1
|
Cartland SP, Patil MS, Kelland E, Le N, Boccanfuso L, Stanley CP, Cholan PM, Dona MI, Patrick R, McGrath J, Su QP, Alwis I, Ganss R, Powell JE, Harvey RP, Pinto AR, Griffith TS, Loa J, Aitken SJ, Robinson DA, Patel S, Kavurma MM. The generation of stable microvessels in ischemia is mediated by endothelial cell derived TRAIL. SCIENCE ADVANCES 2024; 10:eadn8760. [PMID: 39365855 PMCID: PMC11451529 DOI: 10.1126/sciadv.adn8760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/28/2024] [Indexed: 10/06/2024]
Abstract
Reversal of ischemia is mediated by neo-angiogenesis requiring endothelial cell (EC) and pericyte interactions to form stable microvascular networks. We describe an unrecognized role for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in potentiating neo-angiogenesis and vessel stabilization. We show that the endothelium is a major source of TRAIL in the healthy circulation compromised in peripheral artery disease (PAD). EC deletion of TRAIL in vivo or in vitro inhibited neo-angiogenesis, pericyte recruitment, and vessel stabilization, resulting in reduced lower-limb blood perfusion with ischemia. Activation of the TRAIL receptor (TRAIL-R) restored blood perfusion and stable blood vessel networks in mice. Proof-of-concept studies showed that Conatumumab, an agonistic TRAIL-R2 antibody, promoted vascular sprouts from explanted patient arteries. Single-cell RNA sequencing revealed heparin-binding EGF-like growth factor in mediating EC-pericyte communications dependent on TRAIL. These studies highlight unique TRAIL-dependent mechanisms mediating neo-angiogenesis and vessel stabilization and the potential of repurposing TRAIL-R2 agonists to stimulate stable and functional microvessel networks to treat ischemia in PAD.
Collapse
Affiliation(s)
- Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| | - Manisha S. Patil
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| | - Elaina Kelland
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| | - Natalie Le
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| | - Lauren Boccanfuso
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Christopher P. Stanley
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| | | | | | - Ralph Patrick
- Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | - Qian Peter Su
- School of Biomedical Engineering, University of Technology, Sydney, Australia
- Heart Research Institute, Sydney, Australia
| | - Imala Alwis
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Ruth Ganss
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, Australia
| | - Joseph E. Powell
- Garvan-Weizmann Centre for Cellular Genomics, Sydney, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| | | | | | - Jacky Loa
- Royal Prince Alfred Hospital, Sydney, Australia
| | - Sarah J. Aitken
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Concord Institute of Academic Surgery, Concord Hospital, Sydney, Australia
| | - David A. Robinson
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
- Royal Prince Alfred Hospital, Sydney, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Royal Prince Alfred Hospital, Sydney, Australia
| | - Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Sydney, Australia
| |
Collapse
|
2
|
Deng Q, Chen L, Zhang G, Liu L, Luo SM, Gao X. TRIAL-based combination therapies in cancers. Int Immunopharmacol 2024; 138:112570. [PMID: 38971105 DOI: 10.1016/j.intimp.2024.112570] [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: 05/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) shows promising therapeutic potential in cancer treatment as it is able to trigger extrinsic apoptotic pathways by binding to the cognate death receptor, causing broad-spectrum apoptosis in cancer cells with negligible toxicity to normal cells. However, the majority of cancers display resistance to TRAIL, limiting its clinical utility. Overcoming resistance to TRAIL therapies remains a challenge in the development of effective anti-cancer strategies. To address the limitations of TRAIL therapy, a viable alternative approach involves combining TRAIL with more potent drugs compared to monotherapy. This combination strategy aims to induce synergistic effects or sensitize drug-resistant cancer cells. This review provides an overview of relevant modalities of TRAIL combination therapy, highlighting different drug classes. The findings demonstrate that combining TRAIL with other agents can effectively counteract resistance observed with TRAIL therapies in cancer. These findings lay a foundation for future advancements in TRAIL-based therapies for treating various cancers.
Collapse
Affiliation(s)
- Qiumin Deng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Luxuan Chen
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gui Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shi-Ming Luo
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China.
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
3
|
Huang M, Liu Y, Cheng Y, Dai W. Role of inflammatory biomarkers in mediating the effect of lipids on spontaneous intracerebral hemorrhage: a two-step, two-sample Mendelian randomization study. Front Neurol 2024; 15:1411555. [PMID: 39170073 PMCID: PMC11337198 DOI: 10.3389/fneur.2024.1411555] [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: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Background Spontaneous intracerebral hemorrhage (sICH) is a form of stroke with high mortality rates and significant neurological implications for patients. Abnormalities in lipid metabolism have been implicated in various cardiovascular diseases, yet their relationship with sICH remains insufficiently explored, particularly concerning their association with inflammatory factors. Methods Employing a two-sample, two-step Mendelian Randomization approach, combined with data from GWAS datasets, to investigate the causal relationship between plasma lipid levels and sICH. Additionally, the role of inflammatory factors in this relationship was examined, and sensitivity analyses were conducted to ensure the robustness of the results. Results The results indicate a significant causal relationship between 19 plasma lipid metabolites and sICH. Furthermore, mediation analysis revealed that three distinct lipids, namely Sterol ester (27:1/20:2), Phosphatidylcholine (16:0_20:4), and Sphingomyelin (d34:1), exert their influence on sICH through inflammatory factors. TRAIL (OR: 1.078, 95% CI: 1.016-1.144, p = 0.013) and HGF (OR: 1.131, 95% CI: 1.001-1.279, p = 0.049) were identified as significant mediators. Conclusion This study provides new evidence linking abnormalities in lipid metabolism with sICH and elucidates the role of inflammatory factors as mediators. These findings contribute to a better understanding of the pathogenesis of sICH and offer novel insights and therapeutic strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Mingsheng Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiheng Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiran Dai
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Luo C, He S, Shi F, Zhou J, Shang L. The Role of TRAIL Signaling in Cancer: Searching for New Therapeutic Strategies. BIOLOGY 2024; 13:521. [PMID: 39056714 PMCID: PMC11274015 DOI: 10.3390/biology13070521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Cancer continues to pose a significant threat to global health, with its status as a leading cause of death remaining unchallenged. Within the realm of cancer research, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) stands out as a critical player, having been identified in the 1990s as the tenth member of the TNF family. This review examines the pivotal role of TRAIL in cancer biology, focusing on its ability to induce apoptosis in malignant cells through both endogenous and exogenous pathways. We provide an in-depth analysis of TRAIL's intracellular signaling and intercellular communication, underscoring its potential as a selective anticancer agent. Additionally, the review explores TRAIL's capacity to reshape the tumor microenvironment, thereby influencing cancer progression and response to therapy. With an eye towards future developments, we discuss the prospects of harnessing TRAIL's capabilities for the creation of tailored, precision-based cancer treatments, aiming to enhance efficacy and improve patient survival rates.
Collapse
Affiliation(s)
- Cheng Luo
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Shan He
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jianhua Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| | - Li Shang
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| |
Collapse
|
5
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [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: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
6
|
Kelland E, Patil MS, Patel S, Cartland SP, Kavurma MM. The Prognostic, Diagnostic, and Therapeutic Potential of TRAIL Signalling in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24076725. [PMID: 37047698 PMCID: PMC10095395 DOI: 10.3390/ijms24076725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) was originally discovered, almost 20 years ago, for its ability to kill cancer cells. More recent evidence has described pleiotropic functions, particularly in the cardiovascular system. There is potential for TRAIL concentrations in the circulation to act as prognostic and/or diagnostic factors for cardiovascular diseases (CVD). Pre-clinical studies also describe the therapeutic capacity for TRAIL signals, particularly in the context of atherosclerotic disease and diseases of the myocardium. Because diabetes mellitus significantly contributes to the progression and pathogenesis of CVDs, in this review we highlight recent evidence for the prognostic, diagnostic, and therapeutic potential of TRAIL signals in CVDs, and where relevant, the impact of diabetes mellitus. A greater understanding of how TRAIL signals regulate cardiovascular protection and pathology may offer new diagnostic and therapeutic avenues for patients suffering from CVDs.
