1
|
Yeh WC, Tu YC, Chien TC, Hsu PL, Lee CW, Wu SY, Pan BS, Yu HH, Su BC. Vismodegib Potentiates Marine Antimicrobial Peptide Tilapia Piscidin 4-Induced Cytotoxicity in Human Non-Small Cell Lung Cancer Cells. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10282-8. [PMID: 38743208 DOI: 10.1007/s12602-024-10282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
Non-small cell lung cancer (NSCLC) is a common cancer with several accepted treatments, such as chemotherapy, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, and immune checkpoint inhibitors. Nevertheless, NSCLC cells often become insensitive to these treatments, and therapeutic resistance is a major reason NSCLC still has a high mortality rate. The induction of therapeutic resistance in NSCLC often involves hedgehog, and suppression of hedgehog can increase NSCLC cell sensitivity to several conventional therapies. In our previous work, we demonstrated that the marine antimicrobial peptide tilapia piscidin 4 (TP4) exhibits potent anti-NSCLC activity in both EGFR-WT and EGFR-mutant NSCLC cells. Here, we sought to further explore whether hedgehog might influence the sensitivity of NSCLC cells to TP4. Our results showed that hedgehog was activated by TP4 in both WT and EGFR-mutant NSCLC cells and that pharmacological inhibition of hedgehog by vismodegib, a Food and Drug Administration-approved hedgehog inhibitor, potentiated TP4-induced cytotoxicity. Mechanistically, vismodegib acted by enhancing TP4-mediated increases in mitochondrial membrane potential and intracellular reactive oxygen species (ROS). MitoTempo, a specific mitochondrial ROS scavenger, abolished vismodegib/TP4 cytotoxicity. The combination of vismodegib with TP4 also reduced the levels of the antioxidant proteins catalase and superoxide dismutase, and it diminished the levels of chemoresistance-related proteins, Bcl-2 and p21. Thus, we conclude that hedgehog regulates the cytotoxic sensitivity of NSCLC cells to TP4 by protecting against mitochondrial dysfunction and suppressing oxidative stress. These findings suggest that combined treatment of vismodegib and TP4 may be a promising therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Wei-Chen Yeh
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun-Chieh Tu
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Cheng Chien
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pei-Ling Hsu
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Chu-Wan Lee
- Department of Nursing, National Tainan Junior College of Nursing, 78, Section 2, Minzu Road, West Central District, Tainan, 70007, Taiwan
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27157, USA
| | - Bo-Syong Pan
- Department of Pathology, Duke University School of Medicine, Durham, NC27710, USA
| | - Hsin-Hsien Yu
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Paget C, Duret H, Ngiow SF, Kansara M, Thomas DM, Smyth MJ. Studying the role of the immune system on the antitumor activity of a Hedgehog inhibitor against murine osteosarcoma. Oncoimmunology 2021; 1:1313-1322. [PMID: 23243595 PMCID: PMC3518504 DOI: 10.4161/onci.21680] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent evidence demonstrates that the efficacy of conventional anticancer therapies including chemotherapy requires a functional immune system. Here, we addressed the possibility that the antitumor effect of a selective Smoothened antagonist and Hedgehog (Hh) pathway inhibitor (LDE225), a promising anticancer drug, might at least partially depend on the immune system. To this aim, we used tumor cell lines derived from a murine model of radiation-induced osteosarcoma. In vitro treatment of osteosarcoma cells with LDE225 resulted in a decreased ability of tumor cells to proliferate, but had no effect on their viability. Flow cytometry analysis demonstrated that LDE225-treatment did not detectably modulate the immunogenicity of tumor cells. Moreover, LDE225 did not display any pro-apoptotic properties on osteosarcoma cells, highlighting that its antitumor profile mainly derives from a cytostatic effect. Furthermore, calreticulin exposure, a key feature of immunogenic cell death, was not provoked by LDE225, neither alone nor combined with recognized immunogenic drugs. Finally, the oral administration of LDE225 to osteosarcoma-bearing mice did significantly delay the tumor growth even in an immunocompromised setting. These data suggest that inhibiting Hh signaling can control osteosarcoma cell proliferation but does not modulate the immunogenic profile of these cells.
Collapse
Affiliation(s)
- Christophe Paget
- Cancer Immunology Program; Peter MacCallum Cancer Centre; East Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
3
|
O'Hara SP, Splinter PL, Trussoni CE, Guicciardi ME, Splinter NP, Al Suraih MS, Nasser-Ghodsi N, Stollenwerk D, Gores GJ, LaRusso NF. The transcription factor ETS1 promotes apoptosis resistance of senescent cholangiocytes by epigenetically up-regulating the apoptosis suppressor BCL2L1. J Biol Chem 2019; 294:18698-18713. [PMID: 31659122 PMCID: PMC6901313 DOI: 10.1074/jbc.ra119.010176] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is an idiopathic, progressive cholangiopathy. Cholangiocyte senescence is important in PSC pathogenesis, and we have previously reported that senescence is regulated by the transcription factor ETS proto-oncogene 1 (ETS1) and associated with overexpression of BCL2 like 1 (BCL2L1 or BCL-xL), an anti-apoptotic BCL2-family member. Here, we further explored the mechanisms regulating BCL-xL-mediated, apoptosis resistance in senescent cholangiocytes and uncovered that ETS1 and the histone acetyltransferase E1A-binding protein P300 (EP300 or p300) both promote BCL-xL transcription. Using immunofluorescence, we found that BCL-xL protein expression is increased both in cholangiocytes of livers from individuals with PSC and a mouse model of PSC. Using an in vitro model of lipopolysaccharide-induced senescence in normal human cholangiocytes (NHCs), we found increased BCL-xL mRNA and protein levels, and ChIP-PCRs indicated increased occupancy of ETS1, p300, and histone 3 Lys-27 acetylation (H3K27Ac) at the BCL-xL promoter. Using co-immunoprecipitation and proximity ligation assays, we further demonstrate that ETS1 and p300 physically interact in senescent but not control NHCs. Additionally, mutagenesis of predicted ETS1-binding sites within the BCL-xL promoter blocked luciferase reporter activity, and CRISPR/Cas9-mediated genetic deletion of ETS1 reduced senescence-associated BCL-xL expression. In senescent NHCs, TRAIL-mediated apoptosis was reduced ∼70%, and ETS1 deletion or RNAi-mediated BCL-xL suppression increased apoptosis. Overall, our results suggest that ETS1 and p300 promote senescent cholangiocyte resistance to apoptosis by modifying chromatin and inducing BCL-xL expression. These findings reveal ETS1 as a central regulator of both cholangiocyte senescence and the associated apoptosis-resistant phenotype.
