1
|
Dong L, Sun Q, Song F, Song X, Lu C, Li Y, Song X. Identification and verification of eight cancer-associated fibroblasts related genes as a prognostic signature for head and neck squamous cell carcinoma. Heliyon 2023; 9:e14003. [PMID: 36938461 PMCID: PMC10018481 DOI: 10.1016/j.heliyon.2023.e14003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) can exert their immunosuppressive effects by secreting various effectors that are involved in the regulation of tumor-infiltrating immune cells as well as other immune components in the tumor immune microenvironment (TIME), thereby promoting tumorigenesis, progression, metastasis, and drug resistance. Although a large number of studies suggest that CAFs play a key regulatory role in the development of head and neck squamous cell carcinoma (HNSCC), there are limited studies on the relevance of CAFs to the prognosis of HNSCC. In this study, we identified a prognostic signature containing eight CAF-related genes for HNSCC by univariate Cox analysis, lasso regression, stepwise regression, and multivariate Cox analysis. Our validation in primary cultures of CAFs from human HNSCC and four human HNSCC cell lines confirmed that these eight genes are indeed characteristic markers of CAFs. Immune cell infiltration differences analysis between high-risk and low-risk groups according to the eight CAF-related genes signature hinted at CAFs regulatory roles in the TIME, further revealing its potential role on prognosis. The signature of the eight CAF-related genes was validated in different independent validation cohorts and all showed that it was a valid marker for prognosis. The significantly higher overall survival (OS) in the low-risk group compared to the high-risk group was confirmed by Kaplan-Meier (K-M) analysis, suggesting that the signature of CAF-related genes can be used as a non-invasive predictive tool for HNSCC prognosis. The low-risk group had significantly higher levels of tumor-killing immune cell infiltration, as confirmed by CIBERSORT analysis, such as CD8+ T cells, follicular helper T cells, and Dendritic cells (DCs) in the low-risk group. In contrast, the level of infiltration of pro-tumor cells such as M0 macrophages and activated Mast cells (MCs) was lower. It is crucial to delve into the complex mechanisms between CAFs and immune cells to find potential regulatory targets and may provide new evidence for subsequently targeted immunotherapy. These results suggest that the signature of the eight CAF-related genes is a powerful indicator for the assessment of the TIME of HNSCC. It may provide a new and reliable potential indicator for clinicians to predict the prognosis of HNSCC, which may be used to guide treatment and clinical decision-making in HNSCC patients. Meanwhile, CAF-related genes are expected to become tumor biomarkers and effective targets for HNSCC.
Collapse
Key Words
- CAFs, Cancer-associated fibroblasts
- CSCs, cancer stem cells
- Cancer-associated fibroblasts
- DCs, Dendritic cells
- EMT, epithelial mesenchymal transition
- GEO, Gene Expression Omnibus
- GEPIA, Gene Expression Profiling Interactive Analysis
- GO, Gene Ontology
- GSEA, Gene Set Enrichment Analysis
- HNSCC, head and neck squamous cell carcinoma
- HR, Hazard Ratio
- Head and neck squamous cell carcinoma
- Immune cell infiltration
- K-M, Kaplan-Meier
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MCs, Mast cells
- NFs, normal fibroblasts
- OS, overall survival
- OSCC, oral squamous cell carcinomas
- Prognostic signature
- ROC, receiver operating characteristic
- TAMs, tumor-associated macrophages
- TCGA, The Cancer Genome Atlas
- TIME, tumor immune microenvironment
- TME, tumor microenvironment
Collapse
Affiliation(s)
- Lei Dong
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
| | - Qi Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
| | - Fei Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
| | - Xiaoyu Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
| | - Congxian Lu
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
| | - Yumei Li
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
- Corresponding author. Yumei Li: Department of Otorhinolaryngology Head and Neck Surgery. Yantai Yuhuangding Hospital, No.20, Yuhuangding East Road, Zhifu District, Yantai, Shandong, 264000, China.
| | - Xicheng Song
- Department of Otorhinolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Shandong University, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases
- Corresponding author. Xicheng Song: Department of Otorhinolaryngology Head and Neck Surgery. Yantai Yuhuangding Hospital, No.20, Yuhuangding East Road, Zhifu District, Yantai, Shandong, 264000, China.
| |
Collapse
|
2
|
Zhang X, Dai X, Zhao X, Wang J, Dou J, Zhuang H, Chen N, Zhao H. MiR-874-3p represses the migration and invasion yet promotes the apoptosis and cisplatin sensitivity via being sponged by long intergenic non-coding RNA 00922 (LINC00922) and targeting Glycerophosphodiester Phosphodiesterase Domain Containing 5 (GDPD5) in gastric cancer cells. Bioengineered 2022; 13:7082-7104. [PMID: 35282764 PMCID: PMC9208458 DOI: 10.1080/21655979.2022.2045831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Our study mainly reports the specific mechanisms of microRNA (miR)-874-3p on drug resistance in gastric cancer (GC). Clinical specimen was collected. The upstream long non-coding RNA (lncRNA) and the downstream gene of miR-874-3p were predicted using bioinformatic analysis with the results being ascertained with dual-luciferase reporter assay. The viability, apoptosis, migration and invasion of transfected GC cells with or without cisplatin (DDP) treatment were evaluated by Cell Counting Kit-8 (CCK-8), flow cytometric, Scratch, and Transwell assays. An animal xenograft model was constructed. Expressions of long intergenic non-coding RNA 00922 (LINC00922), miR-874-3p and potential target genes were quantified by quantitative real-time polymerase-chain reaction (qRT-PCR) and Western blot. MiR-874-3p, which was lower-expressed in drug-resistant GC tissues and cells, was upregulated to repress the viability, migration and invasion but enhance the apoptosis and sensitivity in GC cells with or without DDP resistance. Downregulation of miR-874-3p eliminated the effects of silenced LINC00922, a upstream lncRNA of miR-874-3p, on cell viability, apoptosis, migration and invasion, as well as the expressions of Glycerophosphodiester Phosphodiesterase Domain Containing 5 (GDPD5) and the downstream gene of miR-874-3p in DDP-resistant GC cells. GDPD5 silencing diminished the effects of miR-874-3p downregulation on GDPD5 expression, viability, migration and invasion of DDP-resistant GC cells. Additionally, LINC00922 silencing enhanced the inhibitory effect of DDP on tumor growth, whereas reversing the effects of DDP on LINC00922, miR-874-3p and GDPD5 expressions in tumors. MiR-874-3p, an miRNA, which is sponged by LINC00922 and targets GDPD5, inhibits the GC progression yet enhances the DDP sensitivity in GC.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Xudong Dai
- Department of General Surgery, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xin Zhao
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Jian Wang
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Jin Dou
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Haiwen Zhuang
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Ning Chen
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Haijian Zhao
- Division of Gastrointestinal Surgery, Department of General Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, Jiangsu, China
| |
Collapse
|
3
|
Parthenolide as Cooperating Agent for Anti-Cancer Treatment of Various Malignancies. Pharmaceuticals (Basel) 2020; 13:ph13080194. [PMID: 32823992 PMCID: PMC7466132 DOI: 10.3390/ph13080194] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Primary and acquired resistance of cancer to therapy is often associated with activation of nuclear factor kappa B (NF-κB). Parthenolide (PN) has been shown to inhibit NF-κB signaling and other pro-survival signaling pathways, induce apoptosis and reduce a subpopulation of cancer stem-like cells in several cancers. Multimodal therapies that include PN or its derivatives seem to be promising approaches enhancing sensitivity of cancer cells to therapy and diminishing development of resistance. A number of studies have demonstrated that several drugs with various targets and mechanisms of action can cooperate with PN to eliminate cancer cells or inhibit their proliferation. This review summarizes the current state of knowledge on PN activity and its potential utility as complementary therapy against different cancers.
