC7027132 DOI: 10.3892/etm.2020.8432] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy and the second leading cause of cancer-associated mortality worldwide. CRC currently has no specific biomarkers to promote its diagnosis and treatment and the underlying mechanisms regulating its pathogenesis have not yet been determined. MicroRNAs (miRs) are small, non-coding RNAs that exhibit regulatory functions and have been demonstrated to serve a crucial role in the post-transcriptional regulatory processes of gene expression that is associated with cell physiology and disease progression. Recently, abnormal miR-20a expression has been identified in a number of cancers types and this has become a novel focus within cancer research. High levels of miR-20a expression have been identified in CRC tissues, serum and plasma. In a recent study, miR-20a was indicated to be present in feces and to exhibit a high sensitivity to CRC. Therefore, miR-20a may be used as a marker for CRC and an indicator that can prevent the invasive examination of patients with this disease. Changes in the expression of miR-20a during chemotherapy can be used as a biomarker for monitoring resistance to treatment. In conclusion, miR-20a exhibits the potential for clinical application as a novel diagnostic biomarker and therapeutic target for use in patients with CRC. The present study focused on the role and mechanisms of miR-20a in CRC.
Collapse
Affiliation(s)
- Zheng Xiao
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shi Chen
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shujun Feng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yukun Li
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui Ling
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ying Zeng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,School of Nursing, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xi Zeng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
9
|
Abstract
OBJECTIVE Identify the molecular mechanism of inflammatory stimuli induced pancreatic cancer progression. METHODS RNA-seq, microarray assay and bioinformatics analyses were used to identify differentially expressed genes. Immunohistochemical staining was performed to evaluate CD68, CD163, β-catenin, CD103, CCL3 markers. Quantitative real-time polymerase chain reaction (qRT-PCR), luciferase reporter assay, apoptosis assay, wound healing assay and immunofluorescence were performed to study the relationship of inflammatory stimuli and WNT/β-catenin pathway. RESULTS Differentially expressed genes of macrophage-conditioned medium-treated pancreatic cancer cells were related with WNT/β-catenin pathway. Inflammatory stimuli could activate WNT/β-catenin signaling pathway. In 106 pancreatic cancer patients, nuclear β-catenin expression of CD68-high group was much higher than CD68-low group (P < 0.05), as same as CD163 (P < 0.05). Inflammatory stimuli downregulated the expression of CCL3 via WNT/β-catenin pathway and inhibited the chemotaxis of CD103 dendritic cells. Six pancreatic cancer prognosis associating genes were upregulated by inflammatory stimuli via WNT/β-catenin pathway. Transforming growth factor-β promoted malignant biological behavior of pancreatic cancer cells through WNT/β-catenin pathway-dependent mechanism. CONCLUSIONS Our present study provided a novel mechanism involved in the inflammation-driven cancer progression through tumor immune escape and downstream gene regulation of WNT/β-catenin pathway-dependent manner.
Collapse
|
10
|
Zhang R, Xu J, Zhao J, Bai J. Knockdown of miR-27a sensitizes colorectal cancer stem cells to TRAIL by promoting the formation of Apaf-1-caspase-9 complex. Oncotarget 2018; 8:45213-45223. [PMID: 28423356 PMCID: PMC5542179 DOI: 10.18632/oncotarget.16779] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/22/2017] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs have been proved to participate in multiple biological processes in cancers. For developing resistance to cytotoxic drug, cancer cells, especially the cancer stem cells, usually change their microRNA expression profile to survive in hostile environments. In the present study, we found that expression of microRNA-27a was increased in colorectal cancer stem cells. High level of microRNA-27a was indicated to induce the resistance to TNF-related apoptosis-inducing ligand (TRAIL). Knockdown of microRNA-27a resensitized colorectal cancer stem cells to TRAIL-induced cell death. Mechanically, the gene of Apaf-1, which is associated with the mitochondrial apoptosis, was demonstrated to be the target of microRNA-27a in colorectal cancer stem cells. Knockdown of microRNA-27a increased the expression level of Apaf-1, thus enhancing the formation of Apaf-1-caspase-9 complex and subsequently promoting the TRAIL-induced apoptosis in colorectal cancer stem cells. These findings suggested that knockdown of microRNA-27a in colorectal cancer stem cells by the specific antioligonucleotides was potential to reverse the chemoresistance to TRAIL. It may represent a novel therapeutic strategy for treating the colorectal cancer more effectively.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang 110042, China