Collapse
Affiliation(s)
- Steven P O'Hara
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905.
| | - Patrick L Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah P Splinter
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Mohammed S Al Suraih
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Navine Nasser-Ghodsi
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Deborah Stollenwerk
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
4
|
Shih S, Dai C, Ansari A, Urso BA, Laughlin AI, Solomon JA. Advances in genetic understanding of gorlin syndrome and emerging treatment options. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1483233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shawn Shih
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Christina Dai
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Ahmed Ansari
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Brittany A Urso
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
| | - Amy I Laughlin
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James A Solomon
- Department of Dermatology, University of Central Florida College of Medicine, Orlando, Florida
- University of Illinois College of Medicine, Urbana, Illinois
- Ameriderm Research, Ormond Beach, FL
- Department of Dermatology, Florida State University College of Medicine, Tallahassee, FL
| |
Collapse
|
5
|
Juntermanns B, Kaiser GM, Orth L, Reis H, Jaradat D, Sydor S, Buechter M, Kasper S, Mathé Z, Sotiropoulos GC, Baba HA, Canbay A, Paul A, Fingas CD. Comparison of the sixth and the seventh editions of the UICC classification for intrahepatic cholangiocarcinoma. Eur J Med Res 2018; 23:29. [PMID: 29859128 PMCID: PMC5984789 DOI: 10.1186/s40001-018-0329-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 05/26/2018] [Indexed: 12/13/2022] Open
Abstract
Background The current seventh edition of the TNM classification for intrahepatic cholangiocarcinoma (ICC) includes tumor number, vascular invasion, lymph node involvement but no longer the tumor size as compared to the sixth edition. The impact of the seventh edition on stage-based prognostic prediction for patients with ICC was evaluated. Methods Between 03/2001 and 02/2013, 98 patients with the diagnosis of an ICC were surgically treated at our center. Median survival times were calculated for these patients after separate classification by both sixth and seventh editions. Results Median overall survival was increased in patients classified to the lower tumor stages I and II using the seventh as compared to the sixth edition: stage I (54.9 vs. 47.3 months), stage II (19.9 vs. 18.9 months), stage III (17.2 vs. 19.9 months), and stage IV (23.2 vs. 15.3 months), respectively. The seventh edition definition of the T category resulted in an increased median survival regarding the T1 (50.4 vs. 47.3 months) as well as the T2 category (19.9 vs. 15.6 months) and revealed a reduced median survival of patients within the T3 (21.6 vs. 24.8 months) as well as the T4 category (19.9 vs. 27.0 months). Conclusions The UICC seventh edition TNM classification for ICC improves separation of patients with intermediate stage tumors as compared to the sixth edition. The prognostic value of the UICC staging system has been improved by the seventh edition. Trial registration The data for this study have been retrospectively registered and the study has been approved by the ethic committee of the medical faculty of the University Hospital of Essen, Germany (license number 15-6353-BO).
Collapse
Affiliation(s)
- Benjamin Juntermanns
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Gernot Maximilian Kaiser
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany. .,Department of General and Visceral Surgery, St. Bernhard-Hospital, Bürgermeister-Schmelzing-Str. 90, 47475, Kamp-Lintfort, Germany.
| | - Lena Orth
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Henning Reis
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Derar Jaradat
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Svenja Sydor
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Matthias Buechter
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefan Kasper
- Department of Medical Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Zoltan Mathé
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Georgios Charalambos Sotiropoulos
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Hideo Andreas Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Paul
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Christian Dominik Fingas
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
6
|
Fritsch J, Zingler P, Särchen V, Heck AL, Schütze S. Role of ubiquitination and proteolysis in the regulation of pro- and anti-apoptotic TNF-R1 signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2138-2146. [PMID: 28765050 DOI: 10.1016/j.bbamcr.2017.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Tumor Necrosis Factor Receptor 1 (TNF-R1) transmits various intracellular signaling cascades leading to diverse biological outcomes, ranging from proliferation, differentiation, survival to the induction of various forms of cell death (i.e. apoptosis, necrosis, necroptosis). These signaling pathways have to be tightly regulated. Proteolysis is an important regulatory mechanism in TNF-R1 pro-apoptotic as well as anti-apoptotic/pro-inflammatory signaling. Some key players in these signaling cascades are known (mainly the caspase-family of proteases and a previously unrecognized "lysosomal death pathway" involving cathepsins), however the interaction of proteases in the regulation of TNF signaling is still enigmatic. Ubiquitination of proteins, both non-degradative degradative, which either results in proteolytic degradation of target substrates or regulates their biological function, represents another layer of regulation in this signaling cascade. We and others found out that the differences in signal quality depend on the localization of the receptors. Plasma membrane resident receptors activate survival signals, while endocytosed receptors can induce cell death. In this article we will review the role of ubiquitination and proteolysis in these diverse events focusing on our own contributions to the lysosomal apoptotic pathway linked to the subcellular compartmentalization of TNF-R1. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
Affiliation(s)
- Jürgen Fritsch
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Philipp Zingler
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Vinzenz Särchen
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Anna Laura Heck
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Stefan Schütze
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany.
