1
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
2
|
Pu KL, Kang H, Li L. Therapeutic targets for age-related macular degeneration: proteome-wide Mendelian randomization and colocalization analyses. Front Neurol 2024; 15:1400557. [PMID: 38903171 PMCID: PMC11187347 DOI: 10.3389/fneur.2024.1400557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Background Currently, effective therapeutic drugs for age-related macular degeneration (AMD) are urgently needed, and it is crucial to explore new treatment targets. The proteome is indispensable for exploring disease targets, so we conducted a Mendelian randomization (MR) of the proteome to identify new targets for AMD and its related subtypes. Methods The plasma protein level data used in this study were obtained from two large-scale studies of protein quantitative trait loci (pQTL), comprising 35,559 and 54,219 samples, respectively. The expression quantitative trait loci (eQTL) data were sourced from eQTLGen and GTEx Version 8. The discovery set for AMD data and subtypes was derived from the FinnGen study, consisting of 9,721 AMD cases and 381,339 controls, 5,239 wet AMD cases and 273,920 controls, and 6,651 dry AMD cases and 272,504 controls. The replication set for AMD data was obtained from the study by Winkler TW et al., comprising 14,034 cases and 91,234 controls. Summary Mendelian randomization (SMR) analysis was employed to assess the association between QTL data and AMD and its subtypes, while colocalization analysis was performed to determine whether they share causal variants. Additionally, chemical exploration and molecular docking were utilized to validate potential drugs targeting the identified proteins. Results SMR and colocalization analysis jointly identified risk-associated proteins for AMD and its subtypes, including 5 proteins (WARS1, BRD2, IL20RB, TGFB1, TNFRSF10A) associated with AMD, 2 proteins (WARS1, IL20RB) associated with Dry-AMD, and 9 proteins (COL10A1, WARS1, VTN, SDF2, LBP, CD226, TGFB1, TNFRSF10A, CSF2) associated with Wet-AMD. The results revealed potential therapeutic chemicals, and molecular docking indicated a good binding between the chemicals and protein structures. Conclusion Proteome-wide MR have identified risk-associated proteins for AMD and its subtypes, suggesting that these proteins may serve as potential therapeutic targets worthy of further clinical investigation.
Collapse
Affiliation(s)
- Kun-Lin Pu
- Pengzhou Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Hong Kang
- Department of Thoracic Surgery, Sichuan Cancer Hospital, Chengdu, China
| | - Li Li
- Pengzhou Hospital of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Abdulrehman T, Qadri S, Haik Y, Sultan A, Skariah S, Kumar S, Mendoza Z, Yadav KK, Titus A, Khader S. Advances in the targeted theragnostics of osteomyelitis caused by Staphylococcus aureus. Arch Microbiol 2024; 206:288. [PMID: 38834761 DOI: 10.1007/s00203-024-04015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
Bone infections caused by Staphylococcus aureus may lead to an inflammatory condition called osteomyelitis, which results in progressive bone loss. Biofilm formation, intracellular survival, and the ability of S. aureus to evade the immune response result in recurrent and persistent infections that present significant challenges in treating osteomyelitis. Moreover, people with diabetes are prone to osteomyelitis due to their compromised immune system, and in life-threatening cases, this may lead to amputation of the affected limbs. In most cases, bone infections are localized; thus, early detection and targeted therapy may prove fruitful in treating S. aureus-related bone infections and preventing the spread of the infection. Specific S. aureus components or overexpressed tissue biomarkers in bone infections could be targeted to deliver active therapeutics, thereby reducing drug dosage and systemic toxicity. Compounds like peptides and antibodies can specifically bind to S. aureus or overexpressed disease markers and combining these with therapeutics or imaging agents can facilitate targeted delivery to the site of infection. The effectiveness of photodynamic therapy and hyperthermia therapy can be increased by the addition of targeting molecules to these therapies enabling site-specific therapy delivery. Strategies like host-directed therapy focus on modulating the host immune mechanisms or signaling pathways utilized by S. aureus for therapeutic efficacy. Targeted therapeutic strategies in conjunction with standard surgical care could be potential treatment strategies for S. aureus-associated osteomyelitis to overcome antibiotic resistance and disease recurrence. This review paper presents information about the targeting strategies and agents for the therapy and diagnostic imaging of S. aureus bone infections.
Collapse
Affiliation(s)
- Tahir Abdulrehman
- eHealth Program, DeGroote School of Business, McMaster University, Hamilton, ON, Canada
- Health Policy, Management and Informatics, Allied Health, Credit Valley Hospital, Mississauga, ON, Canada
| | - Shahnaz Qadri
- School of Pharmacy, Texas A&M University, Kingsville, USA.
| | - Yousef Haik
- Department of Mechanical & Nuclear Engineering, University of Sharjah, Sharjah, UAE.
| | - Ali Sultan
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sini Skariah
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shourya Kumar
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Zachary Mendoza
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Kamlesh K Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Anoop Titus
- Department of Preventive Cardiology, Houston Methodist, Houston, TX, USA
| | - Shameer Khader
- School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
4
|
Zhao Y, He J, Liu X, Yi C, Sun L, Zhu H. Construction and validation of a novel tumor necrosis factor-related apoptosis-inducing ligand mutant MuR5S4-TR. Asia Pac J Clin Oncol 2024; 20:210-219. [PMID: 36658682 DOI: 10.1111/ajco.13926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/06/2022] [Accepted: 12/26/2022] [Indexed: 01/21/2023]
Abstract
AIM Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively kill tumor cells but has no significant effect on normal cells. However, the use of TRAIL is limited for resistance by more than 50% of the tumor cell lines. It's very important to develop a more efficient form of TRAIL for cancer treatment. METHODS The N-terminal in soluble fragments (114-281aa) of TRAIL was redesigned to construct a novel TRAIL mutant-MuR5S4-TR. The Cell Counting Kit-8 method to explore the antitumor effects. The potential mechanisms were also explored. RESULTS Novel TRAIL mutant with cell-penetrating peptides (CPP) like and Second mitochondria-derived activator of caspases (Smac) like structure-MuR5S4-TR was successfully constructed. The prokaryotic expression system was successfully built, and the MuR5S4-TR was purified and reconfirmed by western blot. MuR5S4-TR could enhance the antitumor effects of TRAIL in most of the cancer cell lines significantly, NCI-H460 lung cancer cell line, for instance. After MuR5S4-TR treatment, the expressions of death receptor 4 (DR4), DR5, Caspase-8, and cleaved Caspase-3 were remarkably increased, however, there was no significant difference in X-linked inhibitor of apoptosis expression. CONCLUSION We constructed a novel TRAIL mutant with CPP-like and Smac-like structure-MuR5S4-TR. The MuR5S4-TR showed significantly stronger antitumor effects than TRAIL in many tumor cell lines. The MuR5S4-TR showed strong antitumor effects both in vitro and in vivo. This preliminary study implies that MuR5S4-TR may be a more efficient form of TRAIL for cancer therapy.
Collapse
Affiliation(s)
- Yaqin Zhao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianping He
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Sun
- Department of Osteoporosis, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Anzovino A, Canepa E, Alves M, Lemon NL, Carare RO, Fossati S. Amyloid Beta Oligomers Activate Death Receptors and Mitochondria-Mediated Apoptotic Pathways in Cerebral Vascular Smooth Muscle Cells; Protective Effects of Carbonic Anhydrase Inhibitors. Cells 2023; 12:2840. [PMID: 38132159 PMCID: PMC10741628 DOI: 10.3390/cells12242840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Amyloid beta (Aβ) deposition within the brain vasculature is an early hallmark of Alzheimer's disease (AD), which triggers loss of brain vascular smooth muscle cells (BVSMCs) in cerebral arteries, via poorly understood mechanisms, altering cerebral blood flow, brain waste clearance, and promoting cognitive impairment. We have previously shown that, in brain endothelial cells (ECs), vasculotropic Aβ species induce apoptosis through death receptors (DRs) DR4 and DR5 and mitochondria-mediated mechanisms, while FDA-approved carbonic anhydrase inhibitors (CAIs) prevent mitochondria-mediated EC apoptosis in vitro and in vivo. In this study, we analyzed Aβ-induced extrinsic and intrinsic (DR- and mitochondria-mediated) apoptotic pathways in BVSMC, aiming to unveil new therapeutic targets to prevent BVSMC stress and death. We show that both apoptotic pathways are activated in BVSMCs by oligomeric Aβ42 and Aβ40-Q22 (AβQ22) and mitochondrial respiration is severely impaired. Importantly, the CAIs methazolamide (MTZ) and acetazolamide (ATZ) prevent the pro-apoptotic effects in BVSMCs, while reducing caspase 3 activation and Aβ deposition in the arterial walls of TgSwDI animals, a murine model of cerebral amyloid angiopathy (CAA). This study reveals new molecular targets and a promising therapeutic strategy against BVSMC dysfunction in AD, CAA, and ARIA (amyloid-related imaging abnormalities) complications of recently FDA-approved anti-Aβ antibodies.
Collapse
Affiliation(s)
- Amy Anzovino
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Micaelly Alves
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Roxana O. Carare
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| |
Collapse
|
7
|
de Villalaín L, Álvarez-Teijeiro S, Rodríguez-Santamarta T, Fernández del Valle Á, Allonca E, Rodrigo JP, de Vicente JC, García-Pedrero JM. Emerging Role of Decoy Receptor-2 as a Cancer Risk Predictor in Oral Potentially Malignant Disorders. Int J Mol Sci 2023; 24:14382. [PMID: 37762685 PMCID: PMC10531848 DOI: 10.3390/ijms241814382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this study was to evaluate the expression of the senescence markers, Decoy Receptor 2 (DcR2) and Differentiated Embryo-Chondrocyte expressed gen 1 (DEC1), in oral potentially malignant disorders (OPMDs) to ascertain their possible association with oral cancer risk. The immunohistochemical analysis of DcR2 and DEC1 expression (along with p16 and Ki67 expression) was carried out in 60 patients with clinically diagnosed oral leukoplakia. Fifteen cases (25%) subsequently developed an invasive carcinoma. Correlations between protein marker expression, histological grade and oral cancer risk were assessed. DcR2, DEC1 and Ki67 protein expressions were found to correlate significantly with increased oral cancer risk, and also with an increased grade of dysplasia. Multivariate analysis demonstrated that DcR2 and Ki67 expression are independent predictors of oral cancer development. Our results evidence for the first time the potential of DcR2 as an early biomarker to assess oral cancer risk in patients with oral leukoplakia (HR = 59.7, p = 0.015), showing a superior predictive value to histology (HR = 4.225, p = 0.08). These findings reveal that the increased expression of DcR2 and DEC1 occurred frequently in OPMDs. In addition, DcR2 expression emerges as a powerful biomarker for oral cancer risk assessment in patients with oral leukoplakia.
Collapse
Affiliation(s)
- Lucas de Villalaín
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.d.V.); (T.R.-S.); (Á.F.d.V.)
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (S.Á.-T.); (E.A.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- CIBER de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tania Rodríguez-Santamarta
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.d.V.); (T.R.-S.); (Á.F.d.V.)
| | - Álvaro Fernández del Valle
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.d.V.); (T.R.-S.); (Á.F.d.V.)
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (S.Á.-T.); (E.A.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Juan P. Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (S.Á.-T.); (E.A.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- CIBER de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan Carlos de Vicente
- Department of Oral and Maxillofacial Surgery, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.d.V.); (T.R.-S.); (Á.F.d.V.)
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (S.Á.-T.); (E.A.); (J.P.R.)
| | - Juana M. García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (S.Á.-T.); (E.A.); (J.P.R.)
- Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- CIBER de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Kaczynski TJ, Husami NJ, Au ED, Farkas MH. Dysregulation of a lncRNA within the TNFRSF10A locus activates cell death pathways. Cell Death Discov 2023; 9:242. [PMID: 37443108 DOI: 10.1038/s41420-023-01544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/23/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
TNFRSF10A (tumor necrosis factor receptor superfamily member 10A) encodes a cell surface receptor protein involved in apoptotic, necroptotic, and inflammatory pathways. Dysregulation of TNFRSF10A has been implicated in sensitization to apoptosis and to the development of multiple diseases, yet little is known of the AC100861.1 long noncoding RNA (lncRNA) that lies head-to-head with TNFRSF10A. Given its genomic positioning, we sought to investigate the function of AC100861.1, focusing on its potential relationship with TNFRSF10A and the role it may play in death receptor signaling. Using knockdown and overexpression strategies, we probed cell viability and examined transcript and protein-level changes in key genes involved in apoptosis, necroptosis, and inflammation. Decreased cell viability was observed upon TNFRSF10A overexpression, regardless of whether the cells were subjected to the chemical stressor tunicamycin. Similarly, overexpression of AC100861.1 led to increased cell death, with a further increase observed under conditions of cellular stress. Knockdown of TNFRSF10A increased cell death only when the cells were stressed, and AC100861.1 knockdown exhibited no effect on cell death. Neither knockdown nor overexpression of either of these genes greatly affected the expression of the other. Manipulating AC100861.1, however, led to marked changes in the expression of genes involved in necroptosis and inflammatory cell-signaling pathways. Additionally, RNA fluorescence in situ hybridization (RNA-FISH) revealed that the AC100861.1 transcript is localized primarily to the cytoplasm. Together, these data suggest that AC100861.1 may have a role in regulating necroptotic and inflammatory signaling pathways and that this function is separate from changes in TNFRSF10A expression. Given the importance of this genomic locus for cell survival, these data provide insight into the function of a poorly understood lncRNA with potential implications regarding disease pathology and treatment.
Collapse
Affiliation(s)
- Tadeusz J Kaczynski
- Research Service, VA Medical Center, Buffalo, NY, USA
- Department of Ophthalmology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Nadine J Husami
- Research Service, VA Medical Center, Buffalo, NY, USA
- Department of Ophthalmology, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, USA
| | - Elizabeth D Au
- Department of Ophthalmology, State University of New York at Buffalo, Buffalo, NY, USA
| | - Michael H Farkas
- Research Service, VA Medical Center, Buffalo, NY, USA.
- Department of Ophthalmology, State University of New York at Buffalo, Buffalo, NY, USA.
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
9
|
Boonyarat C, Yenjai C, Reubroycharoen P, Chaiwiwatrakul S, Takomthong P, Pimsa P, Waiwut P. 7-Methoxyheptaphylline Enhances TRAIL-induced Apoptosis of Colorectal Adenocarcinoma Cell via JNK-mediated DR5 Expression. Biol Pharm Bull 2023:b23-00036. [PMID: 37331805 DOI: 10.1248/bpb.b23-00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
A cytokine known as TNF-related apoptosis-inducing ligand (TRAIL) has the ability to precisely cause the death of cancer cells, while normal cells are left undisturbed. Recent studies show that certain cancer cells are sensitive to the apoptotic effect of TRAIL. In this study, HT29 colorectal adenocarcinoma cells exposed to TRAIL were treated with heptaphylline and 7-methoxyheptaphylline from Clausena harmandiana in an effort to comprehend the mechanisms involved behind this activity. The MTT test was utilized to determine cell survival, and phase contrast microscopy was used to examine cell morphology. Through using real-time RT-PCR, Western blotting, and RT-PCR, the molecular mechanisms were investigated. According to the findings, whilst hepataphylline caused cytotoxicity in normal colon FHC cells, in comparison to healthy colon FHC cells, 7-methoxyheptaphylline inhibited cancer cells in a concentration-dependent manner. Heptaphylline alone or in conjunction with TRAIL showed no discernible effect on TRAL-induced HT29 cell death, but 7-methoxyheptaphylline boosted caspase-3 cleavage. The study showed that the JNK pathway was responsible for the 7-methoxyheptaphylline's enhancement of the DR5 (death receptor 5) mRNA, TRAIL receptor, and protein. The results demonstrated that the 7-methoxyheptaphylline of Clausena harmandiana increased the expression of DR5 via the JNK pathway, intensifying TRAIL-induced HT29 cell death.
Collapse
Affiliation(s)
| | - Chavi Yenjai
- Natural Products Research Unit, Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty ofScience, Khon Kaen University
| | | | - Suchada Chaiwiwatrakul
- Department of English, Faculty of Humanities and Social Sciences, Ubon Ratchathani Rajabhat University
| | | | | | - Pornthip Waiwut
- Faculty of Pharmaceutical Sciences, Ubon Ratchathani University
| |
Collapse
|
10
|
Abd-Rabou AA, Edris AE. Frankincense essential oil nanoemulsion specifically induces lung cancer apoptosis and inhibits survival pathways. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00128-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
The volatile fraction of frankincense (Boswellia sacra) oleogum was extracted, formulated in nanoemulsion and tested against lung cancer A549 cell line. First, the gum was hydro-distilled to isolate the volatile fraction (essential oil), which was analyzed via gas chromatography to identify its major volatile constituents. Then, the oil was formulated in two water-based nanoemulsions which differ from one another in the presence of propylene glycol (PG), which is used in the formulation step as a co-surfactant. The pure essential oil as well as its major volatile compound (α-pinene), its two nanoemulsions and a reference drug (Doxorubicin) were evaluated against lung cancer A549 cell lines and WI-38 normal lung cells. The evaluation included cytotoxicity (MTT and IC50), apoptosis (flow cytometric analysis) in addition to genetic assessments for some intrinsic and extrinsic genes relevant to apoptosis and survival pathways.
Results
Chromatographic analysis of frankincense essential oil revealed that α-pinene is the major volatile compound which constituent about 60% of that oil. Emulsification of the oil using the low energy technique gave nanoemulsions having major intense particles population (85–90%) with z-average diameter below 20.0 nm. Frankincense oil nanoemulsion fabricated with (PG) showed the best cytotoxic activity toward lung cancer A549 cell compared to PG-free nanoemulsion, α-pinene and the reference drug doxorubicin, along different incubation periods. Flow cytometric analysis also indicated that PG-containing nanoemulsion can induce cancer cells toward apoptosis better than the other formula and the pure oils. The same nanoemulsion was found to upregulate the pro-apoptotic genes [DR5, FAAD, Caspase 8 (Cas8), p53, and Bax] and downregulate the anti-apoptotic and reoccurrence genes (Bcl-2, NF-kB, and STAT-3). Most importantly, the PG-containing nanoemulsion had the least cytotoxic effect on the normal WI-38 lung cells.
Conclusions
These results point out to the potentials of frankincense essential oil (rich in α-pinene) and its PG-nanoemulsion as a promising adjuvant from plant-source to potentiate the activity of the systematic anti-lung cancer drugs.
Collapse
|
11
|
Xiang T, Qiao M, Xie J, Li Z, Xie H. Emerging Roles of the Unique Molecular Chaperone Cosmc in the Regulation of Health and Disease. Biomolecules 2022; 12:biom12121732. [PMID: 36551160 PMCID: PMC9775496 DOI: 10.3390/biom12121732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/25/2022] Open
Abstract
The core-1 β1-3galactosyltransferase-specific chaperone 1 (Cosmc) is a unique molecular chaperone of core-1 β1-3galactosyltransferase(C1GALT1), which typically functions inside the endoplasmic reticulum (ER). Cosmc helps C1GALT1 to fold correctly and maintain activity. It also participates in the synthesis of the T antigen, O-glycan, together with C1GALT1. Cosmc is a multifaceted molecule with a wide range of roles and functions. It involves platelet production and the regulation of immune cell function. Besides that, the loss of function of Cosmc also facilitates the development of several diseases, such as inflammation diseases, immune-mediated diseases, and cancer. It suggests that Cosmc is a critical control point in diseases and that it should be regarded as a potential target for oncotherapy. It is essential to fully comprehend Cosmc's roles, as they may provide critical information about its involvement in disease development and pathogenesis. In this review, we summarize the recent progress in understanding the role of Cosmc in normal development and diseases.
Collapse
Affiliation(s)
- Ting Xiang
- Hunan Province Key Laboratory of Tumor cellular Molecular Pathology, Cancer Research Institute, Heng yang School of Medicine, University of South China, Hengyang 421009, China
| | - Muchuan Qiao
- Hunan Province Key Laboratory of Tumor cellular Molecular Pathology, Cancer Research Institute, Heng yang School of Medicine, University of South China, Hengyang 421009, China
| | - Jiangbo Xie
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha 410013, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi’an 710069, China
- Correspondence: (Z.L.); (H.X.)
| | - Hailong Xie
- Hunan Province Key Laboratory of Tumor cellular Molecular Pathology, Cancer Research Institute, Heng yang School of Medicine, University of South China, Hengyang 421009, China
- Correspondence: (Z.L.); (H.X.)
| |
Collapse
|
12
|
Wang Y, Huang D, Song T, Qi X, Li M, Zhang H, Liu Y, Yang M. Andrographolide elevates tumor necrosis factor-related apoptosis-inducing ligand lethality through reactive oxygen species accumulation and gasdermin E cleavage in breast cancer cells. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:11. [PMID: 36352155 DOI: 10.1007/s12032-022-01878-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/23/2022] [Indexed: 11/10/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is selectively lethal to cancer cells and harmless to normal cells, making it a potential agent for cancer therapy. However, some breast cancer cells are resistant to TRAIL. This study revealed that andrographolide (Andro), an extract from Andrographis paniculate, a natural compound, sensitized breast cancer cells to TRAIL-induced tumor suppression; it identified apoptosis-associated protein regulation, reactive oxygen species accumulation, mitochondria membrane potential disruption, caspase cascade activation, and gasdermin-E cleavage to be involved in the tumor lethality mediated by Andro combined with TRAIL treatment. The flow cytometry results showed the combination of Andro and TRAIL repressed breast cancer cells by cell death induction, and the assessment of combined index indicated that the combined treatment with Andro and TRAIL repressed breast cancer cells synergistically. Blotting results displayed Andro and TRAIL combination elevated TRAIL-associated receptors, death receptors 4 and 5, at protein levels. These results provided critical insight into breast cancer patients' therapy and exploration direction for TRAIL clinical application.
Collapse
Affiliation(s)
- Yueyuan Wang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Dan Huang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Tingting Song
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xiaoyan Qi
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Hui Zhang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yang Liu
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
13
|
Implication of extrinsic and intrinsic apoptotic pathways in the targeted therapy of hepatocellular carcinoma using aptamer-labeled viramidine nanoparticles. BMC Cancer 2022; 22:1106. [PMID: 36309655 PMCID: PMC9617343 DOI: 10.1186/s12885-022-10201-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a global health problem with regional differences in epidemiological statistics. Co-assembling the drug nanoparticles and targeting moieties could improve the therapeutic delivery of anti-cancer drugs. In this attempt, we tracked the extrinsic and intrinsic apoptotic pathways in HCC cells using viramidine (VRM)-loaded aptamer (APT) nanoparticles. In these NPs, both APT and VRM act as targeted ligands/drugs to HCC cells. The NPs were characterized using TEM, ESI–MS, FTIR, and 1H NMR. The results showed uniform particles with round and smooth shapes on the nano-scale. SRB-based cytotoxicity was performed and IC50 values were measured for HCC versus normal cells upon the proposed treatments. The flow cytometry technique was applied to determine apoptosis, then confirmed using genetic and protein analyses. In addition, nitric oxide (NO) and its enzyme (iNOS) were analyzed to examine the effect of reactive nitrogen species (RNS) on apoptosis induction. The present findings indicated that Huh-7 cells were more sensitive to APT-VRM NPs than HepG2 cells, recording the lowest IC50 values (11.23 ± 0.23 µM and 16.69 ± 1.12 µM), as well as the highest significant increase in the apoptotic cells (61.5% and 42%), respectively. Intriguingely, normal BHK-21 cells recorded undetectable IC50 values in the applied NPs, confirming their targeted delivery ability. The genetic expression and protein levels of c-FLIP, Bcl-2, and TNF-α were down-regulated, while FADD, caspase 8, caspase 3, caspase 9, and Bax were up-regulated upon treatment with APT-VRM NPs. The prepared VRM NPs labeled with APT could significantly elevate NO via activation of iNOS. In conclusion, APT-VRM NPs bioconjugate interferes with HCC cells through NO-mediated extrinsic and intrinsic apoptosis.