| | - Jian Xu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang 110042, China
| | - Jian Zhao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang 110042, China
| | - Jinghui Bai
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang 110042, China
| |
Collapse
|
11
|
Xu C, Shi L, Chen W, Fang P, Li J, Jin L, Pan Z, Pan C. MiR-106b inhibitors sensitize TRAIL-induced apoptosis in hepatocellular carcinoma through increase of death receptor 4. Oncotarget 2018; 8:41921-41931. [PMID: 28410209 PMCID: PMC5522038 DOI: 10.18632/oncotarget.16707] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/20/2017] [Indexed: 12/21/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), which is a member of the TNF superfamily, can induce tumor cell apoptosis. However, multiple types of tumor, including hepatocellular carcinoma, show tolerance to TRAIL. Previous studies have demonstrated that tumor cells usually change their expression profile of microRNA (miRNA) to obtain the ability of tolerance to drugs. However, whether such change of miRNA on TRAIL sensitivity is seen in hepatocellular carcinoma still needs to be explored. In this study, we observed overexpression of miR-106b in both HCC patients’ tumor tissues and cell lines. Furthermore, we found that overexpression of miR-106b is associated with the sensitivity of TRAIL to HCC. Silencing of miR-106b with antisense oligonucleotide (anti-miR-106b) is proved to enhance the TRAIL-induced apoptosis and reduce the acquired drug resistance to TRAIL in HCC. Mechanically, we didn't observe the obvious change of pro-apoptotic proteins (Bax and Bid) and anti-apoptotic proteins (Bcl-2, Mcl-1 and Bcl-xl) after treatment of anti-miR-106b. However, we used the methods of bioinformatics, flow cytometry, cellular and molecular methods to prove that miR-106b directly targeted to death receptor 4 (DR4) 3′-UTR (3′-Untranslated Regions). MiR-106b inhibitors induced increase of DR4 expression and therefore enhancing TRAIL-mediated apoptosis in HCC. In summary, these results suggest the application of miR-106b inhibitors in HCC treatment. Combination with miR-106b inhibitors and TRAIL may be a novel clinical treatment method on HCC treatment in the future.
Collapse
Affiliation(s)
- Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Liang Shi
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Weilai Chen
- Department of Neurology, Wenzhou People's Hospital, Wenzhou 325027, China
| | - Peipei Fang
- Department of Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- Department of Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Lingxiang Jin
- Department of Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenzhen Pan
- Department of Infectious Disease, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chenwei Pan
- Department of Infectious Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
12
|
Predicting the cell death responsiveness and sensitization of glioma cells to TRAIL and temozolomide. Oncotarget 2018; 7:61295-61311. [PMID: 27494880 PMCID: PMC5308652 DOI: 10.18632/oncotarget.10973] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 07/18/2016] [Indexed: 12/28/2022] Open
Abstract
Genotoxic chemotherapy with temozolomide (TMZ) is a mainstay of treatment for glioblastoma (GBM); however, at best, TMZ provides only modest survival benefit to a subset of patients. Recent insight into the heterogeneous nature of GBM suggests a more personalized approach to treatment may be necessary to overcome cancer drug resistance and improve patient care. These include novel therapies that can be used both alone and with TMZ to selectively reactivate apoptosis within malignant cells. For this approach to work, reliable molecular signatures that can accurately predict treatment responsiveness need to be identified first. Here, we describe the first proof-of-principle study that merges quantitative protein-based analysis of apoptosis signaling networks with data- and knowledge-driven mathematical systems modeling to predict treatment responsiveness of GBM cell lines to various apoptosis-inducing stimuli. These include monotherapies with TMZ and TRAIL, which activate the intrinsic and extrinsic apoptosis pathways, respectively, as well as combination therapies of TMZ+TRAIL. We also successfully employed this approach to predict whether individual GBM cell lines could be sensitized to TMZ or TRAIL via the selective targeting of Bcl-2/Bcl-xL proteins with ABT-737. Our findings suggest that systems biology-based approaches could assist in personalizing treatment decisions in GBM to optimize cell death induction.