| |
Collapse
|
7
|
Sydor S, Jafoui S, Wingerter L, Swoboda S, Mertens JC, Gerken G, Canbay A, Paul A, Fingas CD. Bcl-2 degradation is an additional pro-apoptotic effect of polo-like kinase inhibition in cholangiocarcinoma cells. World J Gastroenterol 2017; 23:4007-4015. [PMID: 28652654 PMCID: PMC5473120 DOI: 10.3748/wjg.v23.i22.4007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/05/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To examine the influence on apoptotic mechanisms following inhibition of polo-like kinases as therapeutically approach for cholangiocellular cancer treatment. METHODS As most cholangiocarcinomas are chemotherapy-resistant due to mechanisms preventing tumor cell death, we investigated the effect of Cisplatin on cholangiocellular carcinoma (CCA) cell lines KMCH-1 and Mz-Ch-1. Polo-like kinases (PLK) are important regulators of the cell cycle and their inhibition is discussed as a potential therapy while PLK inhibition can regulate apoptotic mediators. Here, cells were treated with PLK inhibitor BI6727 (Volasertib), Cisplatin, and in combination of both compounds. Cell viability was assessed by MTT; apoptosis was measured by DAPI staining and caspase-3/-7 assay. Western blot and qRT-PCR were used to measure expression levels of apoptosis-related molecules Bax and Bcl-2. RESULTS The cell viability in the CCA cell lines KMCH-1 and Mz-Ch-1 was reduced in all treatment conditions compared to vehicle-treated cells. Co-treatment with BI6727 and cisplatin could even enhance the cytotoxic effect of cisplatin single treatment. Thus, co-treatment of cisplatin with BI6727 could slightly enhance the cytotoxic effect of the cisplatin in both cell lines whereas there was evidence of increased apoptosis induction solely in Mz-Ch-1 as compared to KMCH-1. Moreover, PLK inhibition decreases protein levels of Bcl-2; an effect that can be reversed by the proteasomal degradation inhibitor MG-132. In contrast, protein levels of Bax were not found to be altered by PLK inhibition. These findings indicate that cytotoxic effects of Cisplatin in Mz-Ch-1 cells can be enhanced by cotreatment with BI6727. CONCLUSION In conclusion, BI6727 treatment can sensitize CCA cells to cisplatin-induced apoptosis with proteasomal Bcl-2 degradation as an additional pro-apoptotic effect.
Collapse
|
8
|
Naoum GE, Buchsbaum DJ, Tawadros F, Farooqi A, Arafat WO. Journey of TRAIL from Bench to Bedside and its Potential Role in Immuno-Oncology. Oncol Rev 2017; 11:332. [PMID: 28584572 PMCID: PMC5432952 DOI: 10.4081/oncol.2017.332] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/17/2017] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
Induction of apoptosis in cancer cells has increasingly been the focus of many therapeutic approaches in oncology field. Since its identification as a TNF family member, TRAIL (TNF-related apoptosis-inducing ligand) paved a new path in apoptosis inducing cancer therapies. Its selective ability to activate extrinsic and intrinsic cell death pathways in cancer cells only, independently from p53 mutations responsible for conventional therapeutics resistance, spotted TRAIL as a potent cancer apoptotic agent. Many recombinant preparations of TRAIL and death receptor targeting monoclonal antibodies have been developed and being tested pre-clinically and clinically both as a single agent and in combinations. Of note, the monoclonal antibodies were not the only type of antibodies developed to target TRAIL receptors. Recent technology has brought forth several single chain variable domains (scFv) designs fused recombinantly to TRAIL as well. Also, it is becoming progressively more understandable that field of nanotechnology has revolutionized cancer diagnosis and therapy. The recent breakthroughs in materials science and protein engineering have helped considerably in strategically loading drugs into nanoparticles or conjugating drugs to their surface. In this review we aim to comprehensively highlight the molecular knowledge of TRAIL in the context of its pathway, receptors and resistance factors. We also aim to review the clinical trials that have been done using TRAIL based therapies and to review various scFv designs, the arsenal of nano-carriers and molecules available to selectively target tumor cells with TRAIL.