Collapse
|
14
|
Bixin Prevents Colorectal Cancer Development through AMPK-Activated Endoplasmic Reticulum Stress. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9329151. [PMID: 35252457 PMCID: PMC8894005 DOI: 10.1155/2022/9329151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/14/2022] [Accepted: 02/01/2022] [Indexed: 01/01/2023]
Abstract
Chemicals isolated from natural products have been broadly applied in the treatment of colorectal cancer (CRC). Bixin, an apocarotenoid from the seeds of Bixa orellana, exerts multiple pharmacological properties, including neuroprotective, anti-inflammatory, cardioprotective, and antitumor effects; yet, the therapeutic effects of Bixin on CRC are still unknown. Here, we described that Bixin treatment significantly inhibited the proliferation and motility of two CRC cell lines (CaCO2 and SW480) in vitro and in vivo. In addition, Bixin administration has sensitized CRC cells to TNF-related apoptosis-inducing ligand- (TRAIL-) induced cell apoptosis. Moreover, we showed that Bixin treatment initiated the activation of PERK/eIF-2α signal in CaCO2 and SW480 cells, leading to endoplasmic reticulum stress-associated apoptosis. Pharmacological inhibition of AMP-activated protein kinase (AMPK) abrogated the Bixin-induced activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2 alpha (eIF-2α) pathway, as well as reversed the inhibitory effects of Bixin on CRC development. In conclusion, this study indicated that Bixin treatment inhibits the progression of CRC through activating the AMPK/PERK/eIF-2α pathway, providing a novel potential strategy for clinical prevention of CRC.
Collapse
|
15
|
Dong SXM, Vizeacoumar FS, Bhanumathy KK, Alli N, Gonzalez-Lopez C, Gajanayaka N, Caballero R, Ali H, Freywald A, Cassol E, Angel JB, Vizeacoumar FJ, Kumar A. Identification of novel genes involved in apoptosis of HIV-infected macrophages using unbiased genome-wide screening. BMC Infect Dis 2021; 21:655. [PMID: 34233649 PMCID: PMC8261936 DOI: 10.1186/s12879-021-06346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 06/15/2021] [Indexed: 12/01/2022] Open
Abstract
Background Macrophages, besides resting latently infected CD4+ T cells, constitute the predominant stable, major non-T cell HIV reservoirs. Therefore, it is essential to eliminate both latently infected CD4+ T cells and tissue macrophages to completely eradicate HIV in patients. Until now, most of the research focus is directed towards eliminating latently infected CD4+ T cells. However, few approaches have been directed at killing of HIV-infected macrophages either in vitro or in vivo. HIV infection dysregulates the expression of many host genes essential for the survival of infected cells. We postulated that exploiting this alteration may yield novel targets for the selective killing of infected macrophages. Methods We applied a pooled shRNA-based genome-wide approach by employing a lentivirus-based library of shRNAs to screen novel gene targets whose inhibition should selectively induce apoptosis in HIV-infected macrophages. Primary human MDMs were infected with HIV-eGFP and HIV-HSA viruses. Infected MDMs were transfected with siRNAs specific for the promising genes followed by analysis of apoptosis by flow cytometry using labelled Annexin-V in HIV-infected, HIV-exposed but uninfected bystander MDMs and uninfected MDMs. The results were analyzed using student’s t-test from at least four independent experiments. Results We validated 28 top hits in two independent HIV infection models. This culminated in the identification of four target genes, Cox7a2, Znf484, Cstf2t, and Cdk2, whose loss-of-function induced apoptosis preferentially in HIV-infected macrophages. Silencing these single genes killed significantly higher number of HIV-HSA-infected MDMs compared to the HIV-HSA-exposed, uninfected bystander macrophages, indicating the specificity in the killing of HIV-infected macrophages. The mechanism governing Cox7a2-mediated apoptosis of HIV-infected macrophages revealed that targeting respiratory chain complex II and IV genes also selectively induced apoptosis of HIV-infected macrophages possibly through enhanced ROS production. Conclusions We have identified above-mentioned novel genes and specifically the respiratory chain complex II and IV genes whose silencing may cause selective elimination of HIV-infected macrophages and eventually the HIV-macrophage reservoirs. The results highlight the potential of the identified genes as targets for eliminating HIV-infected macrophages in physiological environment as part of an HIV cure strategy. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06346-7.
Collapse
Affiliation(s)
- Simon X M Dong
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Frederick S Vizeacoumar
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kalpana K Bhanumathy
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Nezeka Alli
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Niranjala Gajanayaka
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ramon Caballero
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Hamza Ali
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Andrew Freywald
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, ON, Canada
| | - Jonathan B Angel
- Department of Medicine, the Ottawa Health Research Institute, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Franco J Vizeacoumar
- Department of Pathology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada. .,Cancer Research, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, SK, Canada.
| | - Ashok Kumar
- Apoptosis Research Center, Children's Hospital of Eastern Ontario, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada. .,Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada. .,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
16
|
Ren Y, Wang X, Huang S, Xu Y, Weng G, Yu R. Alternol Sensitizes Renal Carcinoma Cells to TRAIL-Induced Apoptosis. Front Pharmacol 2021; 12:560903. [PMID: 33841136 PMCID: PMC8026879 DOI: 10.3389/fphar.2021.560903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/05/2021] [Indexed: 01/25/2023] Open
Abstract
Purpose: Tumor necrosis factor–related apoptosis-inducing ligand (TRAIL), a member of the TNF family, can selectively induce cancer cell death while sparing normal cells. However, the application of TRAIL-based antitumor therapies has been hindered due to drug resistance. Alternol is a new compound isolated from microbial fermentation that possesses antitumor activity in different tumors. In our research, we discovered that alternol can sensitize TRAIL-induced apoptosis in renal carcinoma cells (RCCs). Materials and Methods: Cytotoxic activity was measured by MTT assay. Apoptosis was probed using the PI/annexin V method. Real-time PCR and western blot were used to test the levels of mRNA and protein, respectively. Luciferase assay was used to investigate whether CHOP regulated the expression of death receptor (DR) 5 through transcription. A xenogeneic tumor transplantation model was used to evaluate the anticancer effects of alternol/TRAIL in vivo. Results: When the mechanisms were investigated, we discovered that alternol increased DR5 expression. DR5 knockdown by siRNA eliminated the enhanced effect of alternol on TRAIL-mediated apoptosis. Alternol reduced the expression of antiapoptotic proteins and increased the levels of proapoptotic proteins. Moreover, alternol increased the level of CHOP, which is necessary for the enhancing effect of alternol on TRAIL-induced apoptosis, given that downregulation of CHOP abrogated the synergistic effect. DR5 upregulation induced by alternol required the production of reactive oxygen species (ROS). Removing ROS inhibited the induction of DR5 and blocked the antiapoptotic proteins induced by alternol. Conclusion: Taken together, our research suggested that alternol increased TRAIL-mediated apoptosis via inhibiting antiapoptotic proteins and upregulating DR5 levels via ROS generation and the CHOP pathway.
Collapse
Affiliation(s)
- Yu Ren
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo Yinzhou No 2. Hospital, Ningbo, China
| | - Xue Wang
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo Yinzhou No 2. Hospital, Ningbo, China
| | - Shuaishuai Huang
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo Yinzhou No 2. Hospital, Ningbo, China
| | - Yangkai Xu
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo Yinzhou No 2. Hospital, Ningbo, China
| | - Guobin Weng
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo Yinzhou No 2. Hospital, Ningbo, China
| | - Rui Yu
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medical, Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Kim SY, Hwang S, Choi MK, Park S, Nam KY, Kim I. Molecular mechanisms underlying the effects of the small molecule AMC-04 on apoptosis: Roles of the activating transcription factor 4-C/EBP homologous protein-death receptor 5 pathway. Chem Biol Interact 2020; 332:109277. [PMID: 33007289 DOI: 10.1016/j.cbi.2020.109277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/31/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
The unfolded protein response (UPR) is an emerging target pathway for cancer treatment owing to its ability to induce cell death. In our previous analysis of UPR-modulating small molecules, we had reported that piperazine oxalate derivative compounds (AMC-01-04) are able to promote increased phosphorylation of eukaryotic translation initiation factor-2 alpha (eIF2α). In this study, we found that AMC-04 induces apoptotic cell death via the activation of UPR in human breast and liver cancer cells. AMC-04 upregulated the expression of activating transcription factor-4 (ATF4)-C/EBP homologous protein (CHOP) and death receptor 5 (DR5) in cancer cells, as revealed by microarray analysis, small-interference RNA assay, and western blotting. From a mechanistic perspective, cytotoxic UPR pathway activation by AMC-04 is mediated by reactive oxygen species (ROS) and p38 mitogen-activated protein kinase (p38 MAPK) signaling. A chemical informatics approach predicted that AMC-04 modulates histone methyltransferase activity. Based on biochemical analysis, the activity of histone methyltransferases, including SUV39H1, SUV39H2, SETDB1, and EHMT1, was inhibited by AMC-04. Furthermore, chemical inhibition of the identified target proteins induced UPR activation and apoptotic cell death, suggesting that inhibition of histone methyltransferases is a promising strategy for cancer therapy. Taken together, we showed that the small molecule AMC-04 modulates epigenetic enzyme activity and mediates the link between cytotoxic UPR and histone modifications.
Collapse
Affiliation(s)
- So Young Kim
- Biomedical Research Center, ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505, Republic of Korea
| | - Supyong Hwang
- Convergence Medicine Research Center (CREDIT), ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505, Republic of Korea
| | - Min Kyung Choi
- Convergence Medicine Research Center (CREDIT), ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505, Republic of Korea
| | - Sojung Park
- Convergence Medicine Research Center (CREDIT), ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505, Republic of Korea
| | - Ky Youb Nam
- Pharosibio, Heungan Daero 427, Anyang, Gyeonggido, Republic of Korea.
| | - Inki Kim
- Convergence Medicine Research Center (CREDIT), ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
18
|
Wu B, Xiong J, Zhou Y, Wu Y, Song Y, Wang N, Chen L, Zhang J. Luteolin enhances TRAIL sensitivity in non-small cell lung cancer cells through increasing DR5 expression and Drp1-mediated mitochondrial fission. Arch Biochem Biophys 2020; 692:108539. [PMID: 32777260 DOI: 10.1016/j.abb.2020.108539] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/21/2020] [Accepted: 08/02/2020] [Indexed: 12/28/2022]
Abstract
Cancer cells exhibit extreme sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) over normal cells, highlighting TRAIL's potential as a novel and effective cancer drug. However, the therapeutic effect of TRAIL is limited due to drug resistance. In the present study, we sought to investigate the potential effects of luteolin as a TRAIL sensitizer in non-small cell lung cancer (NSCLC) cells. A549 and H1975 cells had low sensitivity or were resistant to TRAIL. Luteolin alone or in combination with TRAIL decreased cell viability and increased apoptosis. Furthermore, luteolin alone or in combination with TRAIL enhanced death receptor 5 (DR5) expression and dynamin-related protein 1 (Drp1)-dependent mitochondrial fission. However, the synergistic effect of luteolin on cell viability and apoptosis was reversed by DR5 and Drp1 inhibition, suggesting that DR5 upregulation and mitochondrial dynamics may be essential for luteolin as a sensitizer of TRAIL-based therapy in NSCLC. Moreover, luteolin treatment alone or in combination with TRAIL increased the phosphorylation of c-Jun N-terminal kinase (JNK), while SP600125 (the JNK inhibitor) significantly abolished the synergistic effect on DR5 expression and Drp1 translocation, indicating that JNK signaling activation was greatly associated with the synergistic effect exerted by luteolin in NSCLC cells. Therefore, TRAIL combined with luteolin could be as an effective chemotherapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Bin Wu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jie Xiong
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yingtong Wu
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yun Song
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ning Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lihua Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Jian Zhang
- Department of Respiratory and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
19
|
Lee WH, Han MW, Kim SH, Seong D, An JH, Chang HW, Kim SY, Kim SW, Lee JC. Tristetraprolin Posttranscriptionally Downregulates TRAIL Death Receptors. Cells 2020; 9:cells9081851. [PMID: 32784606 PMCID: PMC7465502 DOI: 10.3390/cells9081851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has attracted attention as a potential candidate for cancer therapy. However, many primary cancers are resistant to TRAIL, even when combined with standard chemotherapy. The mechanism of TRAIL resistance in cancer cells has not been fully elucidated. The TRAIL death receptor (DR) 3′-untranslated region (3′-UTR) is reported to contain AU-rich elements (AREs) that are important for regulating DR mRNA stability. However, the mechanisms by which DR mRNA stability is determined by its 3′-UTR are unknown. We demonstrate that tristetraprolin (TTP), an ARE-binding protein, has a critical function of regulating DR mRNA stability. DR4 mRNA contains three AREs and DR5 mRNA contains four AREs in 3′-UTR. TTP bound to all three AREs in DR4 and ARE3 in DR5 and enhanced decay of DR4/5 mRNA. TTP overexpression in colon cancer cells changed the TRAIL-sensitive cancer cells to TRAIL-resistant cells, and down-regulation of TTP increased TRAIL sensitivity via DR4/5 expression. Therefore, this study provides a molecular mechanism for enhanced levels of TRAIL DRs in cancer cells and a biological basis for posttranscriptional modification of TRAIL DRs. In addition, TTP status might be a biomarker for predicting TRAIL response when a TRAIL-based treatment is used for cancer.