Collapse
|
13
|
Jin Y, Qiu S, Shao N, Zheng J. Fucoxanthin and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Synergistically Promotes Apoptosis of Human Cervical Cancer Cells by Targeting PI3K/Akt/NF-κB Signaling Pathway. Med Sci Monit 2018; 24:11-18. [PMID: 29291370 PMCID: PMC5759513 DOI: 10.12659/msm.905360] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Fucoxanthin is a carotenoid present in the chloroplasts of brown seaweeds. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine that selectively induces apoptosis in many tumor cells and is an attractive candidate for antitumor therapies. Material/Methods After human cervical cancer cell lines HeLa, SiHa, and CaSki were treated with fucoxanthin or TRAIL. Cell viability was determined by 2,3-bis (2-methoxy-4-nitro-5-sulfophenyl)-2-tetrazolium 5-carboxanilide (XTT) method. Apoptosis was measured by flow cytometry (FCM). Protein expression of phosphatidylinositol 3 kinase (PI3K), protein kinase B (Akt), phosphated Akt (p-Akt), NF-κB nuclear factor-k-gene binding (NF-κB). Phosphated nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (p-IκBa), was measured by Western blot analysis. mRNA expression of Bax and Bcl2 was measured by RNA preparation and quantitative reverse transcription polymerase chain reaction (RT-PCR). Results In the present study, the effectiveness in terms of apoptosis was as follows: TRAIL plus fucoxanthin>fucoxanthin>TRAIL, indicating the combination of fucoxanthin and TRAIL, produced a strong synergistic effect on apoptosis in human cervical cancer cells. Additionally, we found that upstream signaling PI3K/Akt and NF-κB pathways-mediated cell apoptosis was activated by TRAIL and suppressed by fucoxanthin. By using PI3K and NF-κB inhibitors LY49002 and PDTC, we found that fucoxanthin- or TRAIL-induced apoptosis of human cervical cancer cells was obviously down-regulated. Conclusions Taken together, these findings suggest that fucoxanthin and TRAIL increased the apoptosis in human cervical cancer cells by targeting the PI3K/Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ye Jin
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Shuang Qiu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Na Shao
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Jianhua Zheng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
14
|
MiR-126 reverses drug resistance to TRAIL through inhibiting the expression of c-FLIP in cervical cancer. Gene 2017; 627:420-427. [DOI: 10.1016/j.gene.2017.06.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/19/2017] [Accepted: 06/28/2017] [Indexed: 01/20/2023]
|
15
|
miR-20a-directed regulation of BID is associated with the TRAIL sensitivity in colorectal cancer. Oncol Rep 2016; 37:571-578. [DOI: 10.3892/or.2016.5278] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/31/2016] [Indexed: 11/05/2022] Open
|
16
|
Sun X, Li Y, Zheng M, Zuo W, Zheng W. MicroRNA-223 Increases the Sensitivity of Triple-Negative Breast Cancer Stem Cells to TRAIL-Induced Apoptosis by Targeting HAX-1. PLoS One 2016; 11:e0162754. [PMID: 27618431 PMCID: PMC5019415 DOI: 10.1371/journal.pone.0162754] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/26/2016] [Indexed: 01/13/2023] Open
Abstract
Drug resistance remains a significant challenge in the treatment of triple-negative breast cancer (TNBC). Recent studies have demonstrated that this drug resistance is associated with a group of cells known as cancer stem cells (CSCs), which are believed to determine the sensitivity of tumor cells to cancer treatment. MicroRNAs (miRNAs) are small, non-coding RNAs that play significant roles in normal and cancer cells. MiR-223 reportedly acts as a tumor suppressor in a range of cancers. However, the role of miR-223 in TNBC, especially in triple-negative breast cancer stem cells (TNBCSCs), remains unknown. Here, we found that miR-223 expression was down-regulated in CD44+CD24-/low TNBCSCs compared with non-CSCs. Furthermore, we found that miR-223 overexpression resensitized TNBCSCs to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. The HAX-1 gene, which is located in the mitochondria and functions as an anti-apoptotic protein, was found to be directly regulated by miR-223 in MDA-MB-231 cells. We demonstrated that miR-223 overexpression promoted TRAIL-induced apoptosis through the mitochondria/ROS pathway. In conclusion, our results suggest that miR-223 increases the sensitivity of TNBCSCs to TRAIL-induced apoptosis by targeting HAX-1. Our findings have improved our understanding of the role of miR-223 in TNBC and may contribute to TNBC treatment.
Collapse
Affiliation(s)
- Xu Sun
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yongqing Li
- Breast Cancer Center, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Meizhu Zheng
- Breast Cancer Center, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Shandong Academy of Medical Sciences, Jinan, 250117, China
- * E-mail:
| | - Wenshu Zuo
- Breast Cancer Center, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wenzhu Zheng
- Emergency Medicine, Jinan Lixia District People's Hospital, Jinan, 250000, China
| |
Collapse
|
17
|
Matsumoto Y, Miyamoto Y, Cabral H, Matsumoto Y, Nagasaka K, Nakagawa S, Yano T, Maeda D, Oda K, Kawana K, Nishiyama N, Kataoka K, Fujii T. Enhanced efficacy against cervical carcinomas through polymeric micelles physically incorporating the proteasome inhibitor MG132. Cancer Sci 2016; 107:773-81. [PMID: 26987571 PMCID: PMC4968607 DOI: 10.1111/cas.12926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 03/02/2016] [Accepted: 03/08/2016] [Indexed: 12/04/2022] Open
Abstract
Treatment of recurrent or advanced cervical cancer is still limited, and new therapeutic choices are needed for improving prognosis and quality of life of patients. Because human papilloma virus (HPV) infection is critical in cervical carcinogenesis, with the E6 and E7 oncogenes of HPV degrading tumor suppressor proteins through the ubiquitin proteasome system, the inhibition of the ubiquitin proteasome system appears to be an ideal target to suppress the growth of cervical tumors. Herein, we focused on the ubiquitin proteasome inhibitor MG132 (carbobenzoxy‐Leu‐Leu‐leucinal) as an anticancer agent against cervical cancer cells, and physically incorporated it into micellar nanomedicines for achieving selective delivery to solid tumors and improving its in vivo efficacy. These MG132‐loaded polymeric micelles (MG132/m) showed strong tumor inhibitory in vivo effect against HPV‐positive tumors from HeLa and CaSki cells, and even in HPV‐negative tumors from C33A cells. Repeated injection of MG132/m showed no significant toxicity to mice under analysis by weight change or histopathology. Moreover, the tumors treated with MG132/m showed higher levels of tumor suppressing proteins, hScrib and p53, as well as apoptotic degree, than tumors treated with free MG132. This enhanced efficacy of MG132/m was attributed to their prolonged circulation in the bloodstream, which allowed their gradual extravasation and penetration within the tumor tissue, as determined by intravital microscopy. These results support the use of MG132 incorporated into polymeric micelles as a safe and effective therapeutic strategy against cervical tumors.