Collapse
Affiliation(s)
| | | | | | | | - Waleed O. Arafat
- Alexandria Comprehensive Cancer Center, Alexandria, Egypt
- Univeristy of Alabama, Birmingham, AL, USA
- University of Alexandria, Faculty of Medicine, Egypt
| |
Collapse
|
9
|
Naoum GE, Tawadros F, Farooqi AA, Qureshi MZ, Tabassum S, Buchsbaum DJ, Arafat W. Role of nanotechnology and gene delivery systems in TRAIL-based therapies. Ecancermedicalscience 2016; 10:660. [PMID: 27594905 PMCID: PMC4990059 DOI: 10.3332/ecancer.2016.660] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
Since its identification as a member of the tumour necrosis factor (TNF) family, TRAIL (TNF-related apoptosis-inducing ligand) has emerged as a new avenue in apoptosis-inducing cancer therapies. Its ability to circumvent the chemoresistance of conventional therapeutics and to interact with cancer stem cells (CSCs) self-renewal pathways, amplified its potential as a cancer apoptotic agent. Many recombinant preparations of this death ligand and monoclonal antibodies targeting its death receptors have been tested in monotherapy and combinational clinical trials. Gene therapy is a new approach for cancer treatment which implies viral or non-viral functional transgene induction of apoptosis in cancer cells or repair of the underlying genetic abnormality on a molecular level. The role of this approach in overcoming the traditional barriers of radiation and chemotherapeutics systemic toxicity, risk of recurrence, and metastasis made it a promising platform for cancer treatment. The recent first Food Drug Administration (FDA) approved oncolytic herpes virus for melanoma treatment brings forth the potency of the cancer gene therapy approach in the future. Many gene delivery systems have been studied for intratumoural TRAIL gene delivery alone or in combination with chemotherapeutic agents to produce synergistic cancer cytotoxicity. However, there still remain many obstacles to be conquered for this different gene delivery systems. Nanomedicine on the other hand offers a new frontier for clinical trials and biomedical research. The FDA approved nanodrugs motivates horizon exploration for other nanoscale designed particles’ implications in gene delivery. In this review we aim to highlight the molecular role of TRAIL in apoptosis and interaction with cancer stem cells (CSCs) self-renewal pathways. Finally, we also aim to discuss the different roles of gene delivery systems, mesenchymal cells, and nanotechnology designs in TRAIL gene delivery.
Collapse
Affiliation(s)
| | - Fady Tawadros
- East Tennessee State University, 1276 Gilbreath Dr, Johnson City, TN 37604, USA
| | | | | | - Sobia Tabassum
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Donald J Buchsbaum
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA
| | - Waleed Arafat
- University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL 35233, USA; University of Alexandria, El-Gaish Rd, Egypt, Alexandria, Egypt
| |
Collapse
|
10
|
Aoto K, Trainor PA. Co-ordinated brain and craniofacial development depend upon Patched1/XIAP regulation of cell survival. Hum Mol Genet 2014; 24:698-713. [PMID: 25292199 DOI: 10.1093/hmg/ddu489] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Congenital brain and craniofacial defects often occur together as a consequence of their developmental dependency on common progenitor tissue interactions and signaling pathways during embryogenesis. A classic example of this is perturbation of midline embryo development, and disruption of Hedgehog (Hh) signaling in the pathogenesis of holoprosencephaly. However, our understanding of how Hh signaling governs cell and tissue survival remains incomplete. Patched1 (Ptch1) is a well-known receptor for Hh ligands and Ptch1 overexpression is associated with cell and tissue-specific apoptosis. Here, we demonstrate that the X-linked inhibitory apoptosis protein (XIAP) associates with the C terminus of Ptch1 (Ptch1-C) in primary cilia to inhibit Ptch1-mediated cell death. Consistent with this observation, inhibition of XIAP suppresses cell proliferation, resulting in cell death and pathogenesis of an Hh loss-of-function phenotype. Thus, co-ordinated development of the brain and face is dependent in part upon XIAP mediation of Hh/Ptch1-regulated cell survival and apoptosis during embryogenesis.
Collapse
Affiliation(s)
- Kazushi Aoto
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA and
| | - Paul A Trainor
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA and Department of Anatomy & Cell Biology, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66202, USA
| |
Collapse
|
11
|
Wan J, Zhou J, Zhao H, Wang M, Wei Z, Gao H, Wang Y, Cui H. Sonic hedgehog pathway contributes to gastric cancer cell growth and proliferation. Biores Open Access 2014; 3:53-9. [PMID: 24804165 PMCID: PMC3995118 DOI: 10.1089/biores.2014.0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Sonic Hedgehog (Shh) signaling pathway is commonly activated in gastrointestinal cancer. However, our understanding of the Shh pathway in gastric cancer remains limited. Here we examined the effects of cyclopamine, a specific inhibitor of the Shh signaling pathway, on cell growth and proliferation in gastric primary cancer cells GAM-016 and the MKN-45 cell line. The results showed that the Shh signaling molecules SHH, PTCH, SMO, GLI1, and GLI2 were intact and activated in both types of cells. Furthermore, we observed that cyclopamine inhibited gastric cancer cell proliferation through cell cycle arrest and apoptosis. An in vivo study using NOD/SCID mouse xenografts demonstrated that cyclopamine significantly prevented tumor growth and development. Our study indicated that Shh signaling pathway could promote gastric cancer cell proliferation and tumor development, and blocking this pathway may be a potential strategy in gastric cancer treatment.