Collapse
Affiliation(s)
- Won Hyeok Lee
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Myung Woul Han
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Song Hee Kim
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Daseul Seong
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Jae Hee An
- Department of Otorhinolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea; (W.H.L.); (M.W.H.); (S.H.K.); (D.S.); (J.H.A.)
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Korea
| | - Hyo Won Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.W.C.); (S.Y.K.)
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.W.C.); (S.Y.K.)
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Correspondence: (S.W.K.); (J.C.L.); Tel.: +82-2-3010-4270 (S.W.K.); +82-33-610-4372 (J.C.L.); Fax: +82-2-3010-4249 (S.W.K.); +82-33-642-3189 (J.C.L.)
| | - Jong Cheol Lee
- Department of Otorhinolaryngology, GangNeung Asan Hospital, University of Ulsan College of Medicine, Gangneung 25440, Korea
- Correspondence: (S.W.K.); (J.C.L.); Tel.: +82-2-3010-4270 (S.W.K.); +82-33-610-4372 (J.C.L.); Fax: +82-2-3010-4249 (S.W.K.); +82-33-642-3189 (J.C.L.)
| |
Collapse
|
20
|
Setroikromo R, Zhang B, Reis CR, Mistry RH, Quax WJ. Death Receptor 5 Displayed on Extracellular Vesicles Decreases TRAIL Sensitivity of Colon Cancer Cells. Front Cell Dev Biol 2020; 8:318. [PMID: 32509779 PMCID: PMC7248226 DOI: 10.3389/fcell.2020.00318] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
Tumor necrosis factor–related apoptosis inducing ligand (TRAIL) is considered to be a promising antitumor drug because of its selective proapoptotic properties on tumor cells. However, the clinical application of TRAIL is until now limited because of the resistance of several cancer cells, which can occur at various levels in the TRAIL signaling pathway. The role of decoy receptors that can side-track TRAIL, thereby preventing the formation of an activated death receptor, has been extensively studied. In this study, we have focused on extracellular vesicles (EVs) that are known to play a role in cell-to-cell communication and that can be released by donor cells into the medium transferring their components to recipient cells. TRAIL-induced apoptotic signaling is triggered upon the binding of two death receptors, DR4 and DR5. Here, we found that DR5 but not DR4 is present in the conditioned medium (CM)–derived from various cancer cells. Moreover, we observed that DR5 was exposed on EVs and can act as “decoy receptor” for binding to TRAIL. This results in a strongly reduced number of apoptotic cells upon treatment with DR5-specific TRAIL variant DHER in CM. This reduction happened with EVs containing either the long or short isoform of DR5. Taken together, we demonstrated that colon rectal tumor cells can secrete DR5-coated EVs, and this can cause TRAIL resistance. This is to our knowledge a novel finding and provides new insights into understanding TRAIL sensitivity.
Collapse
Affiliation(s)
- Rita Setroikromo
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Baojie Zhang
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Carlos R Reis
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Rima H Mistry
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Wim J Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
Artykov AA, Belov DA, Shipunova VO, Trushina DB, Deyev SM, Dolgikh DA, Kirpichnikov MP, Gasparian ME. Chemotherapeutic Agents Sensitize Resistant Cancer Cells to the DR5-Specific Variant DR5-B more Efficiently than to TRAIL by Modulating the Surface Expression of Death and Decoy Receptors. Cancers (Basel) 2020; 12:cancers12051129. [PMID: 32365976 PMCID: PMC7280987 DOI: 10.3390/cancers12051129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
TRAIL is considered a promising antitumor agent because it causes apoptosis of transformed cells without affecting normal cells. However, many types of tumors are cytokine resistant, and combination therapy with various chemotherapeutic drugs is being developed to overcome the resistance. We have demonstrated that the combination of TRAIL with doxorubicin, bortezomib, and panobinostat dramatically reduced the viability of TRAIL-resistant A549 and HT-29 cells. Chemotherapy even more efficiently sensitized cells to the DR5-specific mutant variant of TRAIL DR5-B, which does not have an affinity for decoy receptors. Bortezomib and doxorubicin greatly enhanced the surface expression of the death receptors DR5 and DR4, while panobinostat increased expression of DR5 and suppressed expression of DR4 in both cell lines. All drugs increased surface expression of the decoy receptors DcR1 and DcR2. Unlike the combined treatment, if the cells were pretreated with chemotherapy for 24 h, the cytotoxic activity of TRAIL was less pronounced, while sequential treatment of cells enhanced the effectiveness of DR5-B. The same results were obtained with agonistic anti-DR5 antibodies. Thus, the effectiveness of TRAIL was rather limited due to changes in the ratio of death and decoy receptors and DR5-specific agonists may be preferred in combination antitumor therapy regimens.
Collapse
Affiliation(s)
- Artem A. Artykov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Dmitry A. Belov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
| | - Victoria O. Shipunova
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (V.O.S.); (S.M.D.)
| | - Daria B. Trushina
- Department of X-ray and Synchrotron Research, A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, 119333 Moscow, Russia;
| | - Sergey M. Deyev
- Department of Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (V.O.S.); (S.M.D.)
| | - Dmitry A. Dolgikh
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Mikhail P. Kirpichnikov
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marine E. Gasparian
- Department of Bioengineering, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.A.); (D.A.B.); (D.A.D.); (M.P.K.)
- Correspondence: ; Tel.: +7-905-515-7494
| |
Collapse
|
22
|
Singh D, Prasad CB, Biswas D, Tewari M, Kar AG, Ansari MA, Singh S, Narayan G. TRAIL receptors are differentially regulated and clinically significant in gallbladder cancer. Pathology 2020; 52:348-358. [PMID: 32111400 DOI: 10.1016/j.pathol.2019.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/15/2022]
Abstract
Deregulation of the receptors of TNF-related apoptosis inducing ligand (TRAIL) has been reported in various cancers. In an effort to define the role of these receptors we profiled their expression in gallbladder cancer (GBC) and explored their clinical significance. Expression of TRAIL receptors' mRNA in GBC was analysed through reverse transcriptase polymerase chain reaction (RT-PCR), and protein through western blotting, immunohistochemistry and enzyme-linked immunosorbent assay (ELISA). mRNA data show frequent higher expression of TRAIL receptors in GBC samples. Death receptors DR4 and DR5 showed significant negative correlation with tumour stage, T stage and tumour grade; DcR1 transcript showed positive correlation with tumour stage, N stage, M stage and tumour grade. Similarly, IHC showed frequent positive staining for DR4, DR5 and DcR1in GBC samples. Cytoplasmic and nuclear DR4 protein showed negative correlation with T stage and tumour grade, whereas cytoplasmic DcR1 protein showed positive correlation with tumour stage and N stage. Nuclear DcR1 showed positive correlation with N stage. ELISA results showed significantly higher expression of secretory DcR1 in GBC patients. Kaplan-Meier analysis demonstrated significantly decreased mean survival of patients with positive staining of cytoplasmic DcR1. High level of death receptors identified the patients with early gallbladder cancer, whereas high DcR1 expression served as a prognostic factor for poor outcome.
Collapse
Affiliation(s)
- Deepika Singh
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Chandra Bhushan Prasad
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Dipanjan Biswas
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mumtaz Ahmed Ansari
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Gopeshwar Narayan
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
23
|
Gao H, Zhang X, Ding Y, Qiu R, Hong Y, Chen W. Synergistic Suppression Effect on Tumor Growth of Colorectal Cancer by Combining Radiotherapy With a TRAIL-Armed Oncolytic Adenovirus. Technol Cancer Res Treat 2019; 18:1533033819853290. [PMID: 31138083 PMCID: PMC6542122 DOI: 10.1177/1533033819853290] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The combination of gene therapy and radiation is a promising new treatment for cancer. This study aimed to clarify the synergistic effect of targeted oncolytic adenovirus (radiotherapy-tumor necrosis factor-related apoptosis-inducing ligand) and radiotherapy on colorectal cancer cells and elucidate the mechanisms of the underlying antitumor activity. Viability, cell cycle status, and apoptosis of treated colorectal cancer cells were determined via MTT and flow cytometric assays. The molecular mechanism underlying apoptotic pathway activation was elucidated through Western blot analysis of caspase-8, caspase-3, and PARP proteins. Combination treatment with radiotherapy-tumor necrosis factor-related apoptosis-inducing ligand and radiotherapy displayed significantly greater antitumor activity than either of the monotherapies. The primary mechanism behind the antitumor activity in the SW480 and Lovo colorectal cancer cell lines was apoptosis induction through the caspase pathway and G1 phase arrest. In an SW480 xenograft model of colorectal cancer, the combination therapy achieved a significantly greater reduction in tumor volume than the monotherapies. Overall, in this study, we demonstrate that the oncolytic radiotherapy-tumor necrosis factor-related apoptosis-inducing ligand construct can sensitize human colorectal cancer cells to radiation-induced apoptosis both in vitro and in vivo. Therefore, our findings point toward a novel synergistic approach to colorectal cancer treatment.
Collapse
Affiliation(s)
- Hangxiang Gao
- 1 Department of Radiology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Zhang
- 2 Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Ding
- 3 Department of Intensive Care Unit, School of Medicine, Sir Run Run Shaw Hospital Xiasha Campus, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rong Qiu
- 4 Department of Pathology and Pathophysiology, Hangzhou Medical University, Hangzhou, Zhejiang, China
| | - Yupeng Hong
- 5 Departments of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wanyuan Chen
- 2 Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Potu H, Kandarpa M, Peterson LF, Donato NJ, Talpaz M. Tumor necrosis factor related apoptosis inducing ligand (TRAIL) regulates deubiquitinase USP5 in tumor cells. Oncotarget 2019; 10:5745-5754. [PMID: 31645897 PMCID: PMC6791380 DOI: 10.18632/oncotarget.27196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/22/2019] [Indexed: 01/08/2023] Open
Abstract
The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway has emerged as a cancer therapeutic target. However, clinical trials have proven that most human cancers are resistant to TRAIL. We show that exposure to recombinant TRAIL resulted in the accumulation of ubiquitinated proteins and free ubiquitin polymers, suggesting a link between TRAIL and the ubiquitin (Ub)-proteasome pathway. TRAIL treatment in cancer cells reduced the activity and cleavage of USP5, a deubiquitinase (DUB) previously shown to target unanchored Ub polymers and regulate p53-mediated transcription. TRAIL was effective in suppressing USP5 activity and cleavage in TRAIL-sensitive cells but not resistant cells. Knockdown of USP5 in TRAIL-resistant cells demonstrated that USP5 controls apoptotic responsiveness to TRAIL. USP5 cleavage and ubiquitination were blocked by caspase-8 specific inhibitors. A small-molecule USP5/9× inhibitor (G9) combined with TRAIL enhanced apoptosis and blocked colony growth in highly TRAIL-resistant cell lines. Finally, USP5 protein levels and activity were found to be frequently deregulated in TRAIL-resistant cells. Together, we conclude that activated TRAIL enhances USP5 activity and induces apoptosis in TRAIL-sensitive and -resistant cells. We also suggest that USP5 inhibition may be effective in inducing apoptotic thresholds to enhance responsiveness to TRAIL.
