1
|
Del Bufalo D, Damia G. Overview of BH3 mimetics in ovarian cancer. Cancer Treat Rev 2024; 129:102771. [PMID: 38875743 DOI: 10.1016/j.ctrv.2024.102771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024]
Abstract
Ovarian carcinoma is the leading cause of gynecological cancer-related death, still with a dismal five-year prognosis, mainly due to late diagnosis and the emergence of resistance to cytotoxic and targeted agents. Bcl-2 family proteins have a key role in apoptosis and are associated with tumor development/progression and response to therapy in different cancer types, including ovarian carcinoma. In tumors, evasion of apoptosis is a possible mechanism of resistance to therapy. BH3 mimetics are small molecules that occupy the hydrophobic pocket on pro-survival proteins, allowing the induction of apoptosis, and are currently under study as single agents and/or in combination with cytotoxic and targeted agents in solid tumors. Here, we discuss recent advances in targeting anti-apoptotic proteins of the Bcl-2 family for the treatment of ovarian cancer, focusing on BH3 mimetics, and how these approaches could potentially offer an alternative/complementary way to treat patients and overcome or delay resistance to current treatments.
Collapse
Affiliation(s)
- Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy.
| | - Giovanna Damia
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy.
| |
Collapse
|
2
|
Sabokrouh A, Hajivand S, Atabi F. Comparison of anti-cancer effects of platinum ribavirin and ribavirin via telomerase and Bcl-2 gene expression. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3907-3915. [PMID: 37975929 DOI: 10.1007/s00210-023-02841-7] [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: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Among the common treatments for cancers, chemotherapy is widely used. One of the ways to evaluate the effectiveness of anti-cancer drugs is by checking the expression of tumor markers. Hence, this study aimed to evaluate the anti-cancer effects of the newly synthesized platinum ribavirin (Pt-Rb) compared to ribavirin (Rb) through biomarkers. In this study, cell lines were divided into four groups: groups A and B as healthy negative control group and untreated cancer group respectively. Group C and D were treated with, Rb and Pt-Rb, a novel anti-cancer drug, respectively. After evaluating LC50 for the drugs by MTT test, the expression of telomerase and Bcl-2 (B cell lymphoma-2) genes was evaluated using real-time PCR (RT-qPCR). The results showed a significant decrease in telomerase (0.020 ± 0.007) and Bcl-2(0.120 ± 0.005) gene expression in cancer cells treated with Pt-Rb (group D) compared to telomerase (0.040 ± 0.014) and Bcl-2(0.220 ± 0.014) treated with Rb (group C) and also between group D and telomerase (70.76 ± 0.330) and Bcl-2 (99.52 ± 0.670) in group B. The majority of the groups under investigation showed a significant difference (p < 0.05), suggesting that Pt-Rb had stronger anti-cancer effects than Rb and untreated cancer cells. Additionally, Pt-Rb treatment results demonstrated more increased apoptosis than Rb. Our results demonstrated that Pt-Rb is an effective medication in cancer treatment by lowering anti-apoptotic indicators. Therefore, this chemical has the potential to be an effective anti-cancer therapy, pending further research on animal models and then human volunteers.