Collapse
Affiliation(s)
- Elaina Kelland
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Manisha S. Patil
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
- Royal Prince Alfred Hospital, Sydney 2006, Australia
| | - Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| |
Collapse
|
7
|
Montinaro A, Walczak H. Harnessing TRAIL-induced cell death for cancer therapy: a long walk with thrilling discoveries. Cell Death Differ 2023; 30:237-249. [PMID: 36195672 PMCID: PMC9950482 DOI: 10.1038/s41418-022-01059-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 02/10/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) can induce apoptosis in a wide variety of cancer cells, both in vitro and in vivo, importantly without killing any essential normal cells. These findings formed the basis for the development of TRAIL-receptor agonists (TRAs) for cancer therapy. However, clinical trials conducted with different types of TRAs have, thus far, afforded only limited therapeutic benefit, as either the respectively chosen agonist showed insufficient anticancer activity or signs of toxicity, or the right TRAIL-comprising combination therapy was not employed. Therefore, in this review we will discuss molecular determinants of TRAIL resistance, the most promising TRAIL-sensitizing agents discovered to date and, importantly, whether any of these could also prove therapeutically efficacious upon cancer relapse following conventional first-line therapies. We will also discuss the more recent progress made with regards to the clinical development of highly active non-immunogenic next generation TRAs. Based thereupon, we next propose how TRAIL resistance might be successfully overcome, leading to the possible future development of highly potent, cancer-selective combination therapies that are based on our current understanding of biology TRAIL-induced cell death. It is possible that such therapies may offer the opportunity to tackle one of the major current obstacles to effective cancer therapy, namely overcoming chemo- and/or targeted-therapy resistance. Even if this were achievable only for certain types of therapy resistance and only for particular types of cancer, this would be a significant and meaningful achievement.
Collapse
Affiliation(s)
- Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany.
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
8
|
Kavurma MM, Bursill C, Stanley CP, Passam F, Cartland SP, Patel S, Loa J, Figtree GA, Golledge J, Aitken S, Robinson DA. Endothelial cell dysfunction: Implications for the pathogenesis of peripheral artery disease. Front Cardiovasc Med 2022; 9:1054576. [PMID: 36465438 PMCID: PMC9709122 DOI: 10.3389/fcvm.2022.1054576] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/24/2022] [Indexed: 08/27/2023] Open
Abstract
Peripheral artery disease (PAD) is caused by occluded or narrowed arteries that reduce blood flow to the lower limbs. The treatment focuses on lifestyle changes, management of modifiable risk factors and vascular surgery. In this review we focus on how Endothelial Cell (EC) dysfunction contributes to PAD pathophysiology and describe the largely untapped potential of correcting endothelial dysfunction. Moreover, we describe current treatments and clinical trials which improve EC dysfunction and offer insights into where future research efforts could be made. Endothelial dysfunction could represent a target for PAD therapy.
Collapse
Affiliation(s)
- Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Christina Bursill
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | | | - Freda Passam
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Central Clinical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jacky Loa
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
- The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia
| | - Sarah Aitken
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Concord Institute of Academic Surgery, Concord Hospital, Sydney, NSW, Australia
| | | |
Collapse
|
9
|
Aggarwal C, Saini K, Reddy ES, Singla M, Nayak K, Chawla YM, Maheshwari D, Singh P, Sharma P, Bhatnagar P, Kumar S, Gottimukkala K, Panda H, Gunisetty S, Davis CW, Kissick HT, Kabra SK, Lodha R, Medigeshi GR, Ahmed R, Murali-Krishna K, Chandele A. Immunophenotyping and Transcriptional Profiling of Human Plasmablasts in Dengue. J Virol 2021; 95:e0061021. [PMID: 34523972 PMCID: PMC8577383 DOI: 10.1128/jvi.00610-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/11/2021] [Indexed: 12/07/2022] Open
Abstract
Plasmablasts represent a specialized class of antibody-secreting effector B cells that transiently appear in blood circulation following infection or vaccination. The expansion of these cells generally tends to be massive in patients with systemic infections such as dengue or Ebola that cause hemorrhagic fever. To gain a detailed understanding of human plasmablast responses beyond antibody expression, here, we performed immunophenotyping and RNA sequencing (RNA-seq) analysis of the plasmablasts from dengue febrile children in India. We found that plasmablasts expressed several adhesion molecules and chemokines or chemokine receptors that are involved in endothelial interactions or homing to inflamed tissues, including skin, mucosa, and intestine, and upregulated the expression of several cytokine genes that are involved in leukocyte extravasation and angiogenesis. These plasmablasts also upregulated the expression of receptors for several B-cell prosurvival cytokines that are known to be induced robustly in systemic viral infections such as dengue, some of which generally tend to be relatively higher in patients manifesting hemorrhage and/or shock than in patients with mild febrile infection. These findings improve our understanding of human plasmablast responses during the acute febrile phase of systemic dengue infection. IMPORTANCE Dengue is globally spreading, with over 100 million clinical cases annually, with symptoms ranging from mild self-limiting febrile illness to more severe and sometimes life-threatening dengue hemorrhagic fever or shock, especially among children. The pathophysiology of dengue is complex and remains poorly understood despite many advances indicating a key role for antibody-dependent enhancement of infection. While serum antibodies have been extensively studied, the characteristics of the early cellular factories responsible for antibody production, i.e., plasmablasts, are only beginning to emerge. This study provides a comprehensive understanding of the transcriptional profiles of human plasmablasts from dengue patients.
Collapse
Affiliation(s)
- Charu Aggarwal
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Keshav Saini
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Elluri Seetharami Reddy
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | - Mohit Singla
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Kaustuv Nayak
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Yadya M. Chawla
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Deepti Maheshwari
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Prabhat Singh
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Pragati Sharma
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Biotechnology, School of Chemical and Life Sciences, New Delhi, India
| | - Priya Bhatnagar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- TERI School of Advanced Studies, New Delhi, India
| | - Sanjeev Kumar
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Kamalvishnu Gottimukkala
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Harekrushna Panda
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sivaram Gunisetty
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Carl W. Davis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Haydn Thomas Kissick
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sushil Kumar Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rakesh Lodha
- Department of Pediatrics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Microbiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
10
|
Cardoso Alves L, Corazza N, Micheau O, Krebs P. The multifaceted role of TRAIL signaling in cancer and immunity. FEBS J 2020; 288:5530-5554. [PMID: 33215853 DOI: 10.1111/febs.15637] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can lead to the induction of apoptosis in tumor or infected cells. However, activation of TRAIL signaling may also trigger nonapoptotic pathways in cancer and in nontransformed cells, that is, immune cells. Here, we review the current knowledge on noncanonical TRAIL signaling. The biological outcomes of TRAIL signaling in immune and malignant cells are presented and explained, with a focus on the role of TRAIL for natural killer (NK) cell function. Furthermore, we highlight the technical difficulties in dissecting the precise molecular mechanisms involved in the switch between apoptotic and nonapoptotic TRAIL signaling. Finally, we discuss the consequences thereof for a therapeutic manipulation of TRAIL in cancer and possible approaches to bypass these difficulties.
Collapse
Affiliation(s)
| | - Nadia Corazza
- Institute of Pathology, University of Bern, Switzerland
| | - Olivier Micheau
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | | |
Collapse
|
11
|
Cartland SP, Lin RCY, Genner S, Patil MS, Martínez GJ, Barraclough JY, Gloss B, Misra A, Patel S, Kavurma MM. Vascular transcriptome landscape of Trail -/- mice: Implications and therapeutic strategies for diabetic vascular disease. FASEB J 2020; 34:9547-9562. [PMID: 32501591 DOI: 10.1096/fj.201902785r] [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: 11/05/2019] [Revised: 04/30/2020] [Accepted: 05/08/2020] [Indexed: 12/20/2022]
Abstract
Circulating plasma TRAIL levels are suppressed in patients with cardiovascular and diabetic diseases. To identify novel targets in vascular metabolic diseases, genome-wide transcriptome of aortic tissue from Trail-/- versus Trail+/+ mice were interrogated. We found 861 genes differentially expressed with TRAIL deletion. Gene enrichment analyses showed many of these genes were related to inflammation, cell-to-cell cytoskeletal interactions, and transcriptional modulation. We identified vascular protective and pathological gene clusters, with Ifi205 as the most significantly reduced vascular protective gene, whereas Glut1, the most significantly increased pathological gene with TRAIL deletion. We hypothesized that therapeutic targets could be devised from such integrated analysis and validated our findings from vascular tissues of diabetic mice. From the differentially expressed gene targets, enriched transcription factor (TF) and microRNA binding motifs were identified. The top two TFs were Elk1 and Sp1, with enrichment to eight gene targets common to both. miR-520d-3p and miR-377-3p were the top enriched microRNAs with TRAIL deletion; with four overlapping genes enriched for both microRNAs. Our findings offer an alternate in silico approach for therapeutic target identification and present a deeper understanding of gene signatures and pathways altered with TRAIL suppression in the vasculature.