Collapse
Affiliation(s)
- Jianhua Wan
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| | - Ji Zhou
- Department of Neurosurgery, Daping Hospital, Third Military Medical University , PLA, Chongqing, China
| | - Hailong Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| | - Mei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| | - Zhuanqin Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| | - Hongyan Gao
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| | - Yongzhong Wang
- Department of Radiology, Fourth Hospital of Hebei Medical University , Shijiazhuang, Hebei Province, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing, China
| |
Collapse
|
12
|
Kim Y, Kim MO, Shin JS, Park SH, Kim SB, Kim J, Park SC, Han CJ, Ryu JK, Yoon YB, Kim YT. Hedgehog signaling between cancer cells and hepatic stellate cells in promoting cholangiocarcinoma. Ann Surg Oncol 2014; 21:2684-98. [PMID: 24682719 DOI: 10.1245/s10434-014-3531-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Aberrant Hedgehog (HH) signaling activation is important in cancer growth and mediates the interaction between cancer cells and the surrounding stromal cells. This study investigated the role of HH signaling on the growth of cholangiocarcinoma (CC), focusing on the interaction of CC cells with stromal cells. METHODS To evaluate the interaction between human CC cells (SNU-1196, SNU-246, SNU-308, SNU-1079, and HuCCT-1) and stromal cells (hepatic stellate cell line, Lx-2), co-culture proliferation, migration, and invasion assays were performed. In vivo nude mice experiments were conducted using two groups-HuCCT-1 single implant xenograft (SX) and co-implant xenograft (CX) with HuCCT-1 and Lx-2. RESULTS When HuCCT-1 cells were co-cultured with Lx-2 cells, the expression of HH signaling-related proteins increased in both HuCCT-1 and Lx-2 cells. Co-culture with Lx-2 cells stimulated the proliferation, migration, and invasion of CC cells, and these effects were mediated by HH signaling. Co-culture of HuCCT-1 and Lx-2 cells increased the secretion of several cytokines. In an ectopic xenograft model, Lx-2 co-implantation increased CC tumor growth and stimulated angiogenesis. Cyclopamine attenuated tumor growth in the CX group, but not in the HuCCT-1 mono-implant (SX) group. Cyclopamine treatment decreased CC cell proliferation, suppressed microvessel density, and increased tumor necrosis in the CX group, but not in the SX group. CONCLUSION Hepatic stellate cells stimulate the proliferation, migration, and invasion of CC cells, promote angiogenesis through HH signaling activation, and render CC more susceptible to necrosis by HH inhibitor.
Collapse
Affiliation(s)
- Younjoo Kim
- Division of Gastroenterology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wehrkamp CJ, Gutwein AR, Natarajan SK, Phillippi MA, Mott JL. XIAP antagonist embelin inhibited proliferation of cholangiocarcinoma cells. PLoS One 2014; 9:e90238. [PMID: 24603802 PMCID: PMC3946004 DOI: 10.1371/journal.pone.0090238] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/26/2014] [Indexed: 01/07/2023] Open
Abstract
Cholangiocarcinoma cells are dependent on antiapoptotic signaling for survival and resistance to death stimuli. Recent mechanistic studies have revealed that increased cellular expression of the E3 ubiquitin-protein ligase X-linked inhibitor of apoptosis (XIAP) impairs TRAIL- and chemotherapy-induced cytotoxicity, promoting survival of cholangiocarcinoma cells. This study was undertaken to determine if pharmacologic antagonism of XIAP protein was sufficient to sensitize cholangiocarcinoma cells to cell death. We employed malignant cholangiocarcinoma cell lines and used embelin to antagonize XIAP protein. Embelin treatment resulted in decreased XIAP protein levels by 8 hours of treatment with maximal effect at 16 hours in KMCH and Mz-ChA-1 cells. Assessment of nuclear morphology demonstrated a concentration-dependent increase in nuclear staining. Interestingly, embelin induced nuclear morphology changes as a single agent, independent of the addition of TNF-related apoptosis inducing ligand (TRAIL). However, caspase activity assays revealed that increasing embelin concentrations resulted in slight inhibition of caspase activity, not activation. In addition, the use of a pan-caspase inhibitor did not prevent nuclear morphology changes. Finally, embelin treatment of cholangiocarcinoma cells did not induce DNA fragmentation or PARP cleavage. Apoptosis does not appear to contribute to the effects of embelin on cholangiocarcinoma cells. Instead, embelin caused inhibition of cell proliferation and cell cycle analysis indicated that embelin increased the number of cells in S and G2/M phase. Our results demonstrate that embelin decreased proliferation in cholangiocarcinoma cell lines. Embelin treatment resulted in decreased XIAP protein expression, but did not induce or enhance apoptosis. Thus, in cholangiocarcinoma cells the mechanism of action of embelin may not be dependent on apoptosis.
Collapse
Affiliation(s)
- Cody J. Wehrkamp
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ashley R. Gutwein
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sathish Kumar Natarajan
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mary Anne Phillippi
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
14
|
Abstract
Because of its unique function and anatomical location, the liver is exposed to a multitude of toxins and xenobiotics, including medications and alcohol, as well as to infection by hepatotropic viruses, and therefore, is highly susceptible to tissue injury. Cell death in the liver occurs mainly by apoptosis or necrosis, with apoptosis also being the physiologic route to eliminate damaged or infected cells and to maintain tissue homeostasis. Liver cells, especially hepatocytes and cholangiocytes, are particularly susceptible to death receptor-mediated apoptosis, given the ubiquitous expression of the death receptors in the organ. In a quite unique way, death receptor-induced apoptosis in these cells is mediated by both mitochondrial and lysosomal permeabilization. Signaling between the endoplasmic reticulum and the mitochondria promotes hepatocyte apoptosis in response to excessive free fatty acid generation during the metabolic syndrome. These cell death pathways are partially regulated by microRNAs. Necrosis in the liver is generally associated with acute injury (i.e., ischemia/reperfusion injury) and has been long considered an unregulated process. Recently, a new form of "programmed" necrosis (named necroptosis) has been described: the role of necroptosis in the liver has yet to be explored. However, the minimal expression of a key player in this process in the liver suggests this form of cell death may be uncommon in liver diseases. Because apoptosis is a key feature of so many diseases of the liver, therapeutic modulation of liver cell death holds promise. An updated overview of these concepts is given in this article.