Collapse
Affiliation(s)
- Yoko Matsumoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichiro Miyamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yu Matsumoto
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazunori Nagasaka
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Nakagawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Teikyo, Tokyo, Japan
| | - Tetsu Yano
- Department of Obstetrics and Gynecology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Daichi Maeda
- Department of Cellular and Organ Pathology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Nishiyama
- Polymer Chemistry Division, Chemical Resources Laboratory, Tokyo Institute of Technology, Yokohama, Japan
| | - Kazunori Kataoka
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,Innovation Center of Nanomedicine, Kawasaki Institute of Industry Promotion, Kawasaki, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Qureshi MZ, Romero MA, Attar R, Javed Z, Farooqi AA. TRAIL and Bortezomib: killing cancer with two stones. Asian Pac J Cancer Prev 2015; 16:1671-4. [PMID: 25743849 DOI: 10.7314/apjcp.2015.16.4.1671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cancer genomics and proteomics have undergone considerable broadening in the past decades and increasingly it is being realized that solid/liquid phase microarrays and high-throughput resequencing have provided platforms to improve our existing knowledge of determinants of cancer development, progression and survival. Loss of apoptosis is a widely and deeply studied process and different approaches are being used to restore apoptosis in resistant cancer phenotype. Modulating the balance between pro-apoptotic and anti-apoptotic proteins is essential to induce apoptosis. It is becoming more understood that pharmacological inhibition of the proteasome might prove to be an effective option in improving TRAIL induced apoptosis in cancer cells. Keeping in view rapidly accumulating evidence of carcinogenesis, metastasis, resistance against wide ranging therapeutics and loss of apoptosis, better knowledge regarding tumor suppressors, oncogenes, pro-apoptotic and anti-apotptic proteins will be helpful in translating the findings from benchtop to bedside.
Collapse
|
19
|
Partial equilibrium approximations in apoptosis. II. The death-inducing signaling complex subsystem. Math Biosci 2015; 270:126-34. [DOI: 10.1016/j.mbs.2015.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 10/01/2015] [Accepted: 10/16/2015] [Indexed: 12/28/2022]
|
20
|
Zhang Y, Huang F, Wang J, Luo H, Wang Z. 2-DG-Regulated RIP and c-FLIP Effect on Liver Cancer Cell Apoptosis Induced by TRAIL. Med Sci Monit 2015; 21:3442-8. [PMID: 26552967 PMCID: PMC4646230 DOI: 10.12659/msm.895034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Cancer cells survival depends on glucose metabolism and ATP. Inhibiting glucose metabolism is a possible anticancer treatment. The phosphorylation of 2-deoxy-D-glucose (2-DG), which is a glycogen analogue, seriously affects the normal glycometabolism phosphorylation process, leading to ATP consumption. Studies showed that 2-DG could regulate RIP and c-FLIP. This paper aimed to investigate the effect of 2-DG on RIP and c-FLIP expression in HepG2 and Hep3B cells, further illustrating the effect and mechanism of 2-DG regulating RIP and c-FLIP expression on liver cancer cell apoptosis induced by TRAIL. Material/Methods RIP and c-FLIP gene silencing HepG2 and Hep3B cell models were established by siRNA and detected by Western blot. Cell viability was determined by MTT and apoptosis rate was measured by flow cytometry. JC-1 fluorescent probe was used to test mitochondrial membrane potential. Results 2-DG or TRAIL alone significantly reduced HepG2 and Hep3B cell survival rate and promoted apoptosis. Compared with the single TRAIL treatment group, the combination of 2-DG and TRAIL could reduce cell survival rate, increase apoptosis rate, and decease mitochondrial membrane potential, which is dependent on Caspases. 2-DG can inhibit RIP and c-FLIP expression, leading to increased TRAIL-induced HepG2 and Hep3B cells apoptosis. Conclusions 2-DG can down-regulate RIP and c-FLIP expression, and change Caspases activities to increase the liver cancer cell apoptosis induced by TRAIL.