Collapse
Affiliation(s)
- Abdolreza Sabokrouh
- Department of Biochemistry, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Soheyla Hajivand
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fereshteh Atabi
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Fiveash JB, Ye X, Peerboom DM, Mikkelsen T, Chowdhary S, Rosenfeld M, Lesser GJ, Fisher J, Desideri S, Grossman S, Leopold L, Nabors LB. Clinical trials of R-(-)-gossypol (AT-101) in newly diagnosed and recurrent glioblastoma: NABTT 0602 and NABTT 0702. PLoS One 2024; 19:e0291128. [PMID: 38285688 PMCID: PMC10824421 DOI: 10.1371/journal.pone.0291128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/21/2023] [Indexed: 01/31/2024] Open
Abstract
PURPOSE AT-101 is an oral bcl-2 family protein inhibitor (Bcl-2, Bcl-XL, Mcl-1, Bcl-W) and potent inducer of proapoptotic proteins. A prior study of the parent compound, racemic gossypol, demonstrated objective and durable responses in patients with malignant glioma. AT-101 has demonstrated synergy with radiation in animal models. The objectives of trial NABTT 0602 were to determine the MTD of AT-101 concurrent with temozolomide (TMZ) and radiation therapy (RT) (Arm I) and to determine the MTD of AT-101 when given with adjuvant TMZ after completion of standard chemoradiation (Arm 2). Separately in trial NABTT 0702, the survival and response rates of single agent AT-101 were evaluated in patients with recurrent glioblastoma. METHODS In NABTT 0602 Phase I, a 3+3 design was used to define MTDs after maximal safe resection, patients with newly diagnosed glioblastoma received standard concurrent RT (60 Gy) and TMZ 75 mg/m2/day followed by adjuvant TMZ 150-200 mg/m2 days 1-5 in 28-day cycles (Stupp regimen). In Arm I, AT-101 was administered M-F during the six weeks of RT beginning 20 mg qd. In Arm 2, concurrent with each adjuvant cycle of TMZ, AT-101 was administered at a starting dose of 20 mg, days 1-21 followed by 7-day break for a maximum of 6 cycles. The PK blood samples were collected in the first three patients in each cohort of arm 1. In NABTT 0702 patients with recurrent glioblastoma received 20 mg p.o. per day for 21 of 28 days in repeated cycles to assess overall survival (OS). RESULTS A total of sixteen patients were enrolled on the two study arms of NABTT 0602. In Arm 1 AT-101 was escalated from 20 to 30 mg where one of six patients experienced DLT (grade 3 GI ulcer). On Arm 2 one patient treated at 20 mg experienced DLT (grade 3 ileus, nausea and diarrhea). The cohort was expanded to include seven patients without observation of DLT. PK results were consistent with drug levels from non-CNS studies. At study closure six patients are still alive. The median survival times for Arm I and Arm II are 15.2 months and 18.2 months, respectively. In NABTT 0702 fifty-six patients were enrolled and forty-three were eligible for imaging response. Sixteen patients (29%) had stable disease as best response and one partial response was observed. The median OS with single agent AT-101 was 5.7 months (95%CI: 3.8-7.6 months) for patients with rGBM. CONCLUSIONS AT-101 can be safely administered with radiation therapy and TMZ in patients with newly diagnosed glioblastoma without toxicity unique to patients with CNS tumors. Because of toxicity observed in non-CNS AT-101 clinical trials, further dose-escalation was not attempted. The recommended dose for future studies that utilize continual AT-101 exposure is 20 mg days M-F concurrent with RT/TMZ and 20 mg days 1-21 for each 28-day cycle of TMZ. AT-101 has limited activity as a single agent in unselected patients with recurrent glioblastoma. Future trials should attempt to better understand resistance mechanisms and consider combination therapy.