Collapse
|
4
|
Liu X, Zhang N, Wang D, Zhu D, Yuan Q, Zhang X, Qian L, Niu H, Lu Y, Ren G, Tian K, Yuan H. Downregulation of reticulocalbin-1 differentially facilitates apoptosis and necroptosis in human prostate cancer cells. Cancer Sci 2018; 109:1147-1157. [PMID: 29453900 PMCID: PMC5891187 DOI: 10.1111/cas.13541] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 02/04/2018] [Accepted: 02/10/2018] [Indexed: 12/16/2022] Open
Abstract
Reticulocalbin 1 (RCN1), an endoplasmic reticulum (ER)‐resident Ca2+‐binding protein, is dysregulated in cancers, but its pathophysiological roles are largely unclear. Here, we demonstrate that RCN1 is overexpressed in clinical prostate cancer (PCa) samples, associated with cyclin B, not cyclin D1 expression, compared to that of benign tissues in a Chinese Han population. Downregulation of endogenous RCN1 significantly suppresses PCa cell viability and arrests the cell cycles of DU145 and LNCaP cells at the S and G2/M phases, respectively. RCN1 depletion causes ER stress, which is evidenced by induction of GRP78, activation of PERK and phosphorylation of eIF2α in PCa cells. Remarkably, RCN1 loss triggers DU145 cell apoptosis in a caspase‐dependent manner but mainly causes necroptosis in LNCaP cells. An animal‐based analysis confirms that RCN1 depletion suppresses cell proliferation and promotes cell death. Further investigations reveal that RCN1 depletion leads to elevation of phosphatase and tensin homolog (PTEN) and inactivation of AKT in DU145 cells. Silencing of PTEN partially restores apoptotic cells upon RCN1 loss. In LNCaP cells, predominant activation of CaMKII is important for necroptosis in response to RCN1 depletion. Thus, RCN1 may promote cell survival and serve as a useful target for cancer therapy.
Collapse
Affiliation(s)
- Xiaofei Liu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Nianzhao Zhang
- Department of Urology, Qilu Hospital, Shandong University, Jinan, China
| | - Dawei Wang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Deyu Zhu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Quan Yuan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Xiulei Zhang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Lilin Qian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Huanmin Niu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Yi Lu
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Guijie Ren
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Keli Tian
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| | - Huiqing Yuan
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
5
|
He J, Cai L, Chen Y, He Y, Wang M, Tang J, Guan H, Wang J, Peng X. Antitumor and radiosensitizing effects of SKLB-163, a novel benzothiazole-2-thiol derivative, on nasopharyngeal carcinoma by affecting the RhoGDI/JNK-1 signaling pathway. Radiother Oncol 2018. [PMID: 29519627 DOI: 10.1016/j.radonc.2018.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE SKLB-163 is a novel benzothiazole-2-thiol derivative with antitumor activities. This study investigated the effects of SKLB-163 on nasopharyngeal carcinoma (NPC) and its mechanisms. MATERIALS AND METHODS Rho GDP-dissociation inhibitor (RhoGDI) expression was examined in NPC cell lines by western blot. Effects of SKLB-163 on proliferation, migration and radiosensitivity were assessed by MTT, wound healing and colony formation assays in vitro. Anti-tumor and anti-metastatic effects, and radiosensitizing effects of SKLB-163 were evaluated in a NPC lung metastatic nude mouse model and a subcutaneous xenograft mouse model. Effects of SKLB-163 on proliferation and apoptosis were assessed by PCNA immunohistochemistry and TUNEL assay in vivo. Key molecules in RhoGDI/c-Jun N-terminal kinases-1 (JNK-1) pathway were examined by western blot. RESULTS RhoGDI was up-regulated in NPC cell lines. SKLB-163 inhibited proliferation and migration, and increased radiosensitivity of NPC cells. SKLB-163 inhibited tumor growth and metastases, and sensitized tumor to irradiation. The radiosensitizing effects were correlated with induction of apoptosis and suppression of proliferation. The molecular mechanism was the down-regulation of RhoGDI and activation of JNK-1 signaling, and the subsequent activation of caspase-3 and the decrease in phosphorylated AKT. CONCLUSIONS SKLB-163 shows strong anti-tumor activities against NPC and sensitizes NPC to irradiation by affecting the RhoGDI/JNK-1 pathway.