Collapse
Affiliation(s)
- Siân P Cartland
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ruby C Y Lin
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Scott Genner
- Heart Research Institute, Sydney, NSW, Australia
| | - Manisha S Patil
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gonzalo J Martínez
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Westmead Institute for Medical Research, Sydney, NSW, Australia.,División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile
| | - Jennifer Y Barraclough
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Brian Gloss
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Mary M Kavurma
- Heart Research Institute, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Nash M, McGrath JP, Cartland SP, Patel S, Kavurma MM. Tumour necrosis factor superfamily members in ischaemic vascular diseases. Cardiovasc Res 2020; 115:713-720. [PMID: 30816914 DOI: 10.1093/cvr/cvz042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/25/2018] [Accepted: 02/26/2019] [Indexed: 12/20/2022] Open
Abstract
Current treatment of ischaemic vascular diseases such as coronary and peripheral artery disease includes angioplasty and bypass grafting, as well as lipid lowering therapies and control of other cardiovascular risk factors. Numerous members of the tumour necrosis factor superfamily (TNFSF) have recently shown emerging roles in both the protection and progression of such diseases. Understanding the role TNFSF members play in ischaemic vascular disease may provide insight into the development of novel therapeutics to prevent or treat diseases relating to atherosclerosis and ischaemia. This review summarizes the most recent findings relating to TNFSF members and the mechanisms that precede ischaemic vascular disease progression, particularly endothelial dysfunction, chronic inflammation, and atherosclerotic plaque development. This review also explores recent translational research on the role of TNFSF therapies in cardiovascular disease.
Collapse
Affiliation(s)
- Megan Nash
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney NSW, Australia.,School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.,Department of Biochemistry, University of Bath, Bath, UK
| | - Jordan P McGrath
- Department of Cardiology, Royal Prince Alfred Hospital, Missenden Rd Camperdown, NSW, Australia
| | - Siân P Cartland
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney NSW, Australia.,School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Sanjay Patel
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney NSW, Australia.,School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Missenden Rd Camperdown, NSW, Australia
| | - Mary M Kavurma
- Heart Research Institute, 7 Eliza Street, Newtown, Sydney NSW, Australia.,School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
13
|
TRAIL in oncology: From recombinant TRAIL to nano- and self-targeted TRAIL-based therapies. Pharmacol Res 2020; 155:104716. [PMID: 32084560 DOI: 10.1016/j.phrs.2020.104716] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 12/18/2022]
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) selectively induces the apoptosis pathway in tumor cells leading to tumor cell death. Because TRAIL induction can kill tumor cells, cancer researchers have developed many agents to target TRAIL and some of these agents have entered clinical trials in oncology. Unfortunately, these trials have failed for many reasons, including drug resistance, off-target toxicities, short half-life, and specifically in gene therapy due to the limited uptake of TRAIL genes by cancer cells. To address these drawbacks, translational researchers have utilized drug delivery platforms. Although, these platforms can improve TRAIL-based therapies, they are unable to sufficiently translate the full potential of TRAIL-targeting to clinically viable products. Herein, we first summarize the complex biology of TRAIL signaling, including TRAILs cross-talk with other signaling pathways and immune cells. Next, we focus on known resistant mechanisms to TRAIL-based therapies. Then, we discuss how nano-formulation has the potential to enhance the therapeutic efficacy of TRAIL protein. Finally, we specify strategies with the potential to overcome the challenges that cannot be addressed via nanotechnology alone, including the alternative methods of TRAIL-expressing circulating cells, tumor-targeting bacteria, viruses, and exosomes.
Collapse
|
14
|
Cartland SP, Genner SW, Martínez GJ, Robertson S, Kockx M, Lin RC, O'Sullivan JF, Koay YC, Manuneedhi Cholan P, Kebede MA, Murphy AJ, Masters S, Bennett MR, Jessup W, Kritharides L, Geczy C, Patel S, Kavurma MM. TRAIL-Expressing Monocyte/Macrophages Are Critical for Reducing Inflammation and Atherosclerosis. iScience 2019; 12:41-52. [PMID: 30665196 PMCID: PMC6348195 DOI: 10.1016/j.isci.2018.12.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/12/2018] [Accepted: 12/28/2018] [Indexed: 12/31/2022] Open
Abstract
Circulating tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) levels are reduced in patients with cardiovascular disease, and TRAIL gene deletion in mice exacerbates atherosclerosis and inflammation. How TRAIL protects against atherosclerosis and why levels are reduced in disease is unknown. Here, multiple strategies were used to identify the protective source of TRAIL and its mechanism(s) of action. Samples from patients with coronary artery disease and bone-marrow transplantation experiments in mice lacking TRAIL revealed monocytes/macrophages as the main protective source. Accordingly, deletion of TRAIL caused a more inflammatory macrophage with reduced migration, displaying impaired reverse cholesterol efflux and efferocytosis. Furthermore, interleukin (IL)-18, commonly increased in plasma of patients with cardiovascular disease, negatively regulated TRAIL transcription and gene expression, revealing an IL-18-TRAIL axis. These findings demonstrate that TRAIL is protective of atherosclerosis by modulating monocyte/macrophage phenotype and function. Manipulating TRAIL levels in these cells highlights a different therapeutic avenue in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Siân P Cartland
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Scott W Genner
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia
| | - Gonzalo J Martínez
- Sydney Medical School, University of Sydney, Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia; División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Stacy Robertson
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | | | - Ruby Cy Lin
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - John F O'Sullivan
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Yen Chin Koay
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Pradeep Manuneedhi Cholan
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Melkam A Kebede
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | | | - Seth Masters
- Walter and Elisa Hall Institute of Medical Research, Melbourne, Australia
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Carolyn Geczy
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Sanjay Patel
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Mary M Kavurma
- Heart Research Institute, 7 Eliza St, Newtown, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia.
| |
Collapse
|
15
|
Manuneedhi Cholan P, Cartland SP, Dang L, Rayner BS, Patel S, Thomas SR, Kavurma MM. TRAIL protects against endothelial dysfunction in vivo and inhibits angiotensin-II-induced oxidative stress in vascular endothelial cells in vitro. Free Radic Biol Med 2018; 126:341-349. [PMID: 30165101 DOI: 10.1016/j.freeradbiomed.2018.08.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022]
Abstract
The vascular endothelium is critical for maintenance of cardiovascular homeostasis. Endothelial dysfunction is a key event of atherosclerosis, with oxidative stress mediated by reactive oxygen species (ROS) playing a major role. Tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is increasingly recognised to play a protective role in atherosclerosis, however the molecular mechanisms by which it exerts its beneficial effects are unclear. Here we examined if TRAIL could attenuate vascular oxidative stress and improve endothelial cell (EC) function. In coronary artery disease patients, plasma TRAIL levels were significantly reduced compared to healthy individuals, and negatively correlated with the levels of circulating 8-iso Prostaglandin F2α, a marker of in vivo oxidative stress. In vivo, high-fat fed, atherosclerotic Trail-/-Apoe-/- mice exhibited a significant impairment in endothelial-dependent vasorelaxation, which correlated with increased vascular ROS and 4-hydroxynonenal compared to Apoe-/- mice. Endothelial permeability measured by Evan's blue dye extravasation was increased in several organs of Trail-/- mice compared to wild-type mice, which correlated with a decrease in VE-cadherin expression. In vitro in ECs, angiotensin II (AngII)-induced ROS generation involving the mitochondria, NADPH oxidase-4 (NOX-4) and eNOS, was inhibited by pre-treatment with TRAIL. Furthermore, AngII-augmented VCAM-1 expression and monocyte adhesion to ECs was inhibited by TRAIL. Finally, AngII reduced VE-cadherin expression and redistributed this protein, all of which was brought back to baseline by TRAIL pre-treatment. These findings demonstrate for the first time that TRAIL protects against several forms of endothelial dysfunction involving its ability to control EC ROS generation. Understanding the role TRAIL plays in normal physiology and disease, may lead to potential new therapies to improve endothelial function and atherosclerosis.