Collapse
Affiliation(s)
- Maria Eugenia Guicciardi
- 1Division of Gastroenterology and Hepatology, College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
15
|
Fingas CD, Mertens JC, Razumilava N, Sydor S, Bronk SF, Christensen JD, Rizvi SH, Canbay A, Treckmann JW, Paul A, Sirica AE, Gores GJ. Polo-like kinase 2 is a mediator of hedgehog survival signaling in cholangiocarcinoma. Hepatology 2013; 58:1362-74. [PMID: 23703673 PMCID: PMC3811036 DOI: 10.1002/hep.26484] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 04/16/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Cholangiocarcinoma (CCA) cells paradoxically express the death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and thus rely on potent survival signals to circumvent cell death by TRAIL. Hedgehog (Hh) signaling is an important survival pathway in CCA. Herein, we further examine the mechanisms whereby Hh signaling mediates apoptosis resistance in CCA, revealing a pivotal role for the cell division regulating serine/threonine kinase polo-like kinase 2 (PLK2). We employed 50 human CCA samples (25 intrahepatic and 25 extrahepatic CCA) as well as human KMCH-1, Mz-CHA-1, and HUCCT-1 CCA cells for these studies. In vivo experiments were conducted using a syngeneic rat orthotopic CCA model. In human samples, polo-like kinase (PLK)1/2/3-immunoreactive cancer cells were present in the preponderance of intra- and extrahepatic CCA specimens. Inhibition of Hh signaling by cyclopamine reduced PLK2, but not PLK1 or PLK3, messenger RNA and protein expression in vehicle-treated and sonic Hh-treated CCA cells, confirming our previous microarray study. PLK2 regulation by Hh signaling appears to be direct, because the Hh transcription factors, glioma-associated oncogene 1 and 2, bind to the PLK2 promotor. Moreover, inhibition of PLK2 by the PLK inhibitor, BI 6727 (volasertib), or PLK2 knockdown was proapoptotic in CCA cells. BI 6727 administration or PLK2 knockdown decreased cellular protein levels of antiapoptotic myeloid cell leukemia 1 (Mcl-1), an effect reversed by the proteasome inhibitor, MG-132. Finally, BI 6727 administration reduced Mcl-1 protein expression in CCA cells, resulting in CCA cell apoptosis and tumor suppression in vivo. CONCLUSION PLK2 appears to be an important mediator of Hh survival signaling. These results suggest PLK inhibitors to be of therapeutic value for treatment of human CCA.
Collapse
Affiliation(s)
- Christian D. Fingas
- Department of General, Visceral, and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Joachim C. Mertens
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Nataliya Razumilava
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Svenja Sydor
- Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany
| | - Steven F. Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - John D. Christensen
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Sumera H. Rizvi
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany
| | - Jürgen W. Treckmann
- Department of General, Visceral, and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Andreas Paul
- Department of General, Visceral, and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Alphonse E. Sirica
- Division of Cellular and Molecular Pathogenesis, Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
16
|
Raychaudhuri S, Raychaudhuri SC. Death ligand concentration and the membrane proximal signaling module regulate the type 1/type 2 choice in apoptotic death signaling. SYSTEMS AND SYNTHETIC BIOLOGY 2013; 8:83-97. [PMID: 24592294 DOI: 10.1007/s11693-013-9124-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 01/06/2023]
Abstract
Apoptotic death pathways are frequently activated by death ligand induction and subsequent activation of the membrane proximal signaling module. Death receptors cluster upon binding to death ligands, leading to formation of a membrane proximal death-inducing-signaling-complex (DISC). In this membrane proximal signalosome, initiator caspases (caspase 8) are processed resulting in activation of both type 1 and type 2 pathways of apoptosis signaling. How the type 1/type 2 choice is made is an important question in the systems biology of apoptosis signaling. In this study, we utilize a Monte Carlo based in silico approach to elucidate the role of membrane proximal signaling module in the type 1/type 2 choice of apoptosis signaling. Our results provide crucial mechanistic insights into the formation of DISC signalosome and caspase 8 activation. Increased concentration of death ligands was shown to correlate with increased type 1 activation. We also study the caspase 6 mediated system level feedback activation of apoptosis signaling and its role in the type 1/type 2 choice. Our results clarify the basis of cell-to-cell stochastic variability in apoptosis activation and ramifications of this issue is further discussed in the context of therapies for cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Subhadip Raychaudhuri
- Indraprastha Institute of Information Technology, Delhi, 110020 Delhi India ; Department of Chemistry, University of California, Davis, Davis, CA 95776 USA
| | | |
Collapse
|
17
|
Schneider-Brachert W, Heigl U, Ehrenschwender M. Membrane trafficking of death receptors: implications on signalling. Int J Mol Sci 2013; 14:14475-503. [PMID: 23852022 PMCID: PMC3742255 DOI: 10.3390/ijms140714475] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/19/2013] [Accepted: 06/27/2013] [Indexed: 12/22/2022] Open
Abstract
Death receptors were initially recognised as potent inducers of apoptotic cell death and soon ambitious attempts were made to exploit selective ignition of controlled cellular suicide as therapeutic strategy in malignant diseases. However, the complexity of death receptor signalling has increased substantially during recent years. Beyond activation of the apoptotic cascade, involvement in a variety of cellular processes including inflammation, proliferation and immune response was recognised. Mechanistically, these findings raised the question how multipurpose receptors can ensure selective activation of a particular pathway. A growing body of evidence points to an elegant spatiotemporal regulation of composition and assembly of the receptor-associated signalling complex. Upon ligand binding, receptor recruitment in specialized membrane compartments, formation of receptor-ligand clusters and internalisation processes constitute key regulatory elements. In this review, we will summarise the current concepts of death receptor trafficking and its implications on receptor-associated signalling events.