Collapse
Affiliation(s)
- Jinlan He
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Lei Cai
- Hepatobiliary Surgery Institute, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ye Chen
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yan He
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Ming Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Tang
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Hui Guan
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Jingjing Wang
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xingchen Peng
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
6
|
Hu X, Li S, He Y, Ai P, Wu S, Su Y, Li X, Cai L, Peng X. Antitumor and antimetastatic activities of a novel benzothiazole-2-thiol derivative in a murine model of breast cancer. Oncotarget 2017; 8:11887-11895. [PMID: 28060755 PMCID: PMC5355312 DOI: 10.18632/oncotarget.14431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 12/21/2016] [Indexed: 02/05/2023] Open
Abstract
The prognosis of metastatic breast cancer is always very poor. Thus, it is urgent to develop novel drugs with less toxicity against metastatic breast cancer. A new drug (XC-591) derived from benzothiazole-2-thiol was designed and synthesized in our lab. In this study, we tried to assess effects of XC-591 treatment on primary breast cancer and pulmonary metastasis in 4T1 mice model. Furthermore, we tried to discover its possible molecular mechanism of action. MTT experiment showed XC-591 had significant anti-cancer activity on diverse cancer cells. Furthermore, XC-591 significantly suppressed the proliferation of 4T1 cells by colony formation assay. The in vivo results displayed that XC-591 could inhibit the growth and metastasis in 4T1 model. Moreover, histological analysis revealed that XC-591 treatment increased apoptosis, inhibited proliferation and angiogenesis in vivo. In addition, XC-591 did not contribute to obvious drug associated toxicity during the whole study. Molecular mechanism showed XC-591 could inhibit RhoGDI, activate caspase-3 and decrease phosphorylated Akt. The present data may be important to further explore this kind of new small-molecule inhibitor.
Collapse
Affiliation(s)
- XiaoLin Hu
- Department of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Sen Li
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital of SouthWest Medical University, Luzhou, China
| | - Yan He
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Ai
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shaoyong Wu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yonglin Su
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolin Li
- Department of Pathophysiology, Basic Medical College, Jilin University, Changchun, China
| | - Lei Cai
- Hepatobiliary Surgery Institute, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xingchen Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Peng XC, Chen XX, Zhang YU, Wang HJ, Feng Y. A novel inhibitor of Rho GDP-dissociation inhibitor α improves the therapeutic efficacy of paclitaxel in Lewis lung carcinoma. Biomed Rep 2015; 3:473-477. [PMID: 26171151 DOI: 10.3892/br.2015.475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 05/21/2015] [Indexed: 02/05/2023] Open
Abstract
Molecular-targeted therapies are considered a promising strategy for the treatment of most types of human cancer. Rho GDP-dissociation inhibitor α (RhoGDIα), which functions mainly by controlling the cellular distribution and activity of Rho GTPases and is associated with tumor progression and poor prognosis of cancer patients, has become a new promising target for anticancer treatment. Recently, a specific RhoGDIα inhibitor (no. SKLB-163) was developed via computer-aided drug design and de novo synthesis. Previous studies have shown that SKLB-163 had extremely good antitumor activities against diverse cancer cell lines. In the present study, SKLB-163 was used in combination with paclitaxel in order to determine the synergistic effect of the antitumor activity. The findings showed that the combination therapy clearly inhibited cell proliferation and induced apoptosis of LL/2 in vitro. The LL/2 mice model also showed that the combination therapy inhibited tumor growth in vivo. Proliferative cell nuclear antigen (PCNA) immunohistochmeistry and terminal deoxynucleotidyl transferase dUTP nick end-labeling showed that combination therapy inhibited cell proliferation and increased apoptosis compared to the treatment with SKLB-163 or paclitaxel alone. The data suggests that the combination therapy exerted synergistic antitumor effects, providing a novel way to augment the antitumor efficacy of cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Xing Chen Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xu Xia Chen
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Y U Zhang
- Department of Oncology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, P.R. China
| | - Hai Jun Wang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu, Sichuan 610041, P.R. China
| | - You Feng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
8
|
Bulatović MZ, Maksimović-Ivanić D, Bensing C, Gómez-Ruiz S, Steinborn D, Schmidt H, Mojić M, Korać A, Golić I, Pérez-Quintanilla D, Momčilović M, Mijatović S, Kaluđerović GN. Organozinn(IV)-beladenes mesoporöses SiO 2als biokompatible Strategie bei der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201400763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
9
|
Bulatović MZ, Maksimović-Ivanić D, Bensing C, Gómez-Ruiz S, Steinborn D, Schmidt H, Mojić M, Korać A, Golić I, Pérez-Quintanilla D, Momčilović M, Mijatović S, Kaluđerović GN. Organotin(IV)-loaded mesoporous silica as a biocompatible strategy in cancer treatment. Angew Chem Int Ed Engl 2014; 53:5982-7. [PMID: 24828572 DOI: 10.1002/anie.201400763] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Indexed: 01/04/2023]
Abstract
The strong therapeutic potential of an organotin(IV) compound loaded in nanostructured silica (SBA-15pSn) is demonstrated: B16 melanoma tumor growth in syngeneic C57BL/6 mice is almost completely abolished. In contrast to apoptosis as the basic mechanism of the anticancer action of numerous chemotherapeutics, the important advantage of this SBA-15pSn mesoporous material is the induction of cell differentiation, an effect unknown for metal-based drugs and nanomaterials alone. This non-aggressive mode of drug action is highly efficient against cancer cells but is in the concentration range used nontoxic for normal tissue. JNK (Jun-amino-terminal kinase)-independent apoptosis accompanied by the development of the melanocyte-like nonproliferative phenotype of survived cells indicates the extraordinary potential of SBA-15pSn to suppress tumor growth without undesirable compensatory proliferation of malignant cells in response to neighboring cell death.
Collapse
Affiliation(s)
- Mirna Z Bulatović
- Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade (Serbia)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Peng X, Xie G, Wang Z, Lin H, Zhou T, Xiang P, Jiang Y, Yang S, Wei Y, Yu L, Zhao Y. SKLB-163, a new benzothiazole-2-thiol derivative, exhibits potent anticancer activity by affecting RhoGDI/JNK-1 signaling pathway. Cell Death Dis 2014; 5:e1143. [PMID: 24675461 PMCID: PMC3973246 DOI: 10.1038/cddis.2014.107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 02/05/2023]
Abstract
Small-molecule inhibitors are an attractive therapeutic approach for most types of human cancers. SKLB-163, a novel benzothiazole-2-thiol derivative, was developed via computer-aided drug design and de novo synthesis. MTT assay showed it had potent anti-proliferative activity on various human cancer cells. Treatment of cancer cells with SKLB-163 induced obvious apoptosis and inhibited proliferation in vitro. SKLB-163 administered p.o. showed a marked antitumor activity in vivo. Proteomic techniques were employed to identify possible drug target proteins. The data showed molecular mechanism of action might be involved in downregulation of RhoGDI, which finally contributed to increased apoptosis and inhibited proliferation. These findings provided the potential value of SKLB-163 as a novel candidate antitumor drug.