Collapse
Affiliation(s)
- Pradeep Manuneedhi Cholan
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Siân P Cartland
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Lei Dang
- School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Benjamin S Rayner
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Sanjay Patel
- Sydney Medical School, The University of Sydney, Sydney 2006, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Shane R Thomas
- School of Medical Sciences, University of New South Wales, Sydney 2052, Australia
| | - Mary M Kavurma
- Heart Research Institute, Sydney 2042, Australia; Sydney Medical School, The University of Sydney, Sydney 2006, Australia.
| |
Collapse
|
16
|
An R, Schmid R, Klausing A, Robering JW, Weber M, Bäuerle T, Detsch R, Boccaccini AR, Horch RE, Boos AM, Weigand A. Proangiogenic effects of tumor cells on endothelial progenitor cells vary with tumor type in an in vitro and in vivo rat model. FASEB J 2018; 32:5587-5601. [PMID: 29746168 DOI: 10.1096/fj.201800135rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endothelial progenitor cells (EPCs) contribute to neovascularization in tumors. However, the relationship of EPCs and tumor-induced angiogenesis still remains to be clarified. The present study aimed at investigating the influence of 4 different tumor types on angiogenic properties of EPCs in an in vitro and in vivo rat model. It could be demonstrated that in vitro proliferation, migration, and angiogenic abilities and genetic modifications of EPCs are controlled in a tumor-type-dependent manner. The proangiogenic effect of mammary carcinoma, osteosarcoma, and rhabdomyosarcoma cells was more pronounced compared to colon carcinoma cells. Coinjection of encapsulated tumor cells, especially mammary carcinoma cells, and EPCs in a rat model confirmed a contributing effect of EPCs in tumor vascularization. Cytokines secreted by tumors such as monocyte chemoattractant protein 1, macrophage inflammatory protein 2, and TNF-related apoptosis-inducing ligand play a pivotal role in the tumor cell-EPC interaction, leading to enhanced migration and angiogenesis. With the present study, we were able to decipher possible underlying mechanisms by which EPCs are stimulated by tumor cells and contribute to tumor vascularization. The present study will contribute to a better understanding of tumor-induced vascularization, thus facilitating the development of therapeutic strategies targeting tumor-EPC interactions.-An, R., Schmid, R., Klausing, A., Robering, J. W., Weber, M., Bäuerle, T., Detsch, R., Boccaccini, A. R., Horch, R. E., Boos, A. M., Weigand, A. Proangiogenic effects of tumor cells on endothelial progenitor cells vary with tumor type in an in vitro and in vivo rat model.
Collapse
Affiliation(s)
- Ran An
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.,Union Plastic and Aesthetic Hospital, Huazhong University of Science and Technology, Wuhan Union Hospital, Wuhan, Hubei, China
| | - Rafael Schmid
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anne Klausing
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jan W Robering
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maximilian Weber
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tobias Bäuerle
- Department of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany; and
| | - Rainer Detsch
- Department of Materials Science and Engineering, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Aldo R Boccaccini
- Department of Materials Science and Engineering, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja M Boos
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Annika Weigand
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
17
|
The Role of NOX4 and TRX2 in Angiogenesis and Their Potential Cross-Talk. Antioxidants (Basel) 2017; 6:antiox6020042. [PMID: 28594389 PMCID: PMC5488022 DOI: 10.3390/antiox6020042] [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: 04/18/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
The nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) family is the major source of reactive oxygen species (ROS) in the vascular system. In this family, NOX4, a constitutive active form of NOXs, plays an important role in angiogenesis. Thioredoxin 2 (TRX2) is a key mitochondrial redox protein that maintains normal protein function and also provides electrons to peroxiredoxin 3 (PRX3) to scavenge H₂O₂ in mitochondria. Angiogenesis, a process of new blood vessel formation, is involved in a variety of physiological processes and pathological conditions. It seems to be paradoxical for ROS-producing NOX4 and ROS-scavenging TRX2 to have a similar role in promoting angiogenesis. In this review, we will focus on data supporting the role of NOX4 and TRX2 in angiogenesis and their cross-talks and discuss how ROS can positively or negatively regulate angiogenesis, depending on their species, levels and locations. NOX4 and TRX2-mediated ROS signaling could be promising targets for the treatment of angiogenesis-related diseases.
Collapse
|
18
|
von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer 2017; 17:352-366. [PMID: 28536452 DOI: 10.1038/nrc.2017.28] [Citation(s) in RCA: 421] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The discovery that the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis of cancer cells without causing toxicity in mice has led to the in-depth study of pro-apoptotic TRAIL receptor (TRAIL-R) signalling and the development of biotherapeutic drug candidates that activate TRAIL-Rs. The outcome of clinical trials with these TRAIL-R agonists has, however, been disappointing so far. Recent evidence indicates that many cancers, in addition to being TRAIL resistant, use the endogenous TRAIL-TRAIL-R system to their own advantage. However, novel insight on two fronts - how resistance of cancer cells to TRAIL-based pro-apoptotic therapies might be overcome, and how the pro-tumorigenic effects of endogenous TRAIL might be countered - gives reasonable hope that the TRAIL system can be harnessed to treat cancer. In this Review we assess the status quo of our understanding of the biology of the TRAIL-TRAIL-R system - as well as the gaps therein - and discuss the opportunities and challenges in effectively targeting this pathway.
Collapse
Affiliation(s)
- Silvia von Karstedt
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
19
|
El Sharkawi FZ, Ewais SM, Fahmy RH, Rashed LA. PTEN and TRAIL genes loaded zein nanoparticles as potential therapy for hepatocellular carcinoma. J Drug Target 2017; 25:513-522. [PMID: 28140697 DOI: 10.1080/1061186x.2017.1289536] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene therapy is one of the recent approaches in treatment of hepatocellular carcinoma (HCC). Development of a vector or vehicle that can selectively and efficiently deliver the gene to target cells with minimal toxicity is an urgent demand. In the present study, phosphatase and tensin homolog (PTEN) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) genes were loaded to zein nanoparticles (ZNPs). The formulated PTEN and TRAIL-loaded ZNPs were tested for their in vitro and in vivo potential antitumor efficacy using liver tumor cells (HepG2) and HCC-induced rats as animal model. Also, mRNA expression of p53, VGEF and MMP-2 were carried out as markers of apoptosis, angiogenesis and metastasis in animal liver tissues. The results of the study showed that both PTEN and TRAIL-loaded ZNPs proved anti-proliferative activity against HepG2 cell lines with IC50 values of 0.09, 0.25 µg/ml, respectively. In vivo assay confirmed decrease in mRNA expression of both VEGF and MMP-2 with increased in P53 expression level in liver tissues of the treated animals. Therefore, authors introduced new integration between gene therapy and nanotechnology in the form of PTEN and TRAIL-loaded ZNPs that proved potential to be used in gene therapy for the treatment of HCC.
Collapse
Affiliation(s)
- Fathia Zaki El Sharkawi
- a Department of Biochemistry and Molecular Biology, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Shaimaa Mohammed Ewais
- a Department of Biochemistry and Molecular Biology, Faculty of Pharmacy , Helwan University , Cairo , Egypt
| | - Rania Hassan Fahmy
- b Department of Pharmaceutics and Industrial pharmacy, Faculty of Pharmacy , Cairo University , Cairo , Egypt.,c Department of Pharmaceutics, Faculty of Pharmacy , Ahram Canadian University , Giza , Egypt
| | - Laila Ahmed Rashed
- d Department of Biochemistry and Molecular Biology, Faculty of Medicine , Cairo University , Cairo , Egypt
| |
Collapse
|
20
|
Kanatli I, Akkaya B, Uysal H, Kahraman S, Sanlioglu AD. Analysis of TNF-related apoptosis-inducing ligand and receptors and implications in thymus biology and myasthenia gravis. Neuromuscul Disord 2016; 27:128-135. [PMID: 28012741 DOI: 10.1016/j.nmd.2016.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/20/2016] [Accepted: 10/31/2016] [Indexed: 11/26/2022]
Abstract
Myasthenia Gravis is an autoantibody-mediated, neuromuscular junction disease, and is usually associated with thymic abnormalities presented as thymic tumors (~10%) or hyperplastic thymus (~65%). The exact role of thymus in Myasthenia Gravis development is not clear, yet many patients benefit from thymectomy. The apoptotic ligand TNF-Related Apoptosis-Inducing Ligand is thought to be involved in the regulation of thymocyte counts, although conflicting results are reported. We investigated differential expression profiles of TNF-Related Apoptosis-Inducing Ligand and its transmembrane receptors, Nuclear Factor-kB activation status, and apoptotic cell counts in healthy thymic tissue and pathological thymus from Myasthenia Gravis patients. All tissues expressed TNF-Related Apoptosis-Inducing Ligand and its receptors, with hyperplastic tissue having the highest expression levels of death receptors DR4 and DR5. No detectable Nuclear Factor-kB activation, at least via the canonical Protein Kinase A-mediated p65 Ser276 phosphorylation, was evident in any of the tissues studied. Apoptotic cell counts were higher in MG-associated tissue compared to the normal thymus. Possible use of the TNF-Related Apoptosis-Inducing Ligand within the concept of an apoptotic ligand-mediated medical thymectomy in thymoma- or thymic hyperplasia-associated Myasthenia Gravis is also discussed.