Collapse
Affiliation(s)
- John B. Fiveash
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Xiaobu Ye
- Departments of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - David M. Peerboom
- Cleveland Clinic Brain Tumor and Neuro-Oncology Center, Cleveland, Ohio, United States of America
| | - Tom Mikkelsen
- Henry Ford Hospital Hermelin Brain Center, Michigan, Indiana, United States of America
| | | | - Myrna Rosenfeld
- University of Pennsylvania Department of Neurology, Philadelphia, Pennsylvania, United States of America
| | - Glenn J. Lesser
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Joy Fisher
- Departments of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Serena Desideri
- Departments of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Stuart Grossman
- Departments of Neurosurgery and Oncology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Lance Leopold
- Incyte, Wilmington, Delaware, United States of America
| | - Louis B. Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
4
|
Bondos SE, Geraldo Mendes G, Jons A. Context-dependent HOX transcription factor function in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 174:225-262. [PMID: 32828467 DOI: 10.1016/bs.pmbts.2020.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During animal development, HOX transcription factors determine the fate of developing tissues to generate diverse organs and appendages. The power of these proteins is striking: mis-expressing a HOX protein causes homeotic transformation of one body part into another. During development, HOX proteins interpret their cellular context through protein interactions, alternative splicing, and post-translational modifications to regulate cell proliferation, cell death, cell migration, cell differentiation, and angiogenesis. Although mutation and/or mis-expression of HOX proteins during development can be lethal, changes in HOX proteins that do not pattern vital organs can result in survivable malformations. In adults, mutation and/or mis-expression of HOX proteins disrupts their gene regulatory networks, deregulating cell behaviors and leading to arthritis and cancer. On the molecular level, HOX proteins are composed of DNA binding homeodomain, and large regions of unstructured, or intrinsically disordered, protein sequence. The primary roles of HOX proteins in arthritis and cancer suggest that mutations associated with these diseases in both the structured and disordered regions of HOX proteins can have substantial functional effects. These insights lead to new questions critical for understanding and manipulating HOX function in physiological and pathological conditions.
Collapse
Affiliation(s)
- Sarah E Bondos
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States.
| | - Gabriela Geraldo Mendes
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Amanda Jons
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| |
Collapse
|
5
|
Zhao W, Li H, Yang S, Guo D, Chen J, Miao S, Xin Y, Liang M. MicroRNA-152 suppresses cisplatin resistance in A549 cells. Oncol Lett 2019; 18:4613-4620. [PMID: 31611969 PMCID: PMC6781694 DOI: 10.3892/ol.2019.10834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to investigate the association between microRNA-152 and cisplatin resistance in non-small cell lung cancer. A549 and cisplatin-resistant A549 cells (A549/cis) were maintained in vitro. Reverse transcription-quantitative PCR (RT-qPCR) was performed to analyze differences in microRNA-152 levels between A549 and A549/cis cells, and changes in Bcl-2 and NF-κB expression levels were analyzed via RT-qPCR and western blot analyses. MicroRNA-152 was overexpressed in A549/cis cells via transfection of a microRNA-152 mimic. Upon treating transfected or untransfected A549/cis cells with 2 µg/l cisplatin for 24 h, a Cell Counting Kit-8 assay, morphological analysis and flow cytometry analysis were performed to evaluate the effect of microRNA-152 on the inhibition of cell proliferation and induction of apoptosis. Furthermore, changes in Bcl-2 and NF-κB expression levels in microRNA-152-overexpressing A549/cis cells were also analyzed. MicroRNA-152 was significantly downregulated and Bcl-2 and NF-κB were significantly upregulated in A549/cis cells (P<0.05). MicroRNA-152 upregulation enhanced the inhibitory effect of cisplatin on A549/cis cells. These results suggest that microRNA-152 downregulates Bcl-2 and NF-κB. MicroRNA-152 downregulation may induce cisplatin resistance in non-small cell lung cancer cells, whereas microRNA-152 upregulation may improve cisplatin sensitivity among A549/cis cells via downregulation of Bcl-2 and NF-κB.