Collapse
Affiliation(s)
- Harish Potu
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Malathi Kandarpa
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Luke F Peterson
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Nicholas J Donato
- Center for Scientific Review, National Institutes of Health, Bethesda, MD 20892, USA
| | - Moshe Talpaz
- Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
25
|
Lee GT, Kim JH, Kwon SJ, Stein MN, Hong JH, Nagaya N, Billakanti S, Kim MM, Kim WJ, Kim IY. Dihydrotestosterone Increases Cytotoxic Activity of Macrophages on Prostate Cancer Cells via TRAIL. Endocrinology 2019; 160:2049-2060. [PMID: 31184711 PMCID: PMC6691685 DOI: 10.1210/en.2019-00367] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/05/2019] [Indexed: 01/03/2023]
Abstract
Although androgen deprivation therapy (ADT) and immunotherapy are potential treatment options in men with metastatic prostate cancer (CaP), androgen has conventionally been proposed to be a suppressor of the immune response. However, we herein report that DHT activates macrophages. When the murine macrophage cell line (RAW 264.7), human monocyte cell line (THP-1), and human peripheral blood monocytes were cultured with androgen-resistant CaP cell lines, DHT increased cytotoxicity of macrophages in a concentration-dependent manner. Further studies revealed that DHT induced M1 polarization and increased the expression levels of TNF-related apoptosis-inducing ligand (TRAIL) in macrophages and that this effect was abrogated when TRAIL was neutralized with a blocking antibody or small interfering RNA. Subsequent experiments demonstrated that induction of TRAIL expression was regulated by direct binding of androgen receptor to the TRAIL promoter region. Finally, an in vivo mouse study demonstrated that castration enhanced the growth of an androgen-resistant murine CaP tumor and that this protumorigenic effect of castration was blocked when macrophages were removed with clodronate liposomes. Collectively, these results demonstrate that DHT activates the cytotoxic activity of macrophages and suggest that immunotherapy may not be optimal when combined with ADT in CaP.
Collapse
Affiliation(s)
- Geun Taek Lee
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Jeong Hyun Kim
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
- Department of Urology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Seok Joo Kwon
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Mark N Stein
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Jeong Hee Hong
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
- Department of Urology, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Naoya Nagaya
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Sachin Billakanti
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Melina Minji Kim
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Isaac Yi Kim
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
- Division of Urology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
26
|
Targeting TRAIL. Bioorg Med Chem Lett 2019; 29:2527-2534. [PMID: 31383590 DOI: 10.1016/j.bmcl.2019.07.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as Apo2L, has been investigated in the past decade for its promising anticancer activity due to its ability to selectively induce apoptosis in tumoral cells by binding to TRAIL receptors (TRAIL-R). Macromolecules such as agonistic monoclonal antibodies and recombinant TRAIL have not proven efficacious in clinical studies, therefore several small molecules acting as TRAIL-R agonists are emerging in the scientific literature. In this work we focus on systemizing these drug molecules described in the past years, in order to better understand and predict the requirements for a novel anti-tumoral therapy based on the TRAIL-R-induced apoptotic mechanism.
Collapse
|
27
|
Kreutzman A, Yadav B, Brummendorf TH, Gjertsen BT, Hee Lee M, Janssen J, Kasanen T, Koskenvesa P, Lotfi K, Markevärn B, Olsson-Strömberg U, Stentoft J, Stenke L, Söderlund S, Udby L, Richter J, Hjorth-Hansen H, Mustjoki S. Immunological monitoring of newly diagnosed CML patients treated with bosutinib or imatinib first-line. Oncoimmunology 2019; 8:e1638210. [PMID: 31428530 PMCID: PMC6685516 DOI: 10.1080/2162402x.2019.1638210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022] Open
Abstract
Changes in the immune system induced by tyrosine kinase inhibitors (TKI) have been shown to positively correlate with therapy responses in chronic myeloid leukemia (CML). However, only a few longitudinal studies exist and no randomized comparisons between two TKIs have been reported. Therefore, we prospectively analyzed the immune system of newly diagnosed CML patients treated with imatinib (n = 20) or bosutinib (n = 13), that participated in the randomized BFORE trial (NCT02130557). Comprehensive immunophenotyping, plasma protein profiling, and functional assays to determine activation levels of T and NK cells were performed at diagnosis, 3, and 12 months after therapy start. All results were correlated with clinical parameters such as Sokal risk and BCR-ABL load measured according to IS%. At diagnosis, low Sokal risk CML patients had a higher frequency of cytotoxic cells (CD8 + T and NK cells), increased cytotoxic potential of NK cells and lower frequency of naïve and central memory CD4 + T cells. Further, soluble plasma protein profile divided patients into two distinct clusters with different disease burden at diagnosis. During treatment, BCR-ABL IS% correlated with immunological parameters such as plasma proteins, together with different memory subsets of CD4+ and CD8 + T cells. Interestingly, the proportion and cytotoxic potential of NK cells together with several soluble proteins increased during imatinib treatment. In contrast, no major immunological changes were observed during bosutinib treatment. In conclusion, imatinib and bosutinib were shown to have differential effects on the immune system in this randomized clinical trial. Increased number and function of NK cells were especially observed during imatinib therapy.
Collapse
Affiliation(s)
- Anna Kreutzman
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Bhagwan Yadav
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Tim H Brummendorf
- Department of Hematology and Oncology, Universitätsklinikum RWTH Aachen, Aachen, Germany
| | - Bjorn Tore Gjertsen
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital and Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Moon Hee Lee
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Jeroen Janssen
- Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Perttu Koskenvesa
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Kourosh Lotfi
- Department of Medical and Health Sciences, Linköping University, Department of Hematology, County Council of Östergötland, Linköping, Sweden
| | - Berit Markevärn
- Department of Hematology, Umeå University Hospital, Umeå, Sweden
| | - Ulla Olsson-Strömberg
- Department of Medical Sciences, Uppsala University and Hematology Section, Uppsala University Hospital, Uppsala, Sweden
| | - Jesper Stentoft
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Leif Stenke
- Department of Hematology, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Stina Söderlund
- Department of Medical Sciences, Uppsala University and Hematology Section, Uppsala University Hospital, Uppsala, Sweden
| | - Lene Udby
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Johan Richter
- Department of Hematology, Skåne University Hospital, Lund, Sweden
| | - Henrik Hjorth-Hansen
- Department of Hematology, St Olavs Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| |
Collapse
|
28
|
Mahalingam D, Carew JS, Espitia CM, Cool RH, Giles FJ, de Jong S, Nawrocki ST. Heightened JNK Activation and Reduced XIAP Levels Promote TRAIL and Sunitinib-Mediated Apoptosis in Colon Cancer Models. Cancers (Basel) 2019; 11:E895. [PMID: 31248045 PMCID: PMC6678293 DOI: 10.3390/cancers11070895] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis that may be a promising agent in cancer therapy due to its selectivity toward tumor cells. However, many cancer cells are resistant to TRAIL due to defects in apoptosis signaling or activation of survival pathways. We hypothesized that a disruption of pro-survival signaling cascades with the multi-tyrosine kinase inhibitor sunitinib and would be an effective strategy to enhance TRAIL-mediated apoptosis. Here we demonstrate that sunitinib significantly augments the anticancer activity of TRAIL in models of colon cancer. The therapeutic benefit of the TRAIL/sunitinib combination was associated with increased apoptosis marked by enhanced caspase-3 cleavage and DNA fragmentation. Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Importantly, the therapeutic benefit of the TRAIL/sunitinib combination was validated in the HCT116-Luc and HCT15 colon cancer xenograft models, which both demonstrated significant anti-tumor activity in response to combination treatment. Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis.
Collapse
Affiliation(s)
- Devalingam Mahalingam
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Jennifer S Carew
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Claudia M Espitia
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Robbert H Cool
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Francis J Giles
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Steven de Jong
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Steffan T Nawrocki
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| |
Collapse
|
29
|
Saini S, Sripada L, Tulla K, Qiao G, Kunda N, Maker AV, Prabhakar BS. MADD silencing enhances anti-tumor activity of TRAIL in anaplastic thyroid cancer. Endocr Relat Cancer 2019; 26:551-563. [PMID: 30999276 DOI: 10.1530/erc-18-0517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/25/2019] [Indexed: 01/03/2023]
Abstract
ATC is an aggressive disease with limited therapeutic options due to drug resistance. TRAIL is an attractive anti-cancer therapy that can trigger apoptosis in a cancer cell-selective manner. However, TRAIL resistance is a major clinical obstacle for its use as a therapeutic drug. Previously, we demonstrated that MADD is a cancer cell pro-survival factor that can modulate TRAIL resistance. However, its role, if any, in overcoming TRAIL resistance in ATC is unknown. First, we characterized ATC cell lines as either TRAIL resistant, TRAIL sensitive or moderately TRAIL sensitive and evaluated MADD expression/cellular localization. We determined the effect of MADD siRNA on cellular growth and investigated its effect on TRAIL treatment. We assessed the effect of combination treatment (MADD siRNA and TRAIL) on mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels. The effect of combination treatment on tumor growth was assessed in vivo. We found increased levels of MADD in ATC cells relative to Nthy-ori 3-1. MADD protein localizes in the cytosol (endoplasmic reticulum and Golgi body) and membrane. MADD knockdown resulted in spontaneous cell death that was synergistically enhanced when combined with TRAIL treatment in otherwise resistant ATC cells. Combination treatment resulted in a significant reduction in MMP and enhanced generation of ROS indicating the putative mechanism of action. In an orthotopic mouse model of TRAIL-resistant ATC, treatment with MADD siRNA alone reduced tumor growth that, when combined with TRAIL, resulted in significant tumor regressions. We demonstrated the potential clinical utility of MADD knockdown in sensitizing cells to TRAIL-induced apoptosis in ATC.