Collapse
Affiliation(s)
- Wulf Schneider-Brachert
- Institute for Clinical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, Regensburg 93053, Germany; E-Mails: (W.S.-B.); (U.H.)
| | - Ulrike Heigl
- Institute for Clinical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, Regensburg 93053, Germany; E-Mails: (W.S.-B.); (U.H.)
| | - Martin Ehrenschwender
- Institute for Clinical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, Regensburg 93053, Germany; E-Mails: (W.S.-B.); (U.H.)
| |
Collapse
|
18
|
Desmoplastic/nodular medulloblastoma associated with anhidrotic ectodermal dysplasia. Int Cancer Conf J 2013. [DOI: 10.1007/s13691-013-0086-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
19
|
Raychaudhuri S, Raychaudhuri SC. Monte carlo study elucidates the type 1/type 2 choice in apoptotic death signaling in healthy and cancer cells. Cells 2013; 2:361-92. [PMID: 24709706 PMCID: PMC3972686 DOI: 10.3390/cells2020361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/10/2013] [Accepted: 05/14/2013] [Indexed: 11/16/2022] Open
Abstract
Apoptotic cell death is coordinated through two distinct (type 1 and type 2) intracellular signaling pathways. How the type 1/type 2 choice is made remains a central problem in the biology of apoptosis and has implications for apoptosis related diseases and therapy. We study the problem of type 1/type 2 choice in silico utilizing a kinetic Monte Carlo model of cell death signaling. Our results show that the type 1/type 2 choice is linked to deterministic versus stochastic cell death activation, elucidating a unique regulatory control of the apoptotic pathways. Consistent with previous findings, our results indicate that caspase 8 activation level is a key regulator of the choice between deterministic type 1 and stochastic type 2 pathways, irrespective of cell types. Expression levels of signaling molecules downstream also regulate the type 1/type 2 choice. A simplified model of DISC clustering elucidates the mechanism of increased active caspase 8 generation and type 1 activation in cancer cells having increased sensitivity to death receptor activation. We demonstrate that rapid deterministic activation of the type 1 pathway can selectively target such cancer cells, especially if XIAP is also inhibited; while inherent cell-to-cell variability would allow normal cells stay protected.
Collapse
|
20
|
Raychaudhuri S, Das S. Low Probability Activation of Bax/Bak Can Induce Selective Killing of Cancer Cells by Generating Heterogeneity in Apoptosis. JOURNAL OF HEALTHCARE ENGINEERING 2013; 4:47-66. [DOI: 10.1260/2040-2295.4.1.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Gradilone SA, Radtke BN, Bogert PS, Huang BQ, Gajdos GB, LaRusso NF. HDAC6 inhibition restores ciliary expression and decreases tumor growth. Cancer Res 2013; 73:2259-70. [PMID: 23370327 DOI: 10.1158/0008-5472.can-12-2938] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Primary cilia are multisensory organelles recently found to be absent in some tumor cells, but the mechanisms of deciliation and the role of cilia in tumor biology remain unclear. Cholangiocytes, the epithelial cells lining the biliary tree, normally express primary cilia and their interaction with bile components regulates multiple processes, including proliferation and transport. Using cholangiocarcinoma as a model, we found that primary cilia are reduced in cholangiocarcinoma by a mechanism involving histone deacetylase 6 (HDAC6). The experimental deciliation of normal cholangiocyte cells increased the proliferation rate and induced anchorage-independent growth. Furthermore, deciliation induced the activation of mitogen-activated protein kinase and Hedgehog signaling, two important pathways involved in cholangiocarcinoma development. We found that HDAC6 is overexpressed in cholangiocarcinoma and overexpression of HDAC6 in normal cholangiocytes induced deciliation and increased both proliferation and anchorage-independent growth. To evaluate the effect of cilia restoration on tumor cells, we targeted HDAC6 by short hairpin RNA (shRNA) or by the pharmacologic inhibitor, tubastatin-A. Both approaches restored the expression of primary cilia in cholangiocarcinoma cell lines and decreased cell proliferation and anchorage-independent growth. The effects of tubastatin-A were abolished when cholangiocarcinoma cells were rendered unable to regenerate cilia by stable transfection of IFT88-shRNA. Finally, inhibition of HDAC6 by tubastatin-A also induced a significant decrease in tumor growth in a cholangiocarcinoma animal model. Our data support a key role for primary cilia in malignant transformation, provide a plausible mechanism for their involvement, and suggest that restoration of primary cilia in tumor cells by HDAC6 targeting may be a potential therapeutic approach for cholangiocarcinoma.