Collapse
Affiliation(s)
- Yuping Zhang
- Department of Normal Surgical, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Feizhou Huang
- Department of Normal Surgical, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Jian Wang
- Department of Normal Surgical, Genetics Research Room of Central South University, Changsha, Hunan, China (mainland)
| | - Hongwu Luo
- Department of Normal Surgical, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| | - Zhichao Wang
- Department of Normal Surgical, The 3rd Xiangya Hospital of Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
21
|
Sun J, Luo H, Nie W, Xu X, Miao X, Huang F, Wu H, Jin X. Protective effect of RIP and c-FLIP in preventing liver cancer cell apoptosis induced by TRAIL. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:6519-6525. [PMID: 26261530 PMCID: PMC4525864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/25/2015] [Indexed: 06/04/2023]
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) is a member of the tumor necrosis factor superfamily that can induce tumor selective death by up-regulating death receptor 4 (DR4) and DR5 expression. The study aimed to explore the role of RIP and c-FLIP genes in TRAIL induced liver cancer cell HepG2 and Hep3B apoptosis and related mechanism. RIP and c-FLIP silenced HepG2 and Hep3B cell model were established through siRNA. Western blot was applied to test c-FLIP, RIP, DR4, DR5, FADD, Caspase-3/8/9, ERK1/2, and DFF45 protein expression. Caspase-8 kit was used to detect Caspase-8 expression. Flow cytometry was performed to measure cell apoptosis rate. Acid phosphatase method was applied to determine cell cycle. TRAIL had no significant effect on Caspase-3/8/9, DR4, DR5, ERK1/2, and DFF45 protein expression, but up-regulated c-FLIP and RIP protein expression and reduced FADD expression level. After treated by the chemotherapy drug mitomycin and adriamycin, c-FLIP and RIP expression decreased significantly, while FADD increased. After knockout c-FLIP and RIP gene, HepG2 and Hep3B cell apoptosis rate induced by TRAIL increased obviously. Meanwhile, cell subG1 percentage increased markedly and exhibited G1 phase growth retardation. In addition, after two kinds of gene knockout, Caspase-8 was activated and produce Caspase-3 P20 and P24, leading DFF45 appeared DNA fragment P17 and P25. c-FLIP and RIP can inhibit Caspase-8 activation and prompting HepG2 and Hep3B resistant to cell apoptosis induced by TRAIL.
Collapse
Affiliation(s)
- Jichun Sun
- Department of Surgery, The 3rd Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Hongwu Luo
- Department of Surgery, The 3rd Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Wanpin Nie
- Department of Surgery, The 3rd Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Xundi Xu
- Department of Surgery, The Second Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Xiongying Miao
- Department of Surgery, The Second Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Feizhou Huang
- Department of Surgery, The 3rd Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Haiyan Wu
- Department of Surgery, The 3rd Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| | - Xiaoxin Jin
- Department of Surgery, The Second Xiangya Hospital, Central South UniversityChangsha, Hunan Province, China
| |
Collapse
|
22
|
Roux J, Hafner M, Bandara S, Sims JJ, Hudson H, Chai D, Sorger PK. Fractional killing arises from cell-to-cell variability in overcoming a caspase activity threshold. Mol Syst Biol 2015; 11:803. [PMID: 25953765 PMCID: PMC4461398 DOI: 10.15252/msb.20145584] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
When cells are exposed to death ligands such as TRAIL, a fraction undergoes apoptosis and a fraction survives; if surviving cells are re-exposed to TRAIL, fractional killing is once again observed. Therapeutic antibodies directed against TRAIL receptors also cause fractional killing, even at saturating concentrations, limiting their effectiveness. Fractional killing arises from cell-to-cell fluctuations in protein levels (extrinsic noise), but how this results in a clean bifurcation between life and death remains unclear. In this paper, we identify a threshold in the rate and timing of initiator caspase activation that distinguishes cells that live from those that die; by mapping this threshold, we can predict fractional killing of cells exposed to natural and synthetic agonists alone or in combination with sensitizing drugs such as bortezomib. A phenomenological model of the threshold also quantifies the contributions of two resistance genes (c-FLIP and Bcl-2), providing new insight into the control of cell fate by opposing pro-death and pro-survival proteins and suggesting new criteria for evaluating the efficacy of therapeutic TRAIL receptor agonists.
Collapse
Affiliation(s)
- Jérémie Roux
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Marc Hafner
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Samuel Bandara
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Joshua J Sims
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Diana Chai
- Merrimack Pharmaceuticals, Cambridge, MA, USA
| | - Peter K Sorger
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Rehm M, Parsons MJ, Bouchier-Hayes L. Measuring caspase activity by Förster resonance energy transfer. Cold Spring Harb Protoc 2015; 2015:pdb.prot082560. [PMID: 25561624 DOI: 10.1101/pdb.prot082560] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Förster resonance energy transfer (FRET) occurs across very short distances (in the nanometer range) between donor and acceptor fluorophores that overlap in their emission and absorption spectra. FRET-compatible green fluorescent protein (GFP) variants that are fused to short peptide linkers containing caspase cleavage sites can be used to measure caspase activity. In the intact probes, the donor and acceptor fluorophores are in close proximity, and FRET is highly efficient. On caspase activation, proteolysis of the linker occurs, and the donor is separated from the acceptor. This results in a disruption of resonance energy transfer and an increase in donor fluorescence quantum yield; this event is typically referred to as sensitized emission or donor unquenching. A number of highly sensitive FRET probes based on the cyan fluorescent protein-yellow fluorescent protein (CFP-YFP) pair, or improved variants thereof, have been developed to detect intracellular caspase activities. In this protocol we describe how to use FRET-based caspase substrates and time-lapse imaging to measure caspase activity in cells undergoing apoptosis.