Collapse
Affiliation(s)
- Shikha Saini
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Lakshmi Sripada
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Kiara Tulla
- Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Guilin Qiao
- Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Nicholas Kunda
- Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Ajay V Maker
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
- Department of Surgery, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
- Jesse Brown VA Medical Centre, Chicago, Illinois, USA
| |
Collapse
|
30
|
Jo EB, Lee YS, Lee H, Park JB, Park H, Choi YL, Hong D, Kim SJ. Combination therapy with c-met inhibitor and TRAIL enhances apoptosis in dedifferentiated liposarcoma patient-derived cells. BMC Cancer 2019; 19:496. [PMID: 31126284 PMCID: PMC6534902 DOI: 10.1186/s12885-019-5713-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Liposarcoma (LPS) is a tumor derived from adipose tissue, and has the highest incidence among soft tissue sarcomas. Dedifferentiated liposarcoma (DDLPS) is a malignant tumor with poor prognosis. Recurrence and metastasis rates in LPS remain high even after chemotherapy and radiotherapy following complete resection. Therefore, the development of advanced treatment strategies for LPS is required. In the present study, we investigated the effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment, and of combination treatment using TRAIL and a c-Met inhibitor on cell viability and apoptosis in LPS and DDLPS cell lines of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment, and of combination treatment using TRAIL and a c-Met inhibitor. Methods We analyzed cell viability after treatment with TRAIL and a c-Met inhibitor by measuring CCK8 and death receptor 5 (DR5) expression levels via fluorescence activated cell sorting (FACS) in both sarcoma cell lines and DDLPS patient-derived cells (PDCs). Moreover, we validated the effects of TRAIL alone and in combination with c-Met inhibitor on apoptosis in LPS cell lines and DDLPS PDCs via FACS. Results Our results revealed that combination treatment with a c-Met inhibitor and human recombinant TRAIL (rhTRAIL) suppressed cell viability and induced cell death in both sarcoma cell lines and DDLPS PDCs, which showed varying sensitivities to rhTRAIL alone. Also, we confirmed that treatment with a c-Met inhibitor upregulated DR5 levels in sarcoma cell lines and DDLPS PDCs. In both TRAIL-susceptible and TRAIL-resistant cells subjected to combination treatment, promotion of apoptosis was dependent on DR5 upregulation. Conclusion From these results, our findings validated that DR5 up-regulation caused by combination therapy with a c-Met inhibitor and rhTRAIL enhanced TRAIL sensitization and promoted apoptosis. We propose the use of this approach to overcome TRAIL resistance and serve as a novel treatment strategy for clinical trials. Electronic supplementary material The online version of this article (10.1186/s12885-019-5713-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eun Byeol Jo
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea
| | - Young Sang Lee
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea
| | - Hyunjoo Lee
- Personalized Medicine, Children's Cancer Institute Australia, Sydney, NSW, Australia
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hyojun Park
- Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Yoon-La Choi
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Pathology, Samsung Medical Center, Seoul, Republic of Korea
| | - Doopyo Hong
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| | - Sung Joo Kim
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea. .,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea. .,Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
31
|
TRAIL, OPG, and TWEAK in kidney disease: biomarkers or therapeutic targets? Clin Sci (Lond) 2019; 133:1145-1166. [PMID: 31097613 PMCID: PMC6526163 DOI: 10.1042/cs20181116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/19/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
Ligands and receptors of the tumor necrosis factor (TNF) superfamily regulate immune responses and homeostatic functions with potential diagnostic and therapeutic implications. Kidney disease represents a global public health problem, whose prevalence is rising worldwide, due to the aging of the population and the increasing prevalence of diabetes, hypertension, obesity, and immune disorders. In addition, chronic kidney disease is an independent risk factor for the development of cardiovascular disease, which further increases kidney-related morbidity and mortality. Recently, it has been shown that some TNF superfamily members are actively implicated in renal pathophysiology. These members include TNF-related apoptosis-inducing ligand (TRAIL), its decoy receptor osteoprotegerin (OPG), and TNF-like weaker inducer of apoptosis (TWEAK). All of them have shown the ability to activate crucial pathways involved in kidney disease development and progression (e.g. canonical and non-canonical pathways of the transcription factor nuclear factor-kappa B), as well as the ability to regulate cell proliferation, differentiation, apoptosis, necrosis, inflammation, angiogenesis, and fibrosis with double-edged effects depending on the type and stage of kidney injury. Here we will review the actions of TRAIL, OPG, and TWEAK on diabetic and non-diabetic kidney disease, in order to provide insights into their full clinical potential as biomarkers and/or therapeutic options against kidney disease.
Collapse
|
32
|
Nieddu V, Piredda R, Bexell D, Barton J, Anderson J, Sebire N, Kolluri K, Janes SM, Karteris E, Sala A. Engineered human mesenchymal stem cells for neuroblastoma therapeutics. Oncol Rep 2019; 42:35-42. [PMID: 31115546 PMCID: PMC6549104 DOI: 10.3892/or.2019.7152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
Drug-resistant neuroblastoma remains a major challenge in paediatric oncology and novel and less toxic therapeutic approaches are urgently needed to improve survival and reduce the side effects of traditional therapeutic interventions. Mesenchymal stem cells (MSCs) are an attractive candidate for cell and gene therapy since they are recruited by and able to infiltrate tumours. This feature has been exploited by creating genetically modified MSCs that are able to combat cancer by delivering therapeutic molecules. Whether neuroblastomas attract systemically delivered MSCs is still controversial. We investigated whether MSCs engineered to express tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) could: i) cause death of classic and primary neuroblastoma cell lines in vitro; ii) migrate to tumour sites in vivo; and iii) reduce neuroblastoma growth in xenotransplantation experiments. We observed that classic and primary neuroblastoma cell lines expressing death receptors could be killed by TRAIL-loaded MSCs in vitro. When injected in the peritoneum of neuroblastoma-bearing mice, TRAIL-MSCs migrated to tumour sites, but were unable to change the course of cancer development. These results indicated that MSCs have the potential to be used to deliver drugs in neuroblastoma patients, but more effective biopharmaceuticals should be used instead of TRAIL.
Collapse
Affiliation(s)
- Valentina Nieddu
- Department of Life Sciences, Research Institute of Environment, Health and Societies, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - Roberta Piredda
- Department of Life Sciences, Research Institute of Environment, Health and Societies, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - Daniel Bexell
- Department of Laboratory Medicine, Translational Cancer Research, Lund University, SE-221 00 Lund, Sweden
| | - Jack Barton
- Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, University College London, London WC1N 1EH, UK
| | - John Anderson
- Institute of Child Health, Unit of Molecular Haematology and Cancer Biology, University College London, London WC1N 1EH, UK
| | - Neil Sebire
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Krishna Kolluri
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Emmanouil Karteris
- Department of Life Sciences, Research Institute of Environment, Health and Societies, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - Arturo Sala
- Department of Life Sciences, Research Institute of Environment, Health and Societies, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| |
Collapse
|
33
|
MSC.sTRAIL Has Better Efficacy than MSC.FL-TRAIL and in Combination with AKTi Blocks Pro-Metastatic Cytokine Production in Prostate Cancer Cells. Cancers (Basel) 2019; 11:cancers11040568. [PMID: 31010082 PMCID: PMC6521093 DOI: 10.3390/cancers11040568] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023] Open
Abstract
Cell therapy is a promising new treatment option for cancer. In particular, mesenchymal stem cells (MSCs) have shown potential in delivering therapeutic genes in various tumour models and are now on the verge of being tested in the clinic. A number of therapeutic genes have been examined in this context, including the death ligand TRAIL. For cell therapy, it can be used in its natural form as a full-length and membrane-bound protein (FL-TRAIL) or as an engineered version commonly referred to as soluble TRAIL (sTRAIL). As to which is more therapeutically efficacious, contradicting results have been reported. We discovered that MSCs producing sTRAIL have significantly higher apoptosis-inducing activity than cells expressing FL-TRAIL and found that FL-TRAIL, in contrast to sTRAIL, is not secreted. We also demonstrated that TRAIL does induce the expression of pro-metastatic cytokines in prostate cancer cells, but that this effect could be overcome through combination with an AKT inhibitor. Thus, a combination consisting of small-molecule drugs specifically targeting tumour cells in combination with MSC.sTRAIL, not only provides a way of sensitising cancer cells to TRAIL, but also reduces the issue of side-effect-causing cytokine production. This therapeutic strategy therefore represents a novel targeted treatment option for advanced prostate cancer and other difficult to treat tumours.
Collapse
|
34
|
Sophonnithiprasert T, Mahabusarakam W, Watanapokasin R. Artonin E sensitizes TRAIL-induced apoptosis by DR5 upregulation and cFLIP downregulation in TRAIL-refractory colorectal cancer LoVo cells. J Gastrointest Oncol 2019; 10:209-217. [PMID: 31032087 DOI: 10.21037/jgo.2018.12.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background The TRAIL treatment is an ideal strategy for colorectal cancer (CRC) therapy because of minimal collateral damage to normal cells. Unfortunately, some CRC is TRAIL-refractory cancer, such as LoVo cells. In an effort to overcome TRAIL-refractory cancer, we investigated the effect of artonin E in regulating death receptor 5 (DR5) and cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (cFLIP), two major mediators regulate TRAIL-induced apoptosis, in LoVo cells as a model of TRAIL refractory CRC. Methods TRAIL-refractory cancer (LoVo cells) was treated with artonin E and TRAIL. Cell viability was determined by MTT assay. Apoptotic chromatin condensation was observed by fluorescent Hoechst33342 staining. The mRNA and protein expression of DR5 and FLIP was determined by quantitative PCR and Western blotting analysis, respectively. Results The combination treatment of artonin E and TRAIL enhanced cytotoxicity and apoptotic chromatin condensation in LoVo cells significantly, while treatment of artonin E or TRAIL alone was not. Artonin E enhanced both mRNA and protein expression of DR5. Interestingly, this is the first report showing that artonin E decreased protein expression of cFLIP. All together we showed that artonin E enhanced TRAIL-induced apoptosis in LoVo cells through DR5 upregulation and cFLIP downregulation. Conclusions Artonin E was able to increase DR5 expression and decrease cFLIP expression in LoVo cells. These results showed that LoVo cells sensitized TRAIL-induced apoptosis in combined treatment with artonin E and TRAIL. Therefore, the combination treatment of artonin E and TRAIL is one of the potential strategies used for TRAIL-refractory CRC therapy in the future.
Collapse
Affiliation(s)
- Thanet Sophonnithiprasert
- Biochemistry Unit, Department of Medical Sciences, Faculty of Science, Rangsit University, Pathum Thani, Thailand
| | - Wilawan Mahabusarakam
- Department of Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| |
Collapse
|
35
|
Hsp90 Inhibitor SNX-2112 Enhances TRAIL-Induced Apoptosis of Human Cervical Cancer Cells via the ROS-Mediated JNK-p53-Autophagy-DR5 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9675450. [PMID: 31019655 PMCID: PMC6452544 DOI: 10.1155/2019/9675450] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent cancer cell apoptosis-inducing factor that can induce apoptosis in a variety of cancer cells. However, resistance to TRAIL in cancer cells is a huge obstacle in creating effective TRAIL-targeted clinical therapies. Thus, agents that can either enhance the effect of TRAIL or overcome its resistance are needed. In this study, we combined TRAIL with SNX-2112, an Hsp90 inhibitor we previously developed, to explore the effect and mechanism that SNX-2112 enhanced TRAIL-induced apoptosis in cervical cancer cells. Our results showed that SNX-2112 markedly enhanced TRAIL-induced cytotoxicity in HeLa cells, and this combination was found to be synergistic. Additionally, we found that SNX-2112 sensitized TRAIL-mediated apoptosis caspase-dependently in TRAIL-resistant HeLa cells. Mechanismly, SNX-2112 downregulated antiapoptosis proteins, including Bcl-2, Bcl-XL, and FLIP, promoted the accumulation of reactive oxygen species (ROS), and increased the expression levels of p-JNK and p53. ROS scavenger NAC rescued SNX-2112/TRAIL-induced apoptosis and suppressed SNX-2112-induced p-JNK and p53. Moreover, SNX-2112 induced the upregulation of death-receptor DR5 in HeLa cells. The silencing of DR5 by siRNA significantly decreased cell apoptosis by the combined effect of SNX-2112 and TRAIL. In addition, SNX-2112 inhibited the Akt/mTOR signaling pathway and induced autophagy in HeLa cells. The blockage of autophagy by bafilomycin A1 or Atg7 siRNA abolished SNX-2112-induced upregulation of DR5. Meanwhile, ROS scavenger NAC, JNK inhibitor SP600125, and p53 inhibitor PFTα were used to verify that autophagy-mediated upregulation of DR5 was regulated by the SNX-2112-stimulated activation of the ROS-JNK-p53 signaling pathway. Thus, the combination of SNX-2112 and TRAIL may provide a novel strategy for the treatment of human cervical cancer by overcoming cellular mechanisms of apoptosis resistance.