Collapse
Affiliation(s)
- Wenfei Zhao
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongyun Li
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shanshan Yang
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Di Guo
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jing Chen
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shaoyi Miao
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Xin
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Miaomiao Liang
- Department of Respiratory, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
6
|
Jia J, Dai S, Sun X, Sang Y, Xu Z, Zhang J, Cui X, Song J, Guo X. A preliminary study of the effect of ECRG4 overexpression on the proliferation and apoptosis of human laryngeal cancer cells and the underlying mechanisms. Mol Med Rep 2015; 12:5058-64. [PMID: 26165988 PMCID: PMC4581775 DOI: 10.3892/mmr.2015.4059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/03/2015] [Indexed: 12/18/2022] Open
Abstract
Human esophageal cancer-related gene 4 (ECRG4) is a potential tumor suppressor gene isolated from human esophageal epithelial cells. Studies have shown that ECRG4 effectively inhibits the proliferation of tumor cells and induces apoptosis. However, the role of ECRG4 in laryngeal cancer has not yet been clearly defined. In this study, a human laryngeal cancer cell line stably overexpressing ECRG4 was established. The effect of ECRG4 on the proliferation and apoptosis of laryngeal cancer cells and the associated mechanisms were investigated. The Hep-2 human laryngeal carcinoma cell line exhibited a low basal level of ECRG4 expression and was selected for the present study. The eukaryotic expression plasmid pcDNA3.1-ECRG4 was constructed and introduced into Hep-2 cells by transfection reagents. Western blot analysis, reverse transcription-quantitative polymerase chain reaction and immunofluorescence staining confirmed high-level expression of ECRG4. The 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assay showed that ECRG4 over-expression suppressed the proliferative capacity of laryngeal cancer cells in vitro. Cell cycle analysis showed that ECRG4 induced cell cycle arrest at the G0/G1 phase. Flow cytometric analysis and Hoechst staining demonstrated that overexpres-sion of ECRG4 significantly induced apoptosis. Western blot analysis confirmed that Bcl-2-associated X protein, cleaved-caspase-3 and cleaved-poly (ADP-ribose) polymerase were upregulated in the apoptotic process, whereas B-cell lymphoma 2 was downregulated. In conclusion, overexpression of ECRG4 inhibited laryngeal cancer cell proliferation and induced cancer cell apoptosis. Therefore, ECRG4 exhibits potential as an effective target in gene therapy for laryngeal cancer.
Collapse
Affiliation(s)
- Jianping Jia
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Song Dai
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xinghe Sun
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Yuehong Sang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Zhenming Xu
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jie Zhang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xiaofeng Cui
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jinhui Song
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xing Guo
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
7
|
Villalobos X, Rodríguez L, Solé A, Lliberós C, Mencia N, Ciudad CJ, Noé V. Effect of Polypurine Reverse Hoogsteen Hairpins on Relevant Cancer Target Genes in Different Human Cell Lines. Nucleic Acid Ther 2015; 25:198-208. [PMID: 26042602 DOI: 10.1089/nat.2015.0531] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We studied the ability of polypurine reverse Hoogsteen hairpins (PPRHs) to silence a variety of relevant cancer-related genes in several human cell lines. PPRHs are hairpins formed by two antiparallel polypurine strands bound by intramolecular Hoogsteen bonds linked by a pentathymidine loop. These hairpins are able to bind to their target DNA sequence through Watson-Crick bonds producing specific silencing of gene expression. We designed PPRHs against the following genes: BCL2, TOP1, mTOR, MDM2, and MYC and tested them for mRNA levels, cytotoxicity, and apoptosis in prostate, pancreas, colon, and breast cancer cell lines. Even though all PPRHs were effective, the most remarkable results were obtained with those against BCL2 and mammalian target of rapamycin (mTOR) in decreasing cell survival and mRNA levels and increasing apoptosis in prostate, colon, and pancreatic cancer cells. In the case of TOP1, MDM2, and MYC, their corresponding PPRHs produced a strong effect in decreasing cell viability and mRNA levels and increasing apoptosis in breast cancer cells. Thus, we confirm that the PPRH technology is broadly useful to silence the expression of cancer-related genes as demonstrated using target genes involved in metabolism (DHFR), proliferation (mTOR), DNA topology (TOP1), lifespan and senescence (telomerase), apoptosis (survivin, BCL2), transcription factors (MYC), and proto-oncogenes (MDM2).