Collapse
Affiliation(s)
- Markus Rehm
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Melissa J Parsons
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics-Hematology, Baylor College of Medicine, Houston, Texas 77030
| | - Lisa Bouchier-Hayes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics-Hematology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
24
|
Bertaux F, Stoma S, Drasdo D, Batt G. Modeling dynamics of cell-to-cell variability in TRAIL-induced apoptosis explains fractional killing and predicts reversible resistance. PLoS Comput Biol 2014; 10:e1003893. [PMID: 25340343 PMCID: PMC4207462 DOI: 10.1371/journal.pcbi.1003893] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/04/2014] [Indexed: 12/22/2022] Open
Abstract
Isogenic cells sensing identical external signals can take markedly different decisions. Such decisions often correlate with pre-existing cell-to-cell differences in protein levels. When not neglected in signal transduction models, these differences are accounted for in a static manner, by assuming randomly distributed initial protein levels. However, this approach ignores the a priori non-trivial interplay between signal transduction and the source of this cell-to-cell variability: temporal fluctuations of protein levels in individual cells, driven by noisy synthesis and degradation. Thus, modeling protein fluctuations, rather than their consequences on the initial population heterogeneity, would set the quantitative analysis of signal transduction on firmer grounds. Adopting this dynamical view on cell-to-cell differences amounts to recast extrinsic variability into intrinsic noise. Here, we propose a generic approach to merge, in a systematic and principled manner, signal transduction models with stochastic protein turnover models. When applied to an established kinetic model of TRAIL-induced apoptosis, our approach markedly increased model prediction capabilities. One obtains a mechanistic explanation of yet-unexplained observations on fractional killing and non-trivial robust predictions of the temporal evolution of cell resistance to TRAIL in HeLa cells. Our results provide an alternative explanation to survival via induction of survival pathways since no TRAIL-induced regulations are needed and suggest that short-lived anti-apoptotic protein Mcl1 exhibit large and rare fluctuations. More generally, our results highlight the importance of accounting for stochastic protein turnover to quantitatively understand signal transduction over extended durations, and imply that fluctuations of short-lived proteins deserve particular attention.
Collapse
Affiliation(s)
| | | | - Dirk Drasdo
- INRIA Paris-Rocquencourt, Le Chesnay, France
- Laboratoire Jacques-Louis Lions (LJLL), University of Paris 6 (UPMC) - CNRS (UMR7598), Paris, France
| | - Gregory Batt
- INRIA Paris-Rocquencourt, Le Chesnay, France
- * E-mail:
| |
Collapse
|
25
|
Lavrik IN. Systems biology of death receptor networks: live and let die. Cell Death Dis 2014; 5:e1259. [PMID: 24874731 PMCID: PMC4047881 DOI: 10.1038/cddis.2014.160] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/11/2014] [Accepted: 03/13/2014] [Indexed: 12/21/2022]
Abstract
The extrinsic apoptotic pathway is initiated by death receptor activation. Death receptor activation leads to the formation of death receptor signaling platforms, resulting in the demolition of the cell. Despite the fact that death receptor-mediated apoptosis has been studied to a high level of detail, its quantitative regulation until recently has been poorly understood. This situation has dramatically changed in the last years. Creation of mathematical models of death receptor signaling led to an enormous progress in the quantitative understanding of the network regulation and provided fascinating insights into the mechanisms of apoptosis control. In the following sections, the models of the death receptor signaling and their biological implications will be addressed. Central attention will be given to the models of CD95/Fas/APO-1, an exemplified member of the death receptor signaling pathways. The CD95 death-inducing signaling complex (DISC) and regulation of CD95 DISC activity by its key inhibitor c-FLIP, have been vigorously investigated by modeling approaches, and therefore will be the major topic here. Furthermore, the non-linear dynamics of the DISC, positive feedback loops and bistability as well as stoichiometric switches in extrinsic apoptosis will be discussed. Collectively, this review gives a comprehensive view how the mathematical modeling supported by quantitative experimental approaches has provided a new understanding of the death receptor signaling network.