Collapse
|
36
|
Deng Y, Bi R, Guo H, Yang J, Du Y, Wang C, Wei W. Andrographolide Enhances TRAIL-Induced Apoptosis via p53-Mediated Death Receptors Up-Regulation and Suppression of the NF-кB Pathway in Bladder Cancer Cells. Int J Biol Sci 2019; 15:688-700. [PMID: 30745855 PMCID: PMC6367587 DOI: 10.7150/ijbs.30847] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/20/2018] [Indexed: 01/22/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an effective chemotherapeutic agent that specifically impairs cancer cells while sparing normal cells; however, some cancer cells develop resistance to TRAIL. Here, we identified Andrographolide, a diterpenoid lactone derived from a traditional herbal medicine Andrographis paniculata, as an ideal sensitizer for TRAIL to overcome bladder cancer. Our results showed that combination treatment of Andro and TRAIL retarded growth, attenuated proliferation, decreased colony formation, inhibited migration and promoted caspases-mediated apoptosis in T24 cells. Additionally, the sensitization by Andro is achieved through up-regulation of death receptors (DR4 and DR5) of TRAIL in a p53-dependent manner. Crucially, Andro is also capable of inactivating NF-κB signaling pathway via transcriptional down-regulation p65/RelA, which is further contributed to enhancement of TRAIL-mediated cytotoxicity. These results indicated that non-toxic doses of Andrographolide sensitized bladder cancer cells to TRAIL-mediated apoptosis, suggesting it as an effective therapeutic agent for TRAIL resistant human bladder cancers.
Collapse
Affiliation(s)
- Yuyou Deng
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China.,Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Ran Bi
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China.,Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Haoran Guo
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Jiaxin Yang
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Yujun Du
- Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Wei Wei
- Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| |
Collapse
|
37
|
Elucidation for modulation of death receptor (DR) 5 to strengthen apoptotic signals in cancer cells. Arch Pharm Res 2019; 42:88-100. [DOI: 10.1007/s12272-018-01103-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
|
38
|
Zhang J, Hu J, Peng K, Song W, Zhi S, Yang E, Zhao J, Hou H. Chemical biology suggests pleiotropic effects for a novel hexanuclear copper(ii) complex inducing apoptosis in hepatocellular carcinoma cells. Chem Commun (Camb) 2019; 55:11944-11947. [DOI: 10.1039/c9cc05995d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new hexanuclear copper(ii) complex proved potential chemotherapeutic applicability in inducing apoptosis in cancer calls by acting on multiple targets and signaling pathways.
Collapse
Affiliation(s)
- Junshuai Zhang
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
| | - Jiyong Hu
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
| | - Kun Peng
- Institute of Inorganic Chemistry
- University of Wuerzburg
- Bavaria 97074
- Germany
| | - Wei Song
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
| | - Shuangcheng Zhi
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
| | - Endian Yang
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
| | - Jin’an Zhao
- College of Material and Chemical Engineering
- Henan University of Urban Construction
- Henan 467036
- P. R. China
- College of Chemistry and Molecular Engineering
| | - Hongwei Hou
- College of Chemistry and Molecular Engineering
- Zhengzhou University
- Zhengzhou 450001
- China
| |
Collapse
|
39
|
Altwaijry N, Somani S, Parkinson JA, Tate RJ, Keating P, Warzecha M, Mackenzie GR, Leung HY, Dufès C. Regression of prostate tumors after intravenous administration of lactoferrin-bearing polypropylenimine dendriplexes encoding TNF-α, TRAIL, and interleukin-12. Drug Deliv 2018; 25:679-689. [PMID: 29493296 PMCID: PMC6058574 DOI: 10.1080/10717544.2018.1440666] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/04/2018] [Accepted: 02/11/2018] [Indexed: 12/22/2022] Open
Abstract
The possibility of using gene therapy for the treatment of prostate cancer is limited by the lack of intravenously administered delivery systems able to safely and selectively deliver therapeutic genes to tumors. Given that lactoferrin (Lf) receptors are overexpressed on prostate cancer cells, we hypothesized that the conjugation of Lf to generation 3-diaminobutyric polypropylenimine dendrimer would improve its transfection and therapeutic efficacy in prostate cancer cells. In this study, we demonstrated that the intravenous administration of Lf-bearing DAB dendriplexes encoding TNFα resulted in the complete suppression of 70% of PC-3 and 50% of DU145 tumors over one month. Treatment with DAB-Lf dendriplex encoding TRAIL led to tumor suppression of 40% of PC-3 tumors and 20% of DU145 tumors. The treatment was well tolerated by the animals. Lf-bearing generation 3-polypropylenimine dendrimer is therefore a highly promising delivery system for non-viral gene therapy of prostate cancer.
Collapse
Affiliation(s)
- Najla Altwaijry
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John A. Parkinson
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Rothwelle J. Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Patricia Keating
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Monika Warzecha
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Graeme R. Mackenzie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
40
|
Patel M, Vlahava VM, Forbes SK, Fielding CA, Stanton RJ, Wang ECY. HCMV-Encoded NK Modulators: Lessons From in vitro and in vivo Genetic Variation. Front Immunol 2018; 9:2214. [PMID: 30327650 PMCID: PMC6174198 DOI: 10.3389/fimmu.2018.02214] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
Human cytomegalovirus (HCMV) is under constant selective pressure from the immune system in vivo. Study of HCMV genes that have been lost in the absence of, or genetically altered by, such selection can focus research toward findings of in vivo significance. We have been particularly interested in the most pronounced change in the highly passaged laboratory strains AD169 and Towne—the deletion of 13–15 kb of sequence (designated the UL/b′ region) that encodes up to 22 canonical genes, UL133-UL150. At least 5 genes have been identified in UL/b′ that inhibit NK cell function. UL135 suppresses formation of the immunological synapse (IS) by remodeling the actin cytoskeleton, thereby illustrating target cell cooperation in IS formation. UL141 inhibits expression of two activating ligands (CD155, CD112) for the activating receptor CD226 (DNAM-1), and two receptors (TRAIL-R1, R2) for the apoptosis-inducing TRAIL. UL142, ectopically expressed in isolation, and UL148A, target specific MICA allotypes that are ligands for NKG2D. UL148 impairs expression of CD58 (LFA-3), the co-stimulatory cell adhesion molecule for CD2 found on T and NK cells. Outside UL/b′, studies on natural variants have shown UL18 mutants change affinity for their inhibitory ligand LIR-1, while mutations in UL40's HLA-E binding peptide differentially drive NKG2C+ NK expansions. Research into HCMV genomic stability and its effect on NK function has provided important insights into virus:host interactions, but future studies will require consideration of genetic variability and the effect of genes expressed in the context of infection to fully understand their in vivo impact.
Collapse
Affiliation(s)
- Mihil Patel
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Virginia-Maria Vlahava
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Simone K Forbes
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ceri A Fielding
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Eddie C Y Wang
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
41
|
Haldar R, Shaashua L, Lavon H, Lyons YA, Zmora O, Sharon E, Birnbaum Y, Allweis T, Sood AK, Barshack I, Cole S, Ben-Eliyahu S. Perioperative inhibition of β-adrenergic and COX2 signaling in a clinical trial in breast cancer patients improves tumor Ki-67 expression, serum cytokine levels, and PBMCs transcriptome. Brain Behav Immun 2018; 73:294-309. [PMID: 29800703 DOI: 10.1016/j.bbi.2018.05.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/10/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Catecholamines and prostaglandins are secreted abundantly during the perioperative period in response to stress and surgery, and were shown by translational studies to promote tumor metastasis. Here, in a phase-II biomarker clinical trial in breast cancer patients (n = 38), we tested the combined perioperative use of the β-blocker, propranolol, and the COX2-inhibitor, etodolac, scheduled for 11 consecutive perioperative days, starting 5 days before surgery. Blood samples were taken before treatment (T1), on the mornings before and after surgery (T2&T3), and after treatment cessation (T4). Drugs were well tolerated. Results based on a-priori hypotheses indicated that already before surgery (T2), serum levels of pro-inflammatory IL-6, CRP, and IFNγ, and anti-inflammatory, cortisol and IL-10, increased. At T2 and/or T3, drug treatment reduced serum levels of the above pro-inflammatory cytokines and of TRAIL, as well as activity of multiple inflammation-related transcription factors (including NFκB, STAT3, ISRE), but not serum levels of cortisol, IL-10, IL-18, IL-8, VEGF and TNFα. In the excised tumor, treatment reduced the expression of the proliferation marker Ki-67, and positively affected its transcription factors SP1 and AhR. Exploratory analyses of transcriptome modulation in PBMCs revealed treatment-induced improvement at T2/T3 in several transcription factors that in primary tumors indicate poor prognosis (CUX1, THRa, EVI1, RORa, PBX1, and T3R), angiogenesis (YY1), EMT (GATA1 and deltaEF1/ZEB1), proliferation (GATA2), and glucocorticoids response (GRE), while increasing the activity of the oncogenes c-MYB and N-MYC. Overall, the drug treatment may benefit breast cancer patients through reducing systemic inflammation and pro-metastatic/pro-growth biomarkers in the excised tumor and PBMCs.
Collapse
Affiliation(s)
- Rita Haldar
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Lee Shaashua
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Hagar Lavon
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel
| | - Yasmin A Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Oded Zmora
- Department of Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Yehudit Birnbaum
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Tanir Allweis
- Department of Surgery, Kaplan Medical Center, Rehovot, Israel
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, M.D. Anderson Cancer Center at University of Texas, Huston, TX, USA
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Steve Cole
- Department of Medicine, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Israel.
| |
Collapse
|
42
|
Lee E, Min K, Chang YT, Kwon Y. Efficient and wash-free labeling of membrane proteins using engineered Npu DnaE split-inteins. Protein Sci 2018; 27:1568-1574. [PMID: 30151847 DOI: 10.1002/pro.3455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 01/01/2023]
Abstract
An efficient and wash-free method to conjugate a fluorescent tag to a target membrane protein is developed, using engineered Npu DnaE split-inteins. This approach allowed fast labeling while avoiding the strenuous synthesis of a long polypeptide. Two different modes of labeling, namely specific binding and covalent conjugation, are observed. The covalent labeling was monitored within 5 min, without background staining.
Collapse
Affiliation(s)
- Euiyeon Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, South Korea
| | - Kyoungmi Min
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, South Korea
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, South Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Youngeun Kwon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, South Korea
| |
Collapse
|
43
|
Zafirlukast and vincamine ameliorate tamoxifen-induced oxidative stress and inflammation: Role of the JNK/ERK pathway. Life Sci 2018; 202:78-88. [DOI: 10.1016/j.lfs.2018.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/27/2018] [Accepted: 04/02/2018] [Indexed: 12/19/2022]
|
44
|
Nazim UM, Moon JH, Lee YJ, Seol JW, Park SY. PPARγ activation by troglitazone enhances human lung cancer cells to TRAIL-induced apoptosis via autophagy flux. Oncotarget 2018; 8:26819-26831. [PMID: 28460464 PMCID: PMC5432299 DOI: 10.18632/oncotarget.15819] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/20/2017] [Indexed: 12/18/2022] Open
Abstract
Members of the tumor necrosis factor (TNF) transmembrane cytokine superfamily, such as TNFα and Fas ligand (FasL), play crucial roles in inflammation and immunity. TRAIL is a member of this superfamily with the ability to selectively trigger cancer cell death but does not motive cytotoxicity to most normal cells. Troglitazone are used in the cure of type II diabetes to reduce blood glucose levels and improve the sensitivity of an amount of tissues to insulin. In this study, we revealed that troglitazone could trigger TRAIL-mediated apoptotic cell death in human lung adenocarcinoma cells. Pretreatment of troglitazone induced activation of PPARγ in a dose-dependent manner. In addition conversion of LC3-I to LC3-II and PPARγ was suppressed in the presence of GW9662, a well-characterized PPARγ antagonist. Treatment with troglitazone resulted in a slight increase in conversion rate of LC3-I to LC3-II and significantly decreased p62 expression levels in a dose-dependent manner. This indicates that troglitazone induced autophagy flux activation in human lung cancer cells. Inhibition of autophagy flux applying a specific inhibitor and genetically modified ATG5 siRNA enclosed troglitazone-mediated enhancing effect of TRAIL. These data demonstrated that activation of PPARγ mediated by troglitazone enhances human lung cancer cells to TRAIL-induced apoptosis via autophagy flux and also suggest that troglitazone may be a combination therapeutic target with TRAIL protein in TRAIL-resistant cancer cells.