Collapse
Affiliation(s)
- Irem Kanatli
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, 07058, Antalya, Turkey; Center for Gene and Cell Therapy, Akdeniz University, 07058 Antalya, Turkey
| | - Bahar Akkaya
- Department of Pathology, Faculty of Medicine, Akdeniz University, 07058 Antalya, Turkey
| | - Hilmi Uysal
- Department of Neurology, Faculty of Medicine, Akdeniz University, 07058 Antalya, Turkey
| | - Sevim Kahraman
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, 07058, Antalya, Turkey; Center for Gene and Cell Therapy, Akdeniz University, 07058 Antalya, Turkey
| | - Ahter Dilsad Sanlioglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, 07058, Antalya, Turkey; Center for Gene and Cell Therapy, Akdeniz University, 07058 Antalya, Turkey.
| |
Collapse
|
21
|
Cartland SP, Genner SW, Zahoor A, Kavurma MM. Comparative Evaluation of TRAIL, FGF-2 and VEGF-A-Induced Angiogenesis In Vitro and In Vivo. Int J Mol Sci 2016; 17:E2025. [PMID: 27918462 PMCID: PMC5187825 DOI: 10.3390/ijms17122025] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/24/2016] [Accepted: 11/25/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis-factor-related apoptosis-inducing ligand (TRAIL) has been implicated in angiogenesis; the growth of new blood vessels from an existing vessel bed. Our aim was to compare pro-angiogenic responses of TRAIL, vascular endothelial growth-factor-A (VEGF-A) and fibroblast growth-factor-2 (FGF-2) either separately (10 ng/mL) or in combination, followed by the assessment of proliferation, migration and tubule formation using human microvascular endothelial-1 (HMEC-1) cells in vitro. Angiogenesis was also measured in vivo using the Matrigel plug assay. TRAIL and FGF-2 significantly augmented HMEC-1 cell proliferation and migration, with combination treatment having an enhanced effect on cell migration only. In contrast, VEGF-A did not stimulate HMEC-1 migration at 10 ng/mL. Tubule formation was induced by all three factors, with TRAIL more effective compared to VEGF-A, but not FGF-2. TRAIL at 400 ng/mL, but not VEGF-A, promoted CD31-positive staining into the Matrigel plug. However, FGF-2 was superior, stimulating cell infiltration and angiogenesis better than TRAIL and VEGF-A in vivo. These findings demonstrate that each growth factor is more effective at different processes of angiogenesis in vitro and in vivo. Understanding how these molecules stimulate different processes relating to angiogenesis may help identify new strategies and treatments aimed at inhibiting or promoting dysregulated angiogenesis in people.
Collapse
Affiliation(s)
- Siân P Cartland
- Heart Research Institute, Sydney 2042, Australia.
- Sydney Medical School, University of Sydney, Sydney 2006, Australia.
| | | | - Amna Zahoor
- Heart Research Institute, Sydney 2042, Australia.
| | - Mary M Kavurma
- Heart Research Institute, Sydney 2042, Australia.
- Sydney Medical School, University of Sydney, Sydney 2006, Australia.
| |
Collapse
|
22
|
From bones to blood pressure, developing novel biologic approaches targeting the osteoprotegein pathway for pulmonary vascular disease. Pharmacol Ther 2016; 169:78-82. [PMID: 27373854 PMCID: PMC5243145 DOI: 10.1016/j.pharmthera.2016.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Osteoprotegerin (tnfsf11b, OPG) is a soluble member of the TNF superfamily originally described as an important regulator of osteoclastogenesis almost 20 years ago. OPG is a heparin-binding secreted glycoprotein that exists as a 55–62 kDa monomer or a 110–120 kDa disulphide-linked homodimer. Acting as a soluble decoy receptor for RANKL, OPG actively regulates RANK signalling, and thereby osteoclastogenesis. OPG has subsequently been shown to also be a decoy receptor TNF related apoptosis inducing-ligand (tnfsf10, TRAIL, Apo2L). TRAIL is a type II transmembrane protein that is widely expressed in a variety of human tissues, including the spleen, lung, and prostate. Through binding to TRAIL, OPG can inhibit TRAIL-induced apoptosis of cancer cells. More recently, OPG has been demonstrated to be secreted by, and influence, vascular smooth muscle cells phenotype particularly related to vascular calcification and pulmonary vascular remodelling. In pulmonary artery smooth muscle cell (PASMC) suppression of BMP, induction of 5-HT and IL-1 signalling have been shown to stimulate the release of OPG in vitro, which causes cell migration and proliferation. Patients with idiopathic PAH (IPAH) demonstrate increased circulating and tissue levels of OPG, and circulating serum levels predict survival. In pre-clinical models, OPG levels correlate with disease severity. Since OPG is a naturally circulating protein, we are investigating the potential of novel biologic antibody therapies to rescue PAH phenotype in disease models. Further pre-clinical and mechanistic data are forthcoming, but we believe current published data identify OPG as an exciting and novel therapeutic target in PAH.
Collapse
|
23
|
Regenfuß B, Dreisow ML, Hos D, Masli S, Bock F, Cursiefen C. The Naïve Murine Cornea as a Model System to Identify Novel Endogenous Regulators of Lymphangiogenesis: TRAIL and rtPA. Lymphat Res Biol 2016; 13:76-84. [PMID: 26091403 DOI: 10.1089/lrb.2015.0004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In the murine cornea, which is an established model for analyzing pathologic lymphatic vessel growth, phenotypic heterogeneity of the endogenous lymphatic vessels in the limbus of the cornea was previously described. In this study, the cornea of BALB/c, C57BL/6, and FVB mice with different limbal lymphangiogenic phenotypes was analyzed to identify novel candidates potentially influencing lymphatic vessel growth. METHODS AND RESULTS Pathway specific expression analysis of the cornea was performed to identify novel candidate genes. Corneal protein expression of the respective candidates was analyzed by fluorescent immunohistochemistry. The effect of the candidates on proliferation of human dermal lymphatic endothelial cells (HDLECs) was analyzed by BrdU proliferation ELISA. Thirteen genes were differentially regulated in corneas of mouse strains with more endogenous limbal lymphatic vessels (high-lymphangiogenic) (C57BL/6) compared to mouse strains with less endogenous limbal lymphatic vessels (low-lymphangiogenic) (BALB/c, FVB). Two candidates, Tumor necrosis factor (ligand) superfamily member 10 (Tnfsf10/Trail) and Plasminogen activator, tissue (Plat/tPA) were expressed in the cornea of BALB/c and C57BL/6 mice on the protein level. In vitro, Trail and recombinant tPA inhibited the proliferation of human dermal lymphatic endothelial cells. CONCLUSION Molecular analysis of the naive cornea in mouse strains with different limbal lymphatic phenotypes is a valuable model to identify novel endogenous regulators of lymphangiogenesis.