Collapse
Affiliation(s)
- I N Lavrik
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
- Faculty of Fundamental Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany. Tel: +49 3916724767; Fax: +49 3916724769; E-mail:
| |
Collapse
|
26
|
Huber HJ, McKiernan RG, Prehn JHM. Harnessing system models of cell death signalling for cytotoxic chemotherapy: towards personalised medicine approaches? J Mol Med (Berl) 2014; 92:227-37. [PMID: 24477766 DOI: 10.1007/s00109-014-1126-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 12/27/2022]
Abstract
Most cytotoxic chemotherapeutics are believed to kill cancer cells by inducing apoptosis. Understanding the factors that contribute to impairment of apoptosis in cancer cells is therefore critical for the development of novel therapies that circumvent the widespread chemoresistance. Apoptosis, however, is a complex and tightly controlled process that can be induced by different classes of chemotherapeutics targeting different signalling nodes and pathways. Moreover, apoptosis initiation and apoptosis execution strongly depend on patient-specific, genomic and proteomic signatures. Here, we will review recent translational studies that suggest a critical link between the sensitivity of cancer cells to initiate apoptosis and clinical outcome. Next we will discuss recent advances in the field of system modelling of apoptosis pathways for the prediction of treatment responses. We propose that initiation of mitochondrial apoptosis, defined as the process of mitochondrial outer membrane permeabilisation (MOMP), is a dose-dependent decision process that allows for a prediction of individual therapy responses and therapeutic windows. We provide evidence in contrast that apoptosis execution post-MOMP may be a binary decision that dictates whether apoptosis is executed or not. We will discuss the implications of this concept for the future use of novel adjuvant therapeutics that specifically target apoptosis signalling pathways or which may be used to reduce the impact of cell-to-cell heterogeneity on therapy responses. Finally, we will discuss the technical and regulatory requirements surrounding the use and implications of system-based patient stratification tools for the future of personalised oncology.
Collapse
Affiliation(s)
- Heinrich J Huber
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland,
| | | | | |
Collapse
|
27
|
Modulation of apoptosis sensitivity through the interplay with autophagic and proteasomal degradation pathways. Cell Death Dis 2014; 5:e1011. [PMID: 24457955 PMCID: PMC4040655 DOI: 10.1038/cddis.2013.520] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 11/16/2013] [Accepted: 11/21/2013] [Indexed: 12/24/2022]
Abstract
Autophagic and proteasomal degradation constitute the major cellular proteolysis pathways. Their physiological and pathophysiological adaptation and perturbation modulates the relative abundance of apoptosis-transducing proteins and thereby can positively or negatively adjust cell death susceptibility. In addition to balancing protein expression amounts, components of the autophagic and proteasomal degradation machineries directly interact with and co-regulate apoptosis signal transduction. The influence of autophagic and proteasomal activity on apoptosis susceptibility is now rapidly gaining more attention as a significant modulator of cell death signalling in the context of human health and disease. Here we present a concise and critical overview of the latest knowledge on the molecular interplay between apoptosis signalling, autophagy and proteasomal protein degradation. We highlight that these three pathways constitute an intricate signalling triangle that can govern and modulate cell fate decisions between death and survival. Owing to rapid research progress in recent years, it is now possible to provide detailed insight into the mechanisms of pathway crosstalk, common signalling nodes and the role of multi-functional proteins in co-regulating both protein degradation and cell death.
Collapse
|
28
|
Flusberg DA, Sorger PK. Modulating cell-to-cell variability and sensitivity to death ligands by co-drugging. Phys Biol 2013; 10:035002. [PMID: 23735516 DOI: 10.1088/1478-3975/10/3/035002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) holds promise as an anti-cancer therapeutic but efficiently induces apoptosis in only a subset of tumor cell lines. Moreover, even in clonal populations of responsive lines, only a fraction of cells dies in response to TRAIL and individual cells exhibit cell-to-cell variability in the timing of cell death. Fractional killing in these cell populations appears to arise not from genetic differences among cells but rather from differences in gene expression states, fluctuations in protein levels and the extent to which TRAIL-induced death or survival pathways become activated. In this study, we ask how cell-to-cell variability manifests in cell types with different sensitivities to TRAIL, as well as how it changes when cells are exposed to combinations of drugs. We show that individual cells that survive treatment with TRAIL can regenerate the sensitivity and death-time distribution of the parental population, demonstrating that fractional killing is a stable property of cell populations. We also show that cell-to-cell variability in the timing and probability of apoptosis in response to treatment can be tuned using combinations of drugs that together increase apoptotic sensitivity compared to treatment with one drug alone. In the case of TRAIL, modulation of cell-to-cell variability by co-drugging appears to involve a reduction in the threshold for mitochondrial outer membrane permeabilization.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Center for Cell Decision Processes, Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | | |
Collapse
|
29
|
Rehm M, Prehn JHM. Systems modelling methodology for the analysis of apoptosis signal transduction and cell death decisions. Methods 2013; 61:165-73. [PMID: 23607991 DOI: 10.1016/j.ymeth.2013.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 04/09/2013] [Accepted: 04/11/2013] [Indexed: 11/25/2022] Open
Abstract
Systems biology and systems medicine, i.e. the application of systems biology in a clinical context, is becoming of increasing importance in biology, drug discovery and health care. Systems biology incorporates knowledge and methods that are applied in mathematics, physics and engineering, but may not be part of classical training in biology. We here provide an introduction to basic concepts and methods relevant to the construction and application of systems models for apoptosis research. We present the key methods relevant to the representation of biochemical processes in signal transduction models, with a particular reference to apoptotic processes. We demonstrate how such models enable a quantitative and temporal analysis of changes in molecular entities in response to an apoptosis-inducing stimulus, and provide information on cell survival and cell death decisions. We introduce methods for analyzing the spatial propagation of cell death signals, and discuss the concepts of sensitivity analyses that enable a prediction of network responses to disturbances of single or multiple parameters.