Collapse
Affiliation(s)
- Uddin Md Nazim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - You-Jin Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| |
Collapse
|
45
|
Staphylococcal Osteomyelitis: Disease Progression, Treatment Challenges, and Future Directions. Clin Microbiol Rev 2018; 31:31/2/e00084-17. [PMID: 29444953 DOI: 10.1128/cmr.00084-17] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Osteomyelitis is an inflammatory bone disease that is caused by an infecting microorganism and leads to progressive bone destruction and loss. The most common causative species are the usually commensal staphylococci, with Staphylococcus aureus and Staphylococcus epidermidis responsible for the majority of cases. Staphylococcal infections are becoming an increasing global concern, partially due to the resistance mechanisms developed by staphylococci to evade the host immune system and antibiotic treatment. In addition to the ability of staphylococci to withstand treatment, surgical intervention in an effort to remove necrotic and infected bone further exacerbates patient impairment. Despite the advances in current health care, osteomyelitis is now a major clinical challenge, with recurrent and persistent infections occurring in approximately 40% of patients. This review aims to provide information about staphylococcus-induced bone infection, covering the clinical presentation and diagnosis of osteomyelitis, pathophysiology and complications of osteomyelitis, and future avenues that are being explored to treat osteomyelitis.
Collapse
|
46
|
Mesothelin's minimal MUC16 binding moiety converts TR3 into a potent cancer therapeutic via hierarchical binding events at the plasma membrane. Oncotarget 2017; 7:31534-49. [PMID: 27120790 PMCID: PMC5058776 DOI: 10.18632/oncotarget.8925] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/10/2016] [Indexed: 12/11/2022] Open
Abstract
TRAIL has been extensively explored as a cancer drug based on its tumor-selective activity profile but it is incapable per se of discriminating between death receptors expressed by normal host cells and transformed cancer cells. Furthermore, it is well documented that surface tethering substantially increases its biologic activity. We have previously reported on Meso-TR3, a constitutive TRAIL trimer targeted to the biomarker MUC16 (CA125), in which the entire ectodomain of human mesothelin was genetically fused to the TR3 platform, facilitating attachment to the cancer cells via the MUC16 receptor. Here, we designed a truncation variant, in which the minimal 64 amino acid MUC16 binding domain of mesothelin was incorporated into TR3. It turned out that the dual-domain biologic Meso64-TR3 retained its high MUC16 affinity and bound to the cancer cells quickly, independent of the TR3/death receptor interaction. Furthermore, it was substantially more potent than Meso-TR3 and TR3 in vitro and in a preclinical xenograft model of MUC16-dependent ovarian cancer. Phenotypically, Meso64-TR3 is more closely related to non-targeted TR3, evident by indistinguishable activity profiles on MUC16-deficient cancers and similar thermal stability characteristics. Overall, Meso64-TR3 represents a fully human, MUC16-targetd TRAIL-based biologic, ideally suited for exploring preclinical and clinical evaluation studies in MUC16-dependent malignancies.
Collapse
|
47
|
Kang CH, Molagoda IMN, Choi YH, Park C, Moon DO, Kim GY. Apigenin promotes TRAIL-mediated apoptosis regardless of ROS generation. Food Chem Toxicol 2017; 111:623-630. [PMID: 29247770 DOI: 10.1016/j.fct.2017.12.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/25/2023]
Abstract
Apigenin is a bioactive flavone in several herbs including parsley, thyme, and peppermint. Apigenin possesses anti-cancer and anti-inflammatory properties; however, whether apigenin enhances TRAIL-mediated apoptosis in cancer cells is unknown. In the current study, we found that apigenin enhanced TRAIL-induced apoptosis by promoting caspase activation and death receptor 5 (DR5) expression and a chimeric antibody against DR5 completely blocked the apoptosis. Apigenin also upregulated reactive oxygen species (ROS) generation; however, intriguingly, ROS inhibitors, glutathione (GSH) or N-acetyl-l-cysteine (NAC), moderately increased apigenin/TRAIL-induced apoptosis. Additional results showed that an autophagy inducer, rapamycin, enhanced apigenin/TRAIL-mediated apoptosis by a slight increase of ROS generation. Accordingly, NAC and GSH rather decreased apigenin-induced autophagy formation, suggesting that apigenin-induced ROS generation increased autophagy formation. However, autophagy inhibitors, bafilomycin (BAF) and 3-methyladenine (3-MA), showed different result in apigenin/TRAIL-mediated apoptosis without ROS generation. 3-MA upregulated the apoptosis but remained ROS levels; however, no changes on apoptosis and ROS generation were observed by BAF treatment. Taken together, these findings reveal that apigenin enhances TRAIL-induced apoptosis by activating apoptotic caspases by upregulating DR5 expression regardless of ROS generation, which may be a promising strategy for an adjuvant of TRAIL.
Collapse
Affiliation(s)
- Chang-Hee Kang
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea; Freshwater Bioresources Utilization Bureau, Bioresources Industrialization Research Division, Sangju-si, Gyeongsangbuk-do 37242, Republic of Korea
| | | | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47340, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dong-Eui University, Busan 47340, Republic of Korea
| | - Dong-Oh Moon
- Department of Biology Education, Daegu University, Gyeongsan, Gyeongbuk 38453, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
48
|
Kim JY, Kim YM, Park JM, Han YM, Lee KC, Hahm KB, Hong S. Cancer preventive effect of recombinant TRAIL by ablation of oncogenic inflammation in colitis-associated cancer rather than anticancer effect. Oncotarget 2017; 9:1705-1716. [PMID: 29416724 PMCID: PMC5788592 DOI: 10.18632/oncotarget.23083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
The potential of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in inducing apoptosis is a hallmark in cancer therapeutics, after which its selective ability to achieve cell death pathways against cancer cells led to hope for recombinant TRAIL in cancer therapeutics. The present data from azoxymethane-initiated, dextran sulfate sodium-promoted colitis associated cancer (CAC) model strongly indicate the potential of rTRAIL in cancer prevention rather than in cancer therapeutics. Early treatment of rTRAIL significantly reduced colitis and CAC by inhibiting the recruitment of macrophages into the damaged mucosa and activating the scavenger activity with efferocytosis and the production of several growth factors. In contrast, late administration of rTRAIL as for anti-cancer effect did not decrease the initiation and development of CAC at all. Significant cancer preventing mechanisms of rTRAIL were identified. In the CAC model, anti-inflammation, regeneration, and efferocytosis was induced by treatment of TRAIL for 6 days, significant inhibitory activity was evident at 4 weeks and anti-oxidative and anti-inflammatory induction were noted at 12 weeks. Most importantly, TRAIL promoted tissue regeneration by enhancing the resolution of pathological inflammation through the activation of the NLRP3 inflammasome pathway. The results indicate that TRAIL reduces the induction of colitis and the initiation of CAC by inhibiting pro-inflammatory signaling and promoting tissue repair to maintain intestinal homeostasis through activation of the NLRP3 inflammasome. Therefore, TRAIL can be used as a chemopreventive agent against CAC, rather than as a therapeutic drug endowing apoptosis.
Collapse
Affiliation(s)
- Joo-Young Kim
- Laboratory of Cancer Cell Biology, Department of Biochemistry, School of Medicine, Gachon University, Incheon, Korea
| | - Young-Mi Kim
- Laboratory of Cancer Cell Biology, Department of Biochemistry, School of Medicine, Gachon University, Incheon, Korea
| | - Jong-Min Park
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Young Min Han
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Kang Choon Lee
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Ki Baik Hahm
- CHA Cancer Prevention Research Center, CHA Cancer Institute, CHA University, Seongnam, Korea
| | - Suntaek Hong
- Laboratory of Cancer Cell Biology, Department of Biochemistry, School of Medicine, Gachon University, Incheon, Korea
| |
Collapse
|
49
|
TAK1 inhibition subverts the osteoclastogenic action of TRAIL while potentiating its antimyeloma effects. Blood Adv 2017; 1:2124-2137. [PMID: 29296860 DOI: 10.1182/bloodadvances.2017008813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) agonists induce tumor-specific apoptosis indicating that they may be an attractive therapeutic strategy against cancers, including multiple myeloma (MM). Osteoclastogenesis is highly induced in MM, which in turn enhances MM growth, thereby forming a vicious cycle between MM tumor expansion and bone destruction. However, the effects of TRAIL on MM-enhanced osteoclastogenesis remain largely unknown. Here, we show that TRAIL induced apoptosis in MM cells, but not in osteoclasts (OCs), and that it rather facilitated receptor activator of NF-κB ligand-induced osteoclastogenesis along with upregulation of cellular FLICE inhibitory protein (c-FLIP). TRAIL did not induce death-inducing signaling complex formation in OCs, but formed secondary complex (complex II) with the phosphorylation of transforming growth factor β-activated kinase-1 (TAK1), and thus activated NF-κB signaling. c-FLIP knockdown abolished complex II formation, thus permitting TRAIL induction of OC cell death. The TAK1 inhibitor LLZ1640-2 abrogated the TRAIL-induced c-FLIP upregulation and NF-κB activation, and triggered TRAIL-induced caspase-8 activation and cell death in OCs. Interestingly, the TRAIL-induced caspase-8 activation caused enzymatic degradation of the transcription factor Sp1 to noticeably reduce c-FLIP expression, which further sensitized OCs to TRAIL-induced apoptosis. Furthermore, the TAK1 inhibition induced antiosteoclastogenic activity by TRAIL even in cocultures with MM cells while potentiating TRAIL's anti-MM effects. These results demonstrated that osteoclastic lineage cells use TRAIL for their differentiation and activation through tilting caspase-8-dependent apoptosis toward NF-κB activation, and that TAK1 inhibition subverts TRAIL-mediated NF-κB activation to resume TRAIL-induced apoptosis in OCs while further enhancing MM cell death in combination with TRAIL.
Collapse
|
50
|
Chen L, Xiong YQ, Xu J, Wang JP, Meng ZL, Hong YQ. Juglanin inhibits lung cancer by regulation of apoptosis, ROS and autophagy induction. Oncotarget 2017; 8:93878-93898. [PMID: 29212196 PMCID: PMC5706842 DOI: 10.18632/oncotarget.21317] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/27/2017] [Indexed: 12/31/2022] Open
Abstract
Juglanin (Jug) is obtained from the crude extract of Polygonum aviculare, exerting suppressive activity against cancer cell progression in vitro and in vivo. Juglanin administration causes apoptosis and reactive oxygen species (ROS) in different types of cells through regulating various signaling pathways. In our study, the effects of juglanin on non-small cell lung cancer were investigated. A significant role of juglanin in suppressing lung cancer growth was observed. Juglanin promoted apoptosis in lung cancer cells through increasing Caspase-3 and poly ADP-ribose polymerase (PARP) cleavage, which is regulated by TNF-related apoptosis-inducing ligand/Death receptors (TRAIL/DRs) relied on p53 activation. Anti-apoptotic members Bcl-2 and Bcl-xl were reduced, and pro-apoptotic members Bax and Bad were enhanced in cells and animals receiving juglanin. Additionally, nuclear factor-κB (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinases (MAPKs) activation were inhibited by juglanin. Further, juglanin improved ROS and induced autophagy. ROS inhibitor N-acetyl-l-cysteine (NAC) reversed apoptosis induced by juglanin in cancer cells. The formation of autophagic vacoules and LC3/autophagy gene7 (ATG7)/Beclin1 (ATG6) over-expression were observed in juglanin-treated cells. Also, juglanin administration to mouse xenograft models inhibited lung cancer progression. Our study demonstrated that juglanin could be a promising candidate against human lung cancer progression.
Collapse
Affiliation(s)
- Liang Chen
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Ya-Qiong Xiong
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Jing Xu
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Ji-Peng Wang
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Zi-Li Meng
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Yong-Qing Hong
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| |
Collapse
|