Collapse
Affiliation(s)
- Sergio A Gradilone
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Center for Cell Signalling in Gastroenterology, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Hedgehog signaling inhibitor cyclopamine induces apoptosis by decreasing Gli2 and Bcl2 expression in human salivary pleomorphic adenoma cells. Biomed Rep 2013; 1:325-329. [PMID: 24648943 DOI: 10.3892/br.2013.61] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/07/2013] [Indexed: 12/20/2022] Open
Abstract
Pleomorphic adenoma is the most common benign neoplasm of the salivary gland. Few studies are currently available on pleomorphic adenoma cell apoptosis. The aim of this study was to investigate the effect of cyclopamine induction apoptosis in human salivary pleomorphic adenoma (HSPA) cells and the impact on Gli2 and Bcl2 mRNA levels. Cells were quantified and cell morphology was visualized under microscope. Flow cytometry was used to detect the apoptotic rate. Cyclopamine is considered an efficient blocker of the hedgehog (Hh) signaling pathway. Following treatment with 10 μmol/l cyclopamine for 48 h, the number of cells were reduced, and nuclear pycnosis or fragmentation, as well as chromatospherite disfiguration apoptotic morphology were observed under microscope. One-way ANOVA test results revealed a significantly greater decrease (P<0.01) of Gli2 and Bcl2 mRNA levels in the cyclopamine-treated group as compared to the blank control group and dimethyl sulfoxide (DMSO)-treated group. Following treatment with 10 μmol/l cyclopamine for 24 h, the apoptotic rate of the cyclopamine-treated group was significantly higher than that of the blank control and DMSO-treated group (P<0.01). Findings of this study showed that cyclopamine affected the mechanism of HSPA cell apoptosis, which may be associated with the downregulation of Gli2 and Bcl2 mRNA expression levels and the activation of the mitochondrial apoptotic pathways.
Collapse
|
23
|
Merchant MS, Geller JI, Baird K, Chou AJ, Galli S, Charles A, Amaoko M, Rhee EH, Price A, Wexler LH, Meyers PA, Widemann BC, Tsokos M, Mackall CL. Phase I trial and pharmacokinetic study of lexatumumab in pediatric patients with solid tumors. J Clin Oncol 2012; 30:4141-7. [PMID: 23071222 DOI: 10.1200/jco.2012.44.1055] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Lexatumumab is an agonistic, fully human monoclonal antibody against tumor necrosis factor-related apoptosis-inducing ligand receptor 2 with preclinical evidence of activity in pediatric solid tumors. PATIENTS AND METHODS This phase I dose-escalation study examined the safety, tolerability, pharmacokinetics, and immunogenicity of lexatumumab at doses up to, but not exceeding, the adult maximum-tolerated dose (3, 5, 8, and 10 mg/kg), administered once every 2 weeks to patients age≤21 years with recurrent or progressive solid tumors. RESULTS Twenty-four patients received a total of 56 cycles of lexatumumab over all four planned dose levels. One patient had grade 2 pericarditis consistent with radiation recall, and one patient developed grade 3 pneumonia with hypoxia during the second cycle. Five patients experienced stable disease for three to 24 cycles. No patients experienced complete or partial response, but several showed evidence of antitumor activity, including one patient with recurrent progressive osteosarcoma who experienced resolution of clinical symptoms and positron emission tomography activity, ongoing more than 1 year off therapy. One patient with hepatoblastoma showed a dramatic biomarker response. CONCLUSION Pediatric patients tolerate 10 mg/kg of lexatumumab administered once every 14 days, the maximum-tolerated dose identified in adults. The drug seems to mediate some clinical activity in pediatric solid tumors and may work with radiation to enhance antitumor effects.
Collapse
Affiliation(s)
- Melinda S Merchant
- National Cancer Institute, National Institutes of Health, 10 Center Dr, Building 10 CRC, Room 1W-3750, Bethesda, MD 20892-1104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bolaños AL, Milla CM, Lira JC, Ramírez R, Checa M, Barrera L, García-Alvarez J, Carbajal V, Becerril C, Gaxiola M, Pardo A, Selman M. Role of Sonic Hedgehog in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2012; 303:L978-90. [PMID: 23023967 DOI: 10.1152/ajplung.00184.2012] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal disease of unknown etiology and uncertain pathogenic mechanisms. Recent studies indicate that the pathogenesis of the disease may involve the abnormal expression of certain developmental pathways. Here we evaluated the expression of Sonic Hedgehog (SHH), Patched-1, Smoothened, and transcription factors glioma-associated oncogene homolog (GLI)1 and GLI2 by RT-PCR, as well as their localization in IPF and normal lungs by immunohistochemistry. The effects of SHH on fibroblast proliferation, migration, collagen and fibronectin production, and apoptosis were analyzed by WST-1, Boyden chamber chemotaxis, RT-PCR, Sircol, and annexin V-propidium iodide binding assays, respectively. Our results showed that all the main components of the Sonic signaling pathway were overexpressed in IPF lungs. With the exception of Smoothened, they were also upregulated in IPF fibroblasts. SHH and GLI2 localized to epithelial cells, whereas Patched-1, Smoothened, and GLI1 were observed mainly in fibroblasts and inflammatory cells. No staining was detected in normal lungs. Recombinant SHH increased fibroblast proliferation (P < 0.05), collagen synthesis, (2.5 ± 0.2 vs. 4.5 ± 1.0 μg of collagen/ml; P < 0.05), fibronectin expression (2-3-fold over control), and migration (190.3 ± 12.4% over control, P < 0.05). No effect was observed on α-smooth muscle actin expression. SHH protected lung fibroblasts from TNF-α/IFN-γ/Fas-induced apoptosis (14.5 ± 3.2% vs. 37.3 ± 7.2%, P < 0.0001). This protection was accompanied by modifications in several apoptosis-related proteins, including increased expression of X-linked inhibitor of apoptosis. These findings indicate that the SHH pathway is activated in IPF lungs and that SHH may contribute to IPF pathogenesis by increasing the proliferation, migration, extracellular matrix production, and survival of fibroblasts.
Collapse
Affiliation(s)
- Alfredo Lozano Bolaños
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|