Collapse
Affiliation(s)
- Birgit Regenfuß
- 1 Department of Ophthalmology, University of Cologne , Germany
| | | | - Deniz Hos
- 1 Department of Ophthalmology, University of Cologne , Germany
| | - Sharmila Masli
- 2 Department of Ophthalmology, Boston University School of Medicine , Boston, Massachusetts
| | - Felix Bock
- 1 Department of Ophthalmology, University of Cologne , Germany
| | - Claus Cursiefen
- 1 Department of Ophthalmology, University of Cologne , Germany
| |
Collapse
|
24
|
Di Bartolo BA, Cartland SP, Prado-Lourenco L, Griffith TS, Gentile C, Ravindran J, Azahri NSM, Thai T, Yeung AWS, Thomas SR, Kavurma MM. Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Promotes Angiogenesis and Ischemia-Induced Neovascularization Via NADPH Oxidase 4 (NOX4) and Nitric Oxide-Dependent Mechanisms. J Am Heart Assoc 2015; 4:e002527. [PMID: 26572549 PMCID: PMC4845240 DOI: 10.1161/jaha.115.002527] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/02/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has the ability to inhibit angiogenesis by inducing endothelial cell death, as well as being able to promote pro-angiogenic activity in vitro. These seemingly opposite effects make its role in ischemic disease unclear. Using Trail(-/-) and wildtype mice, we sought to determine the role of TRAIL in angiogenesis and neovascularization following hindlimb ischemia. METHODS AND RESULTS Reduced vascularization assessed by real-time 3-dimensional Vevo ultrasound imaging and CD31 staining was evident in Trail(-/-) mice after ischemia, and associated with reduced capillary formation and increased apoptosis. Notably, adenoviral TRAIL administration significantly improved limb perfusion, capillary density, and vascular smooth-muscle cell content in both Trail(-/-) and wildtype mice. Fibroblast growth factor-2, a potent angiogenic factor, increased TRAIL expression in human microvascular endothelial cell-1, with fibroblast growth factor-2-mediated proliferation, migration, and tubule formation inhibited with TRAIL siRNA. Both fibroblast growth factor-2 and TRAIL significantly increased NADPH oxidase 4 (NOX4) expression. TRAIL-inducible angiogenic activity in vitro was inhibited with siRNAs targeting NOX4, and consistent with this, NOX4 mRNA was reduced in 3-day ischemic hindlimbs of Trail(-/-) mice. Furthermore, TRAIL-induced proliferation, migration, and tubule formation was blocked by scavenging H2O2, or by inhibiting nitric oxide synthase activity. Importantly, TRAIL-inducible endothelial nitric oxide synthase phosphorylation at Ser-1177 and intracellular human microvascular endothelial cell-1 cell nitric oxide levels were NOX4 dependent. CONCLUSIONS This is the first report demonstrating that TRAIL can promote angiogenesis following hindlimb ischemia in vivo. The angiogenic effect of TRAIL on human microvascular endothelial cell-1 cells is downstream of fibroblast growth factor-2, involving NOX4 and nitric oxide signaling. These data have significant therapeutic implications, such that TRAIL may improve the angiogenic response to ischemia and increase perfusion recovery in patients with cardiovascular disease and diabetes.
Collapse
Affiliation(s)
- Belinda Ann Di Bartolo
- The Heart Research Institute, Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.) The University of Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.)
| | - Siân Peta Cartland
- The Heart Research Institute, Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.) Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.) The University of Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.)
| | - Leonel Prado-Lourenco
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.)
| | | | - Carmine Gentile
- The Heart Research Institute, Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.) The University of Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.)
| | - Jayant Ravindran
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.) School of Medical Sciences, University of New South Wales, Sydney, Australia (J.R., T.T., S.R.T.)
| | - Nor Saadah Muhammad Azahri
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.)
| | - Thuan Thai
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.) School of Medical Sciences, University of New South Wales, Sydney, Australia (J.R., T.T., S.R.T.)
| | - Amanda Wing Shee Yeung
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.)
| | - Shane Ross Thomas
- Centre for Vascular Research, University of New South Wales, Sydney, Australia (S.P.C., L.P.L., J.R., N.S.M.A., T.T., A.W.S.Y., S.R.T.) School of Medical Sciences, University of New South Wales, Sydney, Australia (J.R., T.T., S.R.T.)
| | - Mary Meltem Kavurma
- The Heart Research Institute, Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.) The University of Sydney, NSW, Australia (B.A.D.B., S.P.C., C.G., M.M.K.)
| |
Collapse
|
25
|
Interaction between TNFR1 and TNFR2 dominates the clinicopathologic features of human hypopharyneal carcinoma. Tumour Biol 2015; 36:9421-9. [DOI: 10.1007/s13277-015-3684-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/16/2015] [Indexed: 01/08/2023] Open
|
26
|
Shivinsky A, Bronshtein T, Haber T, Machluf M. The effect of AZD2171- or sTRAIL/Apo2L-loaded polylactic-co-glycolic acid microspheres on a subcutaneous glioblastoma model. Biomed Microdevices 2015; 17:69. [DOI: 10.1007/s10544-015-9969-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Bolkun L, Lemancewicz D, Piszcz J, Moniuszko M, Bolkun-Skornicka U, Szkiladz M, Jablonska E, Kloczko J, Dzieciol J. Relationship between tumour necrosis factor-related apoptosis inducing ligand (TRAIL) and vascular endothelial growth factor in human multiple myeloma patients. Hematol Oncol 2014; 33:199-205. [PMID: 25370722 DOI: 10.1002/hon.2182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 09/26/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
Tumour necrosis factor-alfa (TNF-α) is an inflammatory cytokine with a wide spectrum of biological activity, including angiogenesis. Tumour necrosis factor-related apoptosis inducing ligand (TRAIL), which belongs to the TNF family of proteins, plays a role in the regulation of vascular responses, but its effect on the formation of new blood vessels (angiogenesis) is unclear. We analysed TRAIL concentrations in parallel with pro-angiogenic cytokines in serum and their expression in trephine biopsy (TB) in 56 patients with newly diagnosed IgG MM and 24 healthy volunteers. The study showed statistically higher concentrations of TRAIL and TNF-α, as well as of VEGF and its receptor, in MM patients compared to healthy volunteers and patients in advanced stages of the disease. Furthermore, we observed a significant decrease in all studied pro-angiogenic cytokines and significant increase of TRAIL concentration after anti-angiogenic therapy, with meaningful differences between responders (at least partial remission) and patients with progression during the induction treatment. It was also established that TRAIL correlated statistically and negatively with pro-angiogenic cytokines such as VEGF with its receptor and expression of VEGF and syndecan-1 in TB. In summary, our data indicate that in MM patients, both clinical course and treatment responsiveness are associated with dynamic yet corresponding changes of levels of TRAIL parallel pro-angiogenic mediators such as VEGF with its receptor and expression of VEGF and syndecan-1 in TB.
Collapse
Affiliation(s)
- Lukasz Bolkun
- Department of Haematology, Medical University of Bialystok, Poland
| | - Dorota Lemancewicz
- Department of Haematology, Medical University of Bialystok, Poland.,Department of Human Anatomy, Medical University of Bialystok, Poland
| | - Jaroslaw Piszcz
- Department of Haematology, Medical University of Bialystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Poland.,Department of Allergology and Internal Medicine, Medical University of Bialystok, Poland
| | | | | | - Ewa Jablonska
- Department of Immunology, Medical University of Bialystok, Poland
| | - Janusz Kloczko
- Department of Haematology, Medical University of Bialystok, Poland
| | - Janusz Dzieciol
- Department of Human Anatomy, Medical University of Bialystok, Poland
| |
Collapse
|
28
|
Myśliwska J, Ryba-Stanisławowska M, Smardzewski M, Słomiński B, Myśliwiec M, Siebert J. Enhanced apoptosis of monocytes from complication-free juvenile-onset diabetes mellitus type 1 may be ameliorated by TNF-α inhibitors. Mediators Inflamm 2014; 2014:946209. [PMID: 25053869 PMCID: PMC4099355 DOI: 10.1155/2014/946209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/28/2014] [Accepted: 05/01/2014] [Indexed: 01/17/2023] Open
Abstract
Diabetes mellitus type 1 is associated with an enhanced apoptosis of different cells and tissues, accelerating occurrence of diabetic microvascular complications. The aim of our study was to determine spontaneous apoptotic potential of the monocyte subsets in juvenile-onset complication-free diabetes mellitus type 1 and to compare them with the corresponding values of the healthy. Moreover, we wanted to assess effects of TNF-R1 blocking agents and those of general TNF-α blocker (Infliximab) on spontaneous apoptosis of monocytes. Sixty randomly selected DM1 patients (14.5 ± 3.2 years) and 30 healthy (13.5 ± 2.8 years) volunteers were enrolled in the study. Our results indicate that three monocyte subsets are distinguishable in the groups of young diabetic patients and the healthy, similarly to in the blood of adults. DM1 patients were characterized by higher values of apoptotic monocytes than the healthy. The manipulation with drugs inhibiting TNF-R1 expression diminished the pool of CD16(+) apoptotic monocytes. Infliximab reduced the apoptotic CD16(-) cells. In conclusion, diabetes mellitus type 1 is associated with greater apoptosis of three monocyte subsets which may contribute to the development of microvascular complications. TNF-α modifiers appear to ameliorate monocyte apoptosis. They may be useful for controlling excessive monocyte apoptosis in diabetic patients.