Collapse
Affiliation(s)
- Markus Rehm
- Centre for Systems Medicine and Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | | |
Collapse
|
30
|
FRET-based evaluation of Bid cleavage in a single primary cultured neuron. Neurosci Lett 2013; 536:24-8. [DOI: 10.1016/j.neulet.2012.11.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/25/2012] [Accepted: 11/28/2012] [Indexed: 11/18/2022]
|
31
|
de Wilt LHAM, Kroon J, Jansen G, de Jong S, Peters GJ, Kruyt FAE. Bortezomib and TRAIL: a perfect match for apoptotic elimination of tumour cells? Crit Rev Oncol Hematol 2013; 85:363-372. [PMID: 22944363 DOI: 10.1016/j.critrevonc.2012.08.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/13/2012] [Accepted: 08/06/2012] [Indexed: 01/11/2023] Open
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine that selectively eradicates tumour cells via specific cell surface receptors and is intensively explored for use as a novel anticancer approach. To enhance the efficacy of TRAIL receptor agonists the proteasome inhibitor bortezomib is one of the most potent sensitizers. Here we review the main mechanisms underlying bortezomib-dependent TRAIL sensitization, including stimulation of apoptosis by increasing expression of TRAIL receptors, reduction of cFLIP and enhancement of caspase 8 activation, and modulation of Bcl-2 family proteins and inhibitor of apoptosis proteins (IAPs). Concomitantly, pro-survival signals are suppressed such as elicited by NF-κB and Akt. The different preclinical tumour models explored with this combination, including primary tumour (stem) cells, stroma co-culture and mice models, are discussed, as well as possible hurdles for clinical activity. Collectively, anticipating a solid rationale for bortezomib-TRAIL combination and very promising preclinical results, its clinical activity remains to be demonstrated.
Collapse
Affiliation(s)
- L H A M de Wilt
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
32
|
Kutumova E, Zinovyev A, Sharipov R, Kolpakov F. Model composition through model reduction: a combined model of CD95 and NF-κB signaling pathways. BMC SYSTEMS BIOLOGY 2013; 7:13. [PMID: 23409788 PMCID: PMC3626841 DOI: 10.1186/1752-0509-7-13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 02/05/2013] [Indexed: 12/22/2022]
Abstract
Background Many mathematical models characterizing mechanisms of cell fate decisions have been constructed recently. Their further study may be impossible without development of methods of model composition, which is complicated by the fact that several models describing the same processes could use different reaction chains or incomparable sets of parameters. Detailed models not supported by sufficient volume of experimental data suffer from non-unique choice of parameter values, non-reproducible results, and difficulty of analysis. Thus, it is necessary to reduce existing models to identify key elements determining their dynamics, and it is also required to design the methods allowing us to combine them. Results Here we propose a new approach to model composition, based on reducing several models to the same level of complexity and subsequent combining them together. Firstly, we suggest a set of model reduction tools that can be systematically applied to a given model. Secondly, we suggest a notion of a minimal complexity model. This model is the simplest one that can be obtained from the original model using these tools and still able to approximate experimental data. Thirdly, we propose a strategy for composing the reduced models together. Connection with the detailed model is preserved, which can be advantageous in some applications. A toolbox for model reduction and composition has been implemented as part of the BioUML software and tested on the example of integrating two previously published models of the CD95 (APO-1/Fas) signaling pathways. We show that the reduced models lead to the same dynamical behavior of observable species and the same predictions as in the precursor models. The composite model is able to recapitulate several experimental datasets which were used by the authors of the original models to calibrate them separately, but also has new dynamical properties. Conclusion Model complexity should be comparable to the complexity of the data used to train the model. Systematic application of model reduction methods allows implementing this modeling principle and finding models of minimal complexity compatible with the data. Combining such models is much easier than of precursor models and leads to new model properties and predictions.
Collapse
Affiliation(s)
- Elena Kutumova
- Institute of Systems Biology, Ltd, 15 Detskiy proezd, Novosibirsk 630090, Russia.
| | | | | | | |
Collapse
|