Collapse
Affiliation(s)
- X Peng
- 1] State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China [2] Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - G Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Z Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - H Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - T Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - P Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Y Jiang
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - S Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Y Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - L Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Y Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Findeisen P, Peccerella T, Neumaier M, Schadendorf D. Proteomics for biomarker discovery in malignant melanoma. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.3.2.209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Szaflarski W, Sujka-Kordowska P, Januchowski R, Wojtowicz K, Andrzejewska M, Nowicki M, Zabel M. Nuclear localization of P-glycoprotein is responsible for protection of the nucleus from doxorubicin in the resistant LoVo cell line. Biomed Pharmacother 2013; 67:497-502. [DOI: 10.1016/j.biopha.2013.03.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 03/10/2013] [Indexed: 10/27/2022] Open
|
13
|
Arai K, Sakamoto R, Kubota D, Kondo T. Proteomic approach toward molecular backgrounds of drug resistance of osteosarcoma cells in spheroid culture system. Proteomics 2013; 13:2351-60. [DOI: 10.1002/pmic.201300053] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/12/2013] [Accepted: 04/24/2013] [Indexed: 01/08/2023]
Affiliation(s)
- Kazuya Arai
- Division of Pharmacoproteomics; National Cancer Center Research Institute; Tokyo Japan
- SCIVAX Corporation; Kanagawa Japan
| | - Ruriko Sakamoto
- Division of Pharmacoproteomics; National Cancer Center Research Institute; Tokyo Japan
- SCIVAX Corporation; Kanagawa Japan
| | - Daisuke Kubota
- Division of Pharmacoproteomics; National Cancer Center Research Institute; Tokyo Japan
| | - Tadashi Kondo
- Division of Pharmacoproteomics; National Cancer Center Research Institute; Tokyo Japan
| |
Collapse
|
14
|
Lage H. Proteomic approaches for investigation of therapy resistance in cancer. Proteomics Clin Appl 2012; 3:883-911. [PMID: 21136994 DOI: 10.1002/prca.200800162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Resistance to anticancer therapy is a major obstacle for successful management of patients in oncology. Although in the past, various biological mechanisms involved in therapy resistance, in particular multidrug resistance, have been identified, cancer patients did not really benefit. The mechanisms include the enhanced activity of drug extrusion pumps, modulation of cellular death pathways, alteration and repair of target molecules and various other mechanisms. Together they build a complex network mediating an individual therapy-resistant phenotype. The improved description of this multifactorial network should be useful for prediction of treatment response and would allow to design an individual-tailored therapy regiment. Proteome analyzing technologies appear as powerful tools for identifying new factors and protein expression profiles associated with anticancer therapy resistance. In the last years, the application of proteomic techniques identified multiple new factors or protein expression signatures in drug-resistant cell models and cancerous tissues. However, the functional role and the clinical impact of these findings are not yet clarified. So far, none of the proteomic data were useful for the development of improved diagnostic tests, for prediction of individual therapy response or for development of updated chemosensitizers. Here, the previous therapy resistance-related proteome data and future perspectives will be discussed.
Collapse
Affiliation(s)
- Hermann Lage
- Institute of Pathology, Charité Campus Mitte, Berlin, Germany.
| |
Collapse
|
15
|
Peng X, Gong F, Xie G, Zhao Y, Tang M, Yu L, Tong A. A proteomic investigation into adriamycin chemo-resistance of human leukemia K562 cells. Mol Cell Biochem 2011; 351:233-41. [PMID: 21243406 DOI: 10.1007/s11010-011-0730-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 01/10/2011] [Indexed: 02/05/2023]
Abstract
This study aimed to explore the mechanism of adriamycin resistance in human chronic myelogenous leukemia cells. Proteomic approach was utilized to compare and identify differentially expressed proteins between human chronic myelogenous leukemia K562 cells and their adriamycin-resistant counterparts. The differentially expressed proteins were analyzed by 2-DE (two-dimensional gel electrophoresis), and protein identification were performed on ESI-Q-TOF MS/MS instrument. Out of the 35 differentially expressed proteins between the two cell lines, 29 were identified and grouped into 10 functional classes. Most of identified proteins were related to the categories of metabolism (24%), proteolysis (13%), signal transduction (21%) and calcium ion binding (6%), suggesting that alterations of those biological processes might be involved in adriamycin resistance of K562 cells. We believe this study may provide some clues to a better understanding of the molecular mechanisms underlying adriamycin resistance.
Collapse
Affiliation(s)
- Xingchen Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
16
|
Balasubramani M, Nakao C, Uechi GT, Cardamone J, Kamath K, Leslie KL, Balachandran R, Wilson L, Day BW, Jordan MA. Characterization and detection of cellular and proteomic alterations in stable stathmin-overexpressing, taxol-resistant BT549 breast cancer cells using offgel IEF/PAGE difference gel electrophoresis. Mutat Res 2010; 722:154-64. [PMID: 20816848 DOI: 10.1016/j.mrgentox.2010.08.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 08/25/2010] [Indexed: 12/15/2022]
Abstract
Stathmin/oncoprotein 18, a protein that regulates microtubule dynamics, is highly expressed in a number of tumors including leukemia, lymphoma, neuroblastoma, breast, ovarian, and prostate cancers. High stathmin levels have been associated with the development of resistance to the widely used anti-cancer drug taxol ((®)Taxol, paclitaxel). The mechanisms of stathmin-mediated taxol resistance are not well-understood at the molecular level. To better understand the role of stathmin in taxol resistance, we stably overexpressed stathmin twofold in BT549 human breast cancer cells and characterized several cell processes involved in the mechanism of action of taxol. After stable overexpression of stathmin, neither the cell doubling time nor the mitotic index was altered and the microtubule polymer mass was reduced only modestly (by 18%). Unexpectedly, microtubule dynamicity was reduced by 29% after stathmin overexpression, resulting primarily from reduction in the catastrophe frequency. Sensitivity to taxol was reduced significantly (by 44%) in a clonogenic assay, and stathmin appeared to protect the cells from the spindle-damaging effects of taxol. The results suggest that in the stably stathmin-overexpressing clones, compensatory gene expression occurred that resulted in normal rates of cell proliferation and prevented the increase in catastrophe frequency expected in response to stathmin. Stathmin overexpression protected the cells from taxol-induced abnormal mitoses, and thus induced taxol resistance. Using offgel IEF/PAGE difference gel electrophoresis, we identified a number of proteins whose expression is reduced in the taxol-resistant stathmin-overexpressing cell lines, including proteins involved in the cytoskeleton and cell structure, the stress response, protein folding, glycolysis, and catalysis.
Collapse
|
17
|
Lee WY, Lee PPF, Yan YK, Lau M. Cytotoxic copper(ii) salicylaldehyde semicarbazone complexes: Mode of action and proteomic analysis. Metallomics 2010; 2:694-705. [DOI: 10.1039/c0mt00016g] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
18
|
Honoré B. The rapidly expanding CREC protein family: members, localization, function, and role in disease. Bioessays 2009; 31:262-77. [PMID: 19260022 DOI: 10.1002/bies.200800186] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although many aspects of the physiological and pathophysiological mechanisms remain unknown, recent advances in our knowledge suggest that the CREC proteins are promising disease biomarkers or targets for therapeutic intervention in a variety of diseases. The CREC family of low affinity, Ca2+-binding, multiple EF-hand proteins are encoded by five genes, RCN1, RCN2, RCN3, SDF4, and CALU, resulting in reticulocalbin, ER Ca2+-binding protein of 55 kDa (ERC-55), reticulocalbin-3, Ca2+-binding protein of 45 kDa (Cab45), and calumenin. Alternative splicing increases the number of gene products. The proteins are localized in the cytosol, in various parts of the secretory pathway, secreted to the extracellular space or localized on the cell surface. The emerging functions appear to be highly diverse. The proteins interact with several different ligands. Rather well-described functions are attached to calumenin with the inhibition of several proteins in the endoplasmic or sarcoplasmic reticulum membrane, the vitamin K(1) 2,3-epoxide reductase, the gamma-carboxylase, the ryanodine receptor, and the Ca2+-transporting ATPase. Other functions concern participation in the secretory process, chaperone activity, signal transduction as well as participation in a large variety of disease processes.
Collapse
Affiliation(s)
- Bent Honoré
- Department of Medical Biochemistry, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
19
|
Bermúdez-Crespo J, López JL. A better understanding of molecular mechanisms underlying human disease. Proteomics Clin Appl 2007; 1:983-1003. [PMID: 21136752 DOI: 10.1002/prca.200700086] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Indexed: 01/06/2023]
Abstract
This review summarises and discusses the degree to which proteomics is contributing to medical care, providing examples and signspots for future directions. Why do genomic approaches provide a limited view of gene expression? Because of the multifactorial nature of many diseases, proteomics enables us to understand the molecular basis of disease, not only at the organism, whole-cell or tissue levels, but also in subcellular structures, protein complexes and biological fluids. The application of proteomics in medicine is expected to have a major impact by providing an integrated view of individual disease processes. This review describes several proteomic platforms and examines the role of proteomics as a tool for clinical biomarker discovery, the identification of prognostic and earlier diagnostic markers, their use in monitoring the effects of drug treatments and eventually find more efficient and safer therapeutics for a wide range of pathologies.
Collapse
Affiliation(s)
- José Bermúdez-Crespo
- Department of Genetics, Faculty of Biology, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | |
Collapse
|
20
|
Blumenthal RD, Goldenberg DM. Methods and goals for the use of in vitro and in vivo chemosensitivity testing. Mol Biotechnol 2007; 35:185-97. [PMID: 17435285 DOI: 10.1007/bf02686104] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 10/22/2022]
Abstract
Sensitive, specific, and accurate methods to assay chemosensitivity are needed to (1) screen new therapeutic agents, (2) identify patterns of chemosensitivity for different tumor types, (3) establish patterns of cross-resistance and sensitivity in treatment of naïve and relapsing tumors, (4) identify genomic and proteomic profiles associated with sensitivity, (5) correlate in vitro response with preclinical in vivo effects and clinical outcomes for a particular therapeutic agent, and (6) tailor chemotherapy regimens to individual patients. Various methods are available to achieve these end points, including several in vitro clonogenic and proliferation assays, cell metabolic activity assays, molecular assays to monitor expression of markers for responsiveness, drug resistance, and for induction of apoptosis, in vivo tumor growth and survival assays in metastatic and orthotopic models, and in vivo imaging assays. The advantages and disadvantages of the specific assays are discussed. A summary of research questions related to chemosensitivity testing is also included.
Collapse
|
21
|
Niméus E, Malmström J, Johnsson A, Marko-Varga G, Fernö M. Proteomic analysis identifies candidate proteins associated with distant recurrences in breast cancer after adjuvant chemotherapy. J Pharm Biomed Anal 2007; 43:1086-93. [PMID: 17085005 DOI: 10.1016/j.jpba.2006.09.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 09/10/2006] [Accepted: 09/11/2006] [Indexed: 11/20/2022]
Abstract
Breast cancer is a heterogeneous disease and it is of importance to select patients with regard to different prognosis and treatment sensitivity to individualize treatment regimes. In this study we successfully adapted a protein extraction protocol from mRNA extracted tumor samples enabling two-dimensional gel electrophoresis (2-DE) analysis of samples previously analyzed by cDNA microarray. The aim was to find candidate proteins that distinguish breast cancer patients with or without recurrences after adjuvant CMF (cyclophosphamide, methotrexate and 5-FU) treatment within four years to follow-up. We identified several proteins distinguishing the recurrence group from the non-recurrence group, especially in the ER and PgR positive subgroup (n=7). The induced proteins were involved in translation/folding, iron ion binding, and protease inhibition, whereas proteins involved in signaling, ubiquitination, and splicing were decreased in expression. These results show that it is possible to use 2-DE to separate high abundant proteins in breast cancer tissue and to find discriminating proteins to identify patients with different prognosis after adjuvant CMF treatment.
Collapse
Affiliation(s)
- Emma Niméus
- Department of Oncology, Clinical Sciences, University Hospital, Lund, Sweden
| | | | | | | | | |
Collapse
|
22
|
Busch MHJ, Vollmann W, Grönemeyer DHW. Finite volume analysis of temperature effects induced by active MRI implants: 2. Defects on active MRI implants causing hot spots. Biomed Eng Online 2006; 5:35. [PMID: 16729878 PMCID: PMC1513583 DOI: 10.1186/1475-925x-5-35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 05/26/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Active magnetic resonance imaging implants, for example stents, stent grafts or vena cava filters, are constructed as wireless inductively coupled transmit and receive coils. They are built as a resonator tuned to the Larmor frequency of a magnetic resonance system. The resonator can be added to or incorporated within the implant. This technology can counteract the shielding caused by eddy currents inside the metallic implant structure. This may allow getting diagnostic information of the implant lumen (in stent stenosis or thrombosis for example). The electro magnetic rf-pulses during magnetic resonance imaging induce a current in the circuit path of the resonator. A by material fatigue provoked partial rupture of the circuit path or a broken wire with touching surfaces can set up a relatively high resistance on a very short distance, which may behave as a point-like power source, a hot spot, inside the body part the resonator is implanted to. This local power loss inside a small volume can reach (1/4) of the total power loss of the intact resonating circuit, which itself is proportional to the product of the resonator volume and the quality factor and depends as well from the orientation of the resonator with respect to the main magnetic field and the imaging sequence the resonator is exposed to. METHODS First an analytical solution of a hot spot for thermal equilibrium is described. This analytical solution with a definite hot spot power loss represents the worst case scenario for thermal equilibrium inside a homogeneous medium without cooling effects. Starting with this worst case assumptions additional conditions are considered in a numerical simulation, which are more realistic and may make the results less critical. The analytical solution as well as the numerical simulations use the experimental experience of the maximum hot spot power loss of implanted resonators with a definite volume during magnetic resonance imaging investigations. The finite volume analysis calculates the time developing temperature maps for the model of a broken linear metallic wire embedded in tissue. Half of the total hot spot power loss is assumed to diffuse into both wire parts at the location of a defect. The energy is distributed from there by heat conduction. Additionally the effect of blood perfusion and blood flow is respected in some simulations because the simultaneous appearance of all worst case conditions, especially the absence of blood perfusion and blood flow near the hot spot, is very unlikely for vessel implants. RESULTS The analytical solution as worst case scenario as well as the finite volume analysis for near worst case situations show not negligible volumes with critical temperature increases for part of the modeled hot spot situations. MR investigations with a high rf-pulse density lasting below a minute can establish volumes of several cubic millimeters with temperature increases high enough to start cell destruction. Longer exposure times can involve volumes larger than 100 mm3. Even temperature increases in the range of thermal ablation are reached for substantial volumes. MR sequence exposure time and hot spot power loss are the primary factors influencing the volume with critical temperature increases. Wire radius, wire material as well as the physiological parameters blood perfusion and blood flow inside larger vessels reduce the volume with critical temperature increases, but do not exclude a volume with critical tissue heating for resonators with a large product of resonator volume and quality factor. CONCLUSION The worst case scenario assumes thermal equilibrium for a hot spot embedded in homogeneous tissue without any cooling due to blood perfusion or flow. The finite volume analysis can calculate the results for near and not close to worst case conditions. For both cases a substantial volume can reach a critical temperature increase in a short time. The analytical solution, as absolute worst case, points out that resonators with a small product of inductance volume and quality factor (Q V(ind) < 2 cm3) are definitely save. Stents for coronary vessels or resonators used as tracking devices for interventional procedures therefore have no risk of high temperature increases. The finite volume analysis shows for sure that also conditions not close to the worst case reach physiologically critical temperature increases for implants with a large product of inductance volume and quality factor (Q V(ind) > 10 cm3). Such resonators exclude patients from exactly the MRI investigation these devices are made for.
Collapse
Affiliation(s)
- Martin HJ Busch
- Grönemeyer Institute for Microtherapy, University of Witten/Herdecke, Universitätsstr. 142, D-44799 Bochum, Germany
| | - Wolfgang Vollmann
- Department of mathematics, physics and chemistry, TFH University of Applied Sciences, Luxemburger Straße 10, D-13353 Berlin, Germany
| | - Dietrich HW Grönemeyer
- Grönemeyer Institute for Microtherapy, University of Witten/Herdecke, Universitätsstr. 142, D-44799 Bochum, Germany
| |
Collapse
|
23
|
Righetti PG, Castagna A, Antonioli P, Cecconi D, Campostrini N, Righetti SC. Proteomic approaches for studying chemoresistance in cancer. Expert Rev Proteomics 2006; 2:215-28. [PMID: 15892566 DOI: 10.1586/14789450.2.2.215] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The role of various proteins involved in drug resistance in tumor cells is discussed in this review. Two types of studies are covered: those performed in the preproteomics era and those carried out with modern proteomic tools, namely 2D (electrophoretic) maps and 2D chromatography. In the preproteomic studies, one protein had generally been held responsible for a given chemoresistance. However, analysis via proteomic tools may reveal entire sets of proteins that are up- or downregulated (or switched on/off) in chemoresistant tumor cell lines compared with parental tumor lines. Therefore, it appears more realistic to expect that exposure of cells to drugs results in the activation of different mechanisms of resistance. Such investigations have led to the broadly shared opinion that exposure of cells to drugs results in the activation of different mechanisms of resistance, and that a specific drug-resistant phenotype consists of several molecular mechanisms that are simultaneously active. The proteomic papers reviewed clearly support the hypothesis that many metabolic pathways are affected during the resistance process. Although the modulation of expression levels of such proteins is not clear proof of their role in drug resistance per se, at least some of the themes are very likely to be involved in the resistance phenotype, and thus may be potential targets for new drugs. It is hoped that this review will bring new insight in this field and will stimulate novel and deeper searches with proteomic tools (including prefractionation of subcellular organelles, such as nuclei, to bring to the fore low-abundance proteins that might be responsible for the onset of drug resistance).
Collapse
Affiliation(s)
- Pier Giorgio Righetti
- University of Verona, Department of Industrial & Agricultural Biotechnologies, Laboratory of Proteome Science, Strada Le Grazie 15, Verona, Italy.
| | | | | | | | | | | |
Collapse
|
24
|
Zhang B, Zhang Y, Dagher MC, Shacter E. Rho GDP dissociation inhibitor protects cancer cells against drug-induced apoptosis. Cancer Res 2005; 65:6054-62. [PMID: 16024605 DOI: 10.1158/0008-5472.can-05-0175] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rho GDP dissociation inhibitor (RhoGDI) plays an essential role in control of a variety of cellular functions through interactions with Rho family GTPases, including Rac1, Cdc42, and RhoA. RhoGDI is frequently overexpressed in human tumors and chemo-resistant cancer cell lines, raising the possibility that RhoGDI might play a role in the development of drug resistance in cancer cells. We found that overexpression of RhoGDI increased resistance of cancer cells (MDA-MB-231 human breast cancer cells and JLP-119 lymphoma cells) to the induction of apoptosis by two chemotherapeutic agents: etoposide and doxorubicin. Conversely, silencing of RhoGDI expression by DNA vector-mediated RNA interference (small interfering RNA) sensitized MDA-MB-231 cells to drug-induced apoptosis. Resistance to apoptosis was restored by reintroduction of RhoGDI protein expression. The mechanism for the anti-apoptotic activity of RhoGDI may derive from its ability to inhibit caspase-mediated cleavage of Rac1 GTPase, which is required for maximal apoptosis to occur in response to cytotoxic drugs. Taken together, the data show that RhoGDI is an anti-apoptotic molecule that mediates cellular resistance to these chemotherapy agents.
Collapse
Affiliation(s)
- Baolin Zhang
- Laboratory of Biochemistry, Division of Therapeutic Proteins, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, USA.
| | | | | | | |
Collapse
|
25
|
Busch MHJ, Vollmann W, Schnorr J, Grönemeyer DHW. Finite volume analysis of temperature effects induced by active MRI implants with cylindrical symmetry: 1. Properly working devices. Biomed Eng Online 2005; 4:25. [PMID: 15819973 PMCID: PMC1087857 DOI: 10.1186/1475-925x-4-25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2004] [Accepted: 04/08/2005] [Indexed: 11/10/2022] Open
Abstract
Background Active Magnetic Resonance Imaging implants are constructed as resonators tuned to the Larmor frequency of a magnetic resonance system with a specific field strength. The resonating circuit may be embedded into or added to the normal metallic implant structure. The resonators build inductively coupled wireless transmit and receive coils and can amplify the signal, normally decreased by eddy currents, inside metallic structures without affecting the rest of the spin ensemble. During magnetic resonance imaging the resonators generate heat, which is additional to the usual one described by the specific absorption rate. This induces temperature increases of the tissue around the circuit paths and inside the lumen of an active implant and may negatively influence patient safety. Methods This investigation provides an overview of the supplementary power absorbed by active implants with a cylindrical geometry, corresponding to vessel implants such as stents, stent grafts or vena cava filters. The knowledge of the overall absorbed power is used in a finite volume analysis to estimate temperature maps around different implant structures inside homogeneous tissue under worst-case assumptions. The "worst-case scenario" assumes thermal heat conduction without blood perfusion inside the tissue around the implant and mostly without any cooling due to blood flow inside vessels. Results The additional power loss of a resonator is proportional to the volume and the quality factor, as well as the field strength of the MRI system and the specific absorption rate of the applied sequence. For properly working devices the finite volume analysis showed only tolerable heating during MRI investigations in most cases. Only resonators transforming a few hundred mW into heat may reach temperature increases over 5 K. This requires resonators with volumes of several ten cubic centimeters, short inductor circuit paths with only a few 10 cm and a quality factor above ten. Using MR sequences, for which the MRI system manufacturer declares the highest specific absorption rate of 4 W/kg, vascular implants with a realistic construction, size and quality factor do not show temperature increases over a critical value of 5 K. Conclusion The results show dangerous heating for the assumed "worst-case scenario" only for constructions not acceptable for vascular implants. Realistic devices are safe with respect to temperature increases. However, this investigation discusses only properly working devices. Ruptures or partial ruptures of the wires carrying the electric current of the resonance circuits or other defects can set up a power source inside an extremely small volume. The temperature maps around such possible "hot spots" should be analyzed in an additional investigation.
Collapse
Affiliation(s)
- Martin HJ Busch
- Research and Development Center for Microtherapy (EFMT), D-44799 Bochum, Germany
- Grönemeyer Institute for Microtherapy, University of Witten/Herdecke, D-44799 Bochum, Germany
| | | | - Jörg Schnorr
- Institut für Radiologie, Charité, Medizinische Fakultät, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Dietrich HW Grönemeyer
- Grönemeyer Institute for Microtherapy, University of Witten/Herdecke, D-44799 Bochum, Germany
| |
Collapse
|
26
|
Abstract
Proteomics, the global analysis of expressed cellular proteins, provides powerful tools for the detailed comparison of proteins from normal and neoplastic tissue. In particular, cancer cell culture models are suited for applying proteomics techniques, such as two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and mass spectrometry, to identify specific protein expression profiles and/or proteins that may be associated with a defined phenotype of the cancer cells. As an instance of such an application of proteomics techniques, the detailed proteome analyses of different drug-resistant and thermoresistant cancer cell lines will be discussed. Finally, the potential roles of newly identified factors in a distinct biological mechanism have to be proven by functional studies. This experimental validation strategy will be discussed for two different factors identified by 2D-PAGE analyses of drug-resistant carcinoma cell lines, the "transporter associated with antigen presentation 1" (TAP1) and 14-3-3sigma.
Collapse
Affiliation(s)
- Hermann Lage
- Humboldt University Berlin, Charité Campus Mitte, Institute of Pathology, Schumannstr. 20121, D-10117 Berlin, Germany.
| |
Collapse
|
27
|
Craven RA, Stanley AJ, Hanrahan S, Totty N, Jackson DP, Popescu R, Taylor A, Frey J, Selby PJ, Patel PM, Banks RE. Identification of proteins regulated by interferon-? in resistant and sensitive malignant melanoma cell lines. Proteomics 2004; 4:3998-4009. [PMID: 15449380 DOI: 10.1002/pmic.200400870] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Treatment of patients with malignant melanoma with interferon-alpha achieves a response in a small but significant subset of patients. Currently, although much is known about interferon biology, little is known about either the particular mechanisms of interferon-alpha activity that are crucial for response or why only some patients respond to interferon-alpha therapy. Two melanoma cell lines (MeWo and MM418) that are known to differ in their response to the antiproliferative activity of interferon-alpha, have been used as a model system to investigate interferon-alpha action. Using a proteomics approach based on two-dimensional polyacrylamide gel electrophoresis and mass spectrometry, several proteins induced in response to interferon-alpha have been identified. These include a number of gene products previously known to be type I interferon responsive (tryptophanyl tRNA synthetase, leucine aminopeptidase, ubiquitin cross-reactive protein, gelsolin, FUSE binding protein 2 and hPNPase) as well as a number of proteins not previously reported to be induced by type I interferon (cathepsin B, proteasomal activator 28alpha and alpha-SNAP). Although the proteins upregulated by interferon-alpha were common between the cell lines when examined at the level of Western blotting, the disparity in the basal level of cathepsin B was striking, raising the possibility that the higher level in MM418 may contribute to the sensitivity of this cell line to interferon-alpha treatment.
Collapse
Affiliation(s)
- Rachel A Craven
- Cancer Research UK Clinical Centre, St. James's University Hospital, Leeds, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Proteomics represents a novel methodological approach to investigate the expression of all proteins by a cell or organism in its entireness, similar to global strategies for DNA (genomics) and RNA (transcriptomics). This review focuses on the history of protein analysis, which made up the golden age of pancreatic physiology, the current methodology for proteomics (2D gel electrophoresis, mass spectrometry) and the few published experiences with proteomics in the field of pancreatology until now. Finally, potential applications of proteomics for the pancreas, in concert with other techniques, are cited.
Collapse
Affiliation(s)
- Matthias Löhr
- Molecular Gastroenterology, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
29
|
Mocellin S, Rossi CR, Traldi P, Nitti D, Lise M. Molecular oncology in the post-genomic era: the challenge of proteomics. Trends Mol Med 2004; 10:24-32. [PMID: 14720583 DOI: 10.1016/j.molmed.2003.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Simone Mocellin
- Department of Oncological and Surgical Sciences, University of Padua, via Giustiniani 2, 35128 Padua, Italy.
| | | | | | | | | |
Collapse
|
30
|
Huang Y, Sadée W. Drug sensitivity and resistance genes in cancer chemotherapy: a chemogenomics approach. Drug Discov Today 2003; 8:356-63. [PMID: 12681939 DOI: 10.1016/s1359-6446(03)02654-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Resistance to antineoplastic drugs represents a serious obstacle to successful cancer treatment. Genome-wide studies correlating drug response phenotypes with large DNA/tissue microarray and proteomic datasets have been performed to identify the genes and proteins involved in chemosensitivity or drug resistance. The goal is to identify a set of chemosensitivity and/or resistance genes for each drug that are predictive of treatment response. Therefore, validated pharmacogenomic biomarkers offer the potential for the selection of optimal treatment regimens for individual patients and for identifying novel therapeutic targets to overcome drug resistance.
Collapse
Affiliation(s)
- Ying Huang
- Program of Pharmacogenomics, Dept of Pharmacology, College of Medicine and Public Health, The Ohio State University, 5078 Graves Hall, 333 W. 10th Avenue, Columbus, OH 43210, USA
| | | |
Collapse
|
31
|
Tönnies H, Poland J, Sinha P, Lage H. Association of genomic imbalances with drug resistance and thermoresistance in human gastric carcinoma cells. Int J Cancer 2003; 103:752-8. [PMID: 12516094 DOI: 10.1002/ijc.10905] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Therapy resistance is the major obstacle to advances in successful cancer treatment. To characterize chromosomal alterations associated with different types of acquired MDR and thermoresistance, we applied CGH to compare a unique panel of human gastric carcinoma cells consisting of the parental, drug-sensitive and thermosensitive cancer cell line EPG85-257P, the atypical MDR variant EPG85-257RNOV, the classical MDR subline EPG85-257RDB and their thermoresistant counterparts EPG85-257P-TR, EPG85-257RNOV-TR and EPG85-257RDB-TR. CGH with genomic DNA prepared from these cell lines as probes successfully identified genomic gains and/or losses in chromosomal regions encoding putative genes associated with drug resistance and/or thermoresistance. These genes included various members of the families of ABC transporters and molecular chaperones. The importance of these cell variant-specific genomic imbalances in the development of MDR and thermoresistance is discussed and remains to be elucidated.
Collapse
Affiliation(s)
- Holger Tönnies
- Institute of Human Genetics, Humboldt University Berlin, Charité Campus Virchow-Klinikum, Berlin, Germany
| | | | | | | |
Collapse
|
32
|
Danesi R, de Braud F, Fogli S, de Pas TM, Di Paolo A, Curigliano G, Del Tacca M. Pharmacogenetics of anticancer drug sensitivity in non-small cell lung cancer. Pharmacol Rev 2003; 55:57-103. [PMID: 12615954 DOI: 10.1124/pr.55.1.4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In mammalian cells, the process of malignant transformation is characterized by the loss or down-regulation of tumor-suppressor genes and/or the mutation or overexpression of proto-oncogenes, whose products promote dysregulated proliferation of cells and extend their life span. Deregulation in intracellular transduction pathways generates mitogenic signals that promote abnormal cell growth and the acquisition of an undifferentiated phenotype. Genetic abnormalities in cancer have been widely studied to identify those factors predictive of tumor progression, survival, and response to chemotherapeutic agents. Pharmacogenetics has been founded as a science to examine the genetic basis of interindividual variation in drug metabolism, drug targets, and transporters, which result in differences in the efficacy and safety of many therapeutic agents. The traditional pharmacogenetic approach relies on studying sequence variations in candidate genes suspected of affecting drug response. However, these studies have yielded contradictory results because of the small number of molecular determinants of drug response examined, and in several cases this approach was revealed to be reductionistic. This limitation is now being overcome by the use of novel techniques, i.e., high-density DNA and protein arrays, which allow genome- and proteome-wide tumor profiling. Pharmacogenomics represents the natural evolution of pharmacogenetics since it addresses, on a genome-wide basis, the effect of the sum of genetic variants on drug responses of individuals. Development of pharmacogenomics as a new field has accelerated the progress in drug discovery by the identification of novel therapeutic targets by expression profiling at the genomic or proteomic levels. In addition to this, pharmacogenetics and pharmacogenomics provide an important opportunity to select patients who may benefit from the administration of specific agents that best match the genetic profile of the disease, thus allowing maximum activity.
Collapse
Affiliation(s)
- Romano Danesi
- Division of Pharmacology and Chemotherapy, Department of Oncology, Transplants and Advanced Technologies in Medicine, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|