Collapse
Affiliation(s)
- Jolanta Myśliwska
- Department of Immunology, Medical University of Gdańsk, Ulica Dębinki 1, 80-211 Gdańsk, Poland
| | | | - Marcin Smardzewski
- Department of Immunology, Medical University of Gdańsk, Ulica Dębinki 1, 80-211 Gdańsk, Poland
| | - Bartosz Słomiński
- Department of Immunology, Medical University of Gdańsk, Ulica Dębinki 1, 80-211 Gdańsk, Poland
| | - Małgorzata Myśliwiec
- Academic Clinic of Pediatrics, Hematology, Oncology and Endocrinology, Medical University of Gdańsk, Ulica Dębinki 7, 80-211 Gdańsk, Poland
| | - Janusz Siebert
- Department of Family Medicine, Medical University of Gdańsk, Ulica Dębinki 2, 80-211 Gdańsk, Poland
| |
Collapse
|
29
|
Cantarella G, Di Benedetto G, Ribatti D, Saccani-Jotti G, Bernardini R. Involvement of caspase 8 and c-FLIPL in the proangiogenic effects of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). FEBS J 2014; 281:1505-1513. [PMID: 24438025 DOI: 10.1111/febs.12720] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/02/2013] [Accepted: 01/12/2014] [Indexed: 12/26/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), a cytokine of the tumour necrosis factor superfamily, is a potent cell-apoptosis inducer, although its effects vary as a function of concentration. In fact, low concentrations of TRAIL are associated with non-apoptotic effects, such as cell proliferation. Here, the effects of TRAIL at different concentrations have been evaluated on mitogenesis and migration on human umbilical vein endothelial cells (HUVEC) in vitro, as well as in the chick embryo chorioallantoic membrane (CAM) angiogenesis model in vivo. At low concentrations, TRAIL promoted either mitogenesis or migration of HUVEC, evaluated using the wound healing method. Cleavage of caspase 8 was evaluated along with expression of the caspase 8-like molecule, cellular FLICE-inhibitory protein (long form) (c-FLIPL ). Low concentrations of TRAIL failed to induce caspase 8 processing, whereas high concentrations induced apoptosis of HUVEC and activation of caspase 8. Moreover, TRAIL induced a significant angiogenic response in the CAM assay in vivo, comparable with that of vascular endothelial growth factor. These data suggest that the non-apoptotic effects of TRAIL include mitogenesis and increased mobility of endothelial cells, and eventually angiogenesis. In addition, the results demonstrate that the c-FLIPL level is also modulated by differences in TRAIL concentration, suggesting its involvement in the divergent effects of TRAIL. In conclusion, this study envisions a proangiogenic role of TRAIL, suggesting that TRAIL may represent a target for pharmacological manipulation.
Collapse
Affiliation(s)
- Giuseppina Cantarella
- Department of Clinical and Molecular Biomedicine, University of Catania Medical School, Italy
| | | | | | | | | |
Collapse
|
30
|
Na HJ, Hwang JY, Lee KS, Choi YK, Choe J, Kim JY, Moon HE, Kim KW, Koh GY, Lee H, Jeoung D, Won MH, Ha KS, Kwon YG, Kim YM. TRAIL negatively regulates VEGF-induced angiogenesis via caspase-8-mediated enzymatic and non-enzymatic functions. Angiogenesis 2013; 17:179-94. [PMID: 24097299 DOI: 10.1007/s10456-013-9387-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 09/12/2013] [Indexed: 01/23/2023]
Abstract
Solid tumors supply oxygen and nutrients required for angiogenesis by producing vascular endothelial growth factor (VEGF). Thus, inhibitors of VEGF signaling abrogate tumor angiogenesis, resulting in the suppression of tumor growth and metastasis. We here investigated the effects of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on VEGF-induced angiogenesis. TRAIL inhibited VEGF-induced in vitro angiogenesis of human umbilical vein endothelial cells (HUVECs) and in vivo neovascularization in chicken embryos and mice. TRAIL blocked VEGF-induced angiogenic signaling by inhibiting ERK, Src, FAK, paxillin, Akt, and eNOS. Further, TRAIL blocked intracellular Ca(2+) elevation and actin reorganization in HUVECs stimulated with VEGF, without inhibiting VEGF receptor-2 tyrosine phosphorylation. TRAIL increased caspase-8 activity, without inducing caspase-9/-3 activation and apoptosis. Moreover, TRAIL resulted in cleavage of FAK into FAK-related non-kinase-like fragments in VEGF-stimulated HUVECs, which was blocked by a caspase-8 inhibitor and cellular caspase-8-like inhibitory protein. Biochemical and pharmacological inhibition of caspase-8 and FAK blocked the inhibitory effects of TRAIL on VEGF-stimulated anti-angiogenic signaling and events. In addition, caspase-8 knockdown also suppressed VEGF-mediated signaling and angiogenesis, suggesting that procaspase-8 plays a role of a non-apoptotic modulator in VEGF-induced angiogenic signaling. These results suggest that TRAIL inhibits VEGF-induced angiogenesis by increasing caspase-8 activity and subsequently decreasing non-apoptotic signaling functions of procaspase-8, without inducing caspase-3 activation and endothelial cell cytotoxicity. These data indicate that caspase-8 may be used as an anti-angiogenic drug for solid tumors resistant to TRAIL and anti-tumor drugs.
Collapse
Affiliation(s)
- Hee-Jun Na
- Vascular Homeostasis Laboratory and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Guo L, Fan L, Ren J, Pang Z, Ren Y, Li J, Wen Z, Qian Y, Zhang L, Ma H, Jiang X. Combination of TRAIL and actinomycin D liposomes enhances antitumor effect in non-small cell lung cancer. Int J Nanomedicine 2012; 7:1449-60. [PMID: 22619505 PMCID: PMC3356168 DOI: 10.2147/ijn.s24711] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The intractability of non-small cell lung cancer (NSCLC) to multimodality treatments plays a large part in its extremely poor prognosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising cytokine for selective induction of apoptosis in cancer cells; however, many NSCLC cell lines are resistant to TRAIL-induced apoptosis. The therapeutic effect can be restored by treatments combining TRAIL with chemotherapeutic agents. Actinomycin D (ActD) can sensitize NSCLC cells to TRAIL-induced apoptosis by upregulation of death receptor 4 (DR4) or 5 (DR5). However, the use of ActD has significant drawbacks due to the side effects that result from its nonspecific biodistribution in vivo. In addition, the short half-life of TRAIL in serum also limits the antitumor effect of treatments combining TRAIL and ActD. In this study, we designed a combination treatment of long-circulating TRAIL liposomes and ActD liposomes with the aim of resolving these problems. The combination of TRAIL liposomes and ActD liposomes had a synergistic cytotoxic effect against A-549 cells. The mechanism behind this combination treatment includes both increased expression of DR5 and caspase activation. Moreover, systemic administration of the combination of TRAIL liposomes and ActD liposomes suppressed both tumor formation and growth of established subcutaneous NSCLC xenografts in nude mice, inducing apoptosis without causing significant general toxicity. These results provide preclinical proof-of-principle for a novel therapeutic strategy in which TRAIL liposomes are safely combined with ActD liposomes.
Collapse
Affiliation(s)
- Liangran Guo
- School of Pharmacy, Fudan University, Zhangheng Road, Shanghai, People’s Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Groth A, Salnikov AV, Ottinger S, Gladkich J, Liu L, Kallifatidis G, Salnikova O, Ryschich E, Giese N, Giese T, Momburg F, Büchler MW, Moldenhauer G, Herr I. New Gene-Immunotherapy Combining TRAIL-Lymphocytes and EpCAMxCD3 Bispecific Antibody for Tumor Targeting. Clin Cancer Res 2012; 18:1028-38. [DOI: 10.1158/1078-0432.ccr-11-2767] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Yang H, Wang Y, Qian H, Zhang P, Huang C. Pim protein kinase-3 is regulated by TNF-α and promotes endothelial cell sprouting. Mol Cells 2011; 32:235-41. [PMID: 21870113 PMCID: PMC3887632 DOI: 10.1007/s10059-011-1026-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 01/06/2023] Open
Abstract
Tumor necrosis factor-α (TNF-α) plays an important role in pathological angiogenesis associated with inflammatory response. Pim-3 kinase belonging to serine/threonine protein kinases is a potent suppressor of myc-induced apoptosis. We have recently demonstrated that Pim-3 plays an essential role in endothelial cell (EC) spreading and migration. In this study, we showed that TNF-α transiently increased Pim-3 mRNA expression, and this was mediated through Tumor necrosis factor-α receptor-1 (TNFR1) pathway in ECs. TNF-α could promote stabilization of Pim- 3 mRNA in ECs. Small-interfering RNA (siRNA)-mediated gene knockdown of Pim-3 significantly impaired TNF-α-induced formation of EC membrane protrusions in vitro. Furthermore, Pim-3 silencing inhibited EC sprouting in subcutaneous Matrigel in vivo. eNOS mRNA abundance was lower in Pim-3 siRNA transfected ECs compared with the control ECs. These observations suggest that Pim-3 plays a role in TNF-α-induced angiogenesis.