Collapse
Affiliation(s)
- Xenia Villalobos
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Laura Rodríguez
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Anna Solé
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Carolina Lliberós
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Núria Mencia
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Carlos J Ciudad
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| | - Véronique Noé
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Barcelona , Barcelona, Spain
| |
Collapse
|
8
|
Pojo M, Gonçalves CS, Xavier-Magalhães A, Oliveira AI, Gonçalves T, Correia S, Rodrigues AJ, Costa S, Pinto L, Pinto AA, Lopes JM, Reis RM, Rocha M, Sousa N, Costa BM. A transcriptomic signature mediated by HOXA9 promotes human glioblastoma initiation, aggressiveness and resistance to temozolomide. Oncotarget 2015; 6:7657-74. [PMID: 25762636 PMCID: PMC4480707 DOI: 10.18632/oncotarget.3150] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/16/2015] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma is the most malignant brain tumor, exhibiting remarkable resistance to treatment. Here we investigated the oncogenic potential of HOXA9 in gliomagenesis, the molecular and cellular mechanisms by which HOXA9 renders glioblastoma more aggressive, and how HOXA9 affects response to chemotherapy and survival. The prognostic value of HOXA9 in glioblastoma patients was validated in two large datasets from TCGA and Rembrandt, where high HOXA9 levels were associated with shorter survival. Transcriptomic analyses identified novel HOXA9-target genes with key roles in cancer-related processes, including cell proliferation, DNA repair, and stem cell maintenance. Functional studies with HOXA9-overexpressing and HOXA9-silenced glioblastoma cell models revealed that HOXA9 promotes cell viability, stemness and invasion, and inhibits apoptosis. Additionally, HOXA9 promoted the malignant transformation of human immortalized astrocytes in an orthotopic in vivo model, and caused tumor-associated death. HOXA9 also mediated resistance to temozolomide treatment in vitro and in vivo via upregulation of BCL2. Importantly, the pharmacological inhibition of BCL2 with the BH3 mimetic ABT-737 reverted temozolomide resistance in HOXA9-positive cells. These data establish HOXA9 as a driver of glioma initiation, aggressiveness and resistance to therapy. In the future, the combination of BH3 mimetics with temozolomide should be further explored as an alternative treatment for glioblastoma.
Collapse
Affiliation(s)
- Marta Pojo
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Ana Xavier-Magalhães
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Ana Isabel Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Tiago Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Sara Correia
- Centre of Biological Engineering/Department of Informatics, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
| | - Ana J. Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Sandra Costa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Afonso A. Pinto
- Department of Neurosurgery, Hospital de Braga, Sete Fontes, 4710-243 São Victor, Braga, Portugal
| | - José M. Lopes
- Department of Pathology, Hospital S. João, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Molecular Pathology and Immunology at the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n 4200-465 Porto, Portugal
- Medical Faculty, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Rui M. Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
- Barretos Cancer Hospital, Molecular Oncology Research Center, Rua Antenor Duarte Vilela, 1331 - Doutor Paulo Prata, Barretos - SP, 14780-000, Brasil
| | - Miguel Rocha
- Centre of Biological Engineering/Department of Informatics, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar 4710-057 Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Campus de Gualtar 4710-057 Braga, Portugal
| |
Collapse
|
9
|
Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, Bartsch G, Michaelis M, Cinatl J, Haferkamp A, Kögel D. Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (-)-Gossypol (AT-101). BMC Cancer 2015; 15:224. [PMID: 25885284 PMCID: PMC4409725 DOI: 10.1186/s12885-015-1239-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 03/20/2015] [Indexed: 12/19/2022] Open
Abstract
Background Acquired resistance to standard chemotherapy causes treatment failure in patients with metastatic bladder cancer. Overexpression of pro-survival Bcl-2 family proteins has been associated with a poor chemotherapeutic response, suggesting that Bcl-2-targeted therapy may be a feasible strategy in patients with these tumors. The small-molecule pan-Bcl-2 inhibitor (−)-gossypol (AT-101) is known to induce apoptotic cell death, but can also induce autophagy through release of the pro-autophagic BH3 only protein Beclin-1 from Bcl-2. The potential therapeutic effects of (−)-gossypol in chemoresistant bladder cancer and the role of autophagy in this context are hitherto unknown. Methods Cisplatin (5637rCDDP1000, RT4rCDDP1000) and gemcitabine (5637rGEMCI20, RT4rGEMCI20) chemoresistant sub-lines of the chemo-sensitive bladder cancer cell lines 5637 and RT4 were established for the investigation of acquired resistance mechanisms. Cell lines carrying a stable lentiviral knockdown of the core autophagy regulator ATG5 were created from chemosensitive 5637 and chemoresistant 5637rGEMCI20 and 5637rCDDP1000 cell lines. Cell death and autophagy were quantified by FACS analysis of propidium iodide, Annexin and Lysotracker staining, as well as LC3 translocation. Results Here we demonstrate that (−)-gossypol induces an apoptotic type of cell death in 5637 and RT4 cells which is partially inhibited by the pan-caspase inhibitor z-VAD. Cisplatin- and gemcitabine-resistant bladder cancer cells exhibit enhanced basal and drug-induced autophagosome formation and lysosomal activity which is accompanied by an attenuated apoptotic cell death after treatment with both (−)-gossypol and ABT-737, a Bcl-2 inhibitor which spares Mcl-1, in comparison to parental cells. Knockdown of ATG5 and inhibition of autophagy by 3-MA had no discernible effect on apoptotic cell death induced by (−)-gossypol and ABT-737 in parental 5637 cells, but evoked a significant increase in early apoptosis and overall cell death in BH3 mimetic-treated 5637rGEMCI20 and 5637rCDDP1000 cells. Conclusions Our findings show for the first time that (−)-gossypol concomitantly triggers apoptosis and a cytoprotective type of autophagy in bladder cancer and support the notion that enhanced autophagy may underlie the chemoresistant phenotype of these tumors. Simultaneous targeting of Bcl-2 proteins and the autophagy pathway may be an efficient new strategy to overcome their “autophagy addiction” and acquired resistance to current therapy.
Collapse
Affiliation(s)
- Jens Mani
- Department of Urology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Stefan Vallo
- Department of Urology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Stefanie Rakel
- Experimental Neurosurgery, Neuroscience Center, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Patrick Antonietti
- Experimental Neurosurgery, Neuroscience Center, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Florian Gessler
- Experimental Neurosurgery, Neuroscience Center, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Roman Blaheta
- Department of Urology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Georg Bartsch
- Department of Urology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Martin Michaelis
- Institute for Medical Virology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany. .,School of Biosciences, The University of Kent, Canterbury, Kent, CT2 7NZ, UK.
| | - Jindrich Cinatl
- Institute for Medical Virology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Axel Haferkamp
- Department of Urology, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| | - Donat Kögel
- Experimental Neurosurgery, Neuroscience Center, Goethe University Hospital, Theodor-Stern-Kai 7, D-60590, Frankfurt am Main, Germany.
| |
Collapse
|
10
|
Gao J, Yan Q, Liu S, Yang X. Knockdown of EpCAM enhances the chemosensitivity of breast cancer cells to 5-fluorouracil by downregulating the antiapoptotic factor Bcl-2. PLoS One 2014; 9:e102590. [PMID: 25019346 PMCID: PMC4097402 DOI: 10.1371/journal.pone.0102590] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/19/2014] [Indexed: 12/19/2022] Open
Abstract
Resistance to fluoropyrimidine-based chemotherapy is the main reason for the failure of cancer treatment, and drug resistance is associated with an inability of tumor cells to undergo apoptosis in response to treatment. Alterations in the expression of epithelial cell adhesion molecule (EpCAM) affect the sensitivity or resistance of tumor cells to anticancer treatment and the activity of intracellular signaling pathways. However, the role of EpCAM in the induction of apoptosis in breast cancer cells remains unclear. Here, we investigated the effect of EpCAM gene knockdown on chemosensitivity to 5-fluorouracil (5-FU) in MCF-7 cells and explored the underlying mechanisms. Our results showed that knockdown of