Collapse
MESH Headings
- Blotting, Western
- Cell Movement
- Cells, Cultured
- Collagen/chemistry
- Collagen/metabolism
- Drug Combinations
- Female
- Gene Expression
- Gene Silencing/drug effects
- Human Umbilical Vein Endothelial Cells/cytology
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Laminin/chemistry
- Laminin/metabolism
- Neoplasms/blood supply
- Neoplasms/pathology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proteoglycans/chemistry
- Proteoglycans/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA Stability
- RNA, Messenger/analysis
- RNA, Messenger/biosynthesis
- RNA, Small Interfering/pharmacology
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Handong Yang
- Department of Cardiovascular Diseases, Renmin Hospital of Wuhan University, Wuhan City, China
| | - Yinfang Wang
- Institute of Cardiovascular Science, Dongfeng Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Hang Qian
- Institute of Cardiovascular Science, Dongfeng Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Peng Zhang
- Institute of Cardiovascular Science, Dongfeng Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Congxin Huang
- Department of Cardiovascular Diseases, Renmin Hospital of Wuhan University, Wuhan City, China
| |
Collapse
|
34
|
TRAIL and doxorubicin combination enhances anti-glioblastoma effect based on passive tumor targeting of liposomes. J Control Release 2011; 154:93-102. [DOI: 10.1016/j.jconrel.2011.05.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 03/23/2011] [Accepted: 05/06/2011] [Indexed: 11/22/2022]
|
35
|
Guo L, Fan L, Ren J, Pang Z, Ren Y, Li J, Wen Z, Jiang X. A novel combination of TRAIL and doxorubicin enhances antitumor effect based on passive tumor-targeting of liposomes. NANOTECHNOLOGY 2011; 22:265105. [PMID: 21586819 DOI: 10.1088/0957-4484/22/26/265105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a novel anticancer agent for non-small cell lung cancer (NSCLC). However, approximately half of NSCLC cell lines are highly resistant to TRAIL. Doxorubicin (DOX) can sensitize NSCLC cells to TRAIL-induced apoptosis, indicating the possibility of combination therapy. Unfortunately, the therapeutic effect of a DOX and TRAIL combination is limited by multiple factors including the short serum half-life of TRAIL, poor compliance and application difficulty in the clinic, chronic DOX-induced cardiac toxicity, and the multidrug resistance (MDR) property of NSCLC cells. To solve such problems, we developed the combination of TRAIL liposomes (TRAIL-LP) and DOX liposomes (DOX-LP). An in vitro cytotoxicity study indicated that DOX-LP sensitized the NSCLC cell line A-549 to TRAIL-LP-induced apoptosis. Furthermore, this combination therapy of TRAIL-LP and DOX-LP displayed a stronger antitumor effect on NSCLC in xenografted mice when compared with free drugs or liposomal drugs alone. Therefore, the TRAIL-LP and DOX-LP combination therapy has excellent potential to become a new therapeutic approach for patients with advanced NSCLC.
Collapse
Affiliation(s)
- Liangran Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Two different docetaxel resistant MCF-7 sublines exhibited different gene expression pattern. Mol Biol Rep 2011; 39:3505-16. [PMID: 21720762 DOI: 10.1007/s11033-011-1123-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/20/2011] [Indexed: 12/20/2022]
Abstract
The objective of the present study was to investigate gene expression pattern of two docetaxel resistant MCF-7 breast carcinoma sublines step wisely selected in 30 and 120 nM docetaxel. Cell proliferation assay was performed in order to demonstrate development of docetaxel resistance. cDNA microarray analysis was performed using Affymetrix(®) Human Genome U133 Plus 2.0 Arrays in duplicate experiments. Quantitative and semi-quantitative gene expression analysis was also performed to confirm gene expression analysis for selected genes. XTT results demonstrated that 30 (MCF-7/30nM DOC) and 120 nM (MCF-7/120nM DOC) docetaxel selected cells were 13- and 47-fold resistant, respectively. cDNA microarray analysis demonstrated that expression profiles of MCF-7 and MCF-7/30nM DOC were more similar to each other where expression profile of MCF-7/120nM DOC was different as examined by line graphs and scatter plots. 2,837 and 4,036 genes were significantly altered in 30 and 120 nM docetaxel resistant sublines, respectively. Among these, 849 genes were altered in common in two docetaxel resistant sublines. Antiapoptotic gene expression (e.g., Bcl-2 and APRIL) were noticeably altered in MCF-7/30nM DOC. However, docetaxel resistance in MCF-7/120nM DOC were more complicated with the involvement of ECM related gene expression, cytokine and growth factor signaling, ROS metabolism and EMT related gene expression together with higher level of MDR1 expression. Expression profiles in 30 and 120 nM docetaxel resistant sublines changed gradually with increasing resistance index. Drug resistance development seems to be step wise event in MCF-7 cells.
Collapse
|
37
|
Röder C, Trauzold A, Kalthoff H. Impact of death receptor signaling on the malignancy of pancreatic ductal adenocarcinoma. Eur J Cell Biol 2011; 90:450-5. [DOI: 10.1016/j.ejcb.2010.10.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 10/13/2010] [Indexed: 12/22/2022] Open
|
38
|
Increased expression of TRAIL and its death receptors DR4 and DR5 in plaque psoriasis. Arch Dermatol Res 2011; 303:389-97. [DOI: 10.1007/s00403-011-1125-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 01/09/2011] [Accepted: 01/14/2011] [Indexed: 12/18/2022]
|
39
|
Wang S, Ren W, Liu J, Lahat G, Torres K, Lopez G, Lazar AJ, Hayes-Jordan A, Liu K, Bankson J, Hazle JD, Lev D. TRAIL and doxorubicin combination induces proapoptotic and antiangiogenic effects in soft tissue sarcoma in vivo. Clin Cancer Res 2010; 16:2591-604. [PMID: 20406839 PMCID: PMC2967255 DOI: 10.1158/1078-0432.ccr-09-2443] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Novel therapeutic approaches for complex karyotype soft tissue sarcoma (STS) are crucially needed. Consequently, we assessed the efficacy of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), in combination with chemotherapy, on local and metastatic growth of human STS xenografts in vivo. EXPERIMENTAL DESIGN TRAIL was evaluated alone and combined with low-dose doxorubicin in two human STS severe combined immunodeficient mouse xenograft models using fibrosarcoma (HT1080; wild-type p53) and leiomyosarcoma (SKLMS1; mutated p53), testing for effects on local growth, metastasis, and overall survival. Magnetic resonance imaging was used to evaluate local growth and bioluminescence was used to longitudinally assess lung metastases. Tissues were evaluated through immunohistocemistry and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining for treatment effects on tumor cell proliferation, apoptosis, angiogenesis, angiogenic factors, and TRAIL receptor expression. Quantitative real-time polymerase chain reaction (QRTPCR) angiogenesis array was used to assess therapy-induced gene expression changes. RESULTS TRAIL/doxorubicin combination induced marked STS local and metastatic growth inhibition in a p53-independent manner. Significantly increased (P < 0.001) host survival was also demonstrable. Combined therapy induced significant apoptosis, decreased tumor cell proliferation, and increased TRAIL receptor (DR4 and DR5) expression in all treated tumors. Moreover, decreased microvessel density was observed, possibly secondary to increased expression of the antiangiogenic factor CXCL10 and decreased proangiogenic interleukin-8 cytokine in response to TRAIL/doxorubicin combination, as was also observed in vitro. CONCLUSIONS Given the urgent need for better systemic approaches to STS, clinical trials evaluating TRAIL in combination with low-dose chemotherapy are potentially warranted.
Collapse
Affiliation(s)
- Suizhao Wang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wenhong