EpCAM promoted apoptosis, inhibited cell proliferation and caused cell-cycle arrest. EpCAM knockdown enhanced the cytotoxic effect of 5-FU, promoting apoptosis by downregulating the expression of the anti-apoptotic protein Bcl-2 and upregulating the expression of the pro-apoptotic proteins Bax, and caspase3 via the ERK1/2 and JNK MAPK signaling pathways in MCF-7 cells. These results indicate that knockdown of EpCAM may have a tumor suppressor effect and suggest EpCAM as a potential target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jiujiao Gao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Shuai Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian, People’s Republic of China
- * E-mail:
| |
Collapse
|
11
|
Activated Ras as a Therapeutic Target: Constraints on Directly Targeting Ras Isoforms and Wild-Type versus Mutated Proteins. ISRN ONCOLOGY 2013; 2013:536529. [PMID: 24294527 PMCID: PMC3833460 DOI: 10.1155/2013/536529] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 10/04/2013] [Indexed: 12/12/2022]
Abstract
The ability to selectively and directly target activated Ras would provide immense utility for treatment of the numerous cancers that are driven by oncogenic Ras mutations. Patients with disorders driven by overactivated wild-type Ras proteins, such as type 1 neurofibromatosis, might also benefit from progress made in that context. Activated Ras is an extremely challenging direct drug target due to the inherent difficulties in disrupting the protein:protein interactions that underlie its activation and function. Major investments have been made to target Ras through indirect routes. Inhibition of farnesyl transferase to block Ras maturation has failed in large clinical trials. Likely reasons for this disappointing outcome include the significant and underappreciated differences in the isoforms of Ras. It is still plausible that inhibition of farnesyl transferase will prove effective for disease that is driven by activated H-Ras. The principal current focus of drugs entering clinic trial is inhibition of pathways downstream of activated Ras, for example, trametinib, a first-in-class MEK inhibitor. The complexity of signaling that is driven by activated Ras indicates that effective inhibition of oncogenic transduction through this approach will be difficult, with resistance being likely to emerge through switch to parallel pathways. Durable disease responses will probably require combinatorial block of several downstream targets.
Collapse
|
12
|
Liao J, Xu T, Zheng JX, Lin JM, Cai QY, Yu DB, Peng J. Nitidine chloride inhibits hepatocellular carcinoma cell growth in vivo through the suppression of the JAK1/STAT3 signaling pathway. Int J Mol Med 2013; 32:79-84. [PMID: 23613111 DOI: 10.3892/ijmm.2013.1358] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/15/2013] [Indexed: 11/06/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is persistently activated in cancer cells and contributes to malignant progression in various types of cancer. The Janus-activated kinase (JAK) family phosphorylates STAT3 in response to stimulation by cytokines or growth factors. The JAK1-STAT3 signaling pathway plays an important role in cell proliferation and apoptosis. Nitidine chloride (NC) is a benzophenanthridine alkaloid that has been reported as an antitumor agent due to its its inhibitory effects on topoisomerase I. Using a mouse xenograft model of hepatocellular carcinoma (HCC), this study aimed to evaluate the effects of NC on tumor growth in vivo and to elucidate the underlying mechanisms. The analysis of the effects of NC on apoptosis in HCC tumor xenografts in mice was carried out by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assay; the expression of Bcl-2, Bax, cyclin-dependent kinase (CDK)4, cyclin D1, p21 and proliferating cell nuclear antigen (PCNA) was analyzed by immunohistochemistry; and the protein expression of JAK1 and STAT3 was examined by western blot analysis. Our results revealed that treatment with NC decreased the tumor volume and tumor weight, suggesting that NC inhibits HCC cell growth in vivo. In addition, NC blocked the activation of JAK1-STAT3 in the tumor tissues, which in turn resulted in the induction of cancer cell apoptosis and the inhibition of proliferation. Consequently, treatment with NC downregulated the expression of cyclin D1, CDK4 and Bcl-2 and increased the level of p21 and Bax. Our data provide a molecular basis for the antitumor activity of NC.
Collapse
Affiliation(s)
- Jun Liao
- Department of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.
| | | | | | | | | | | | | |
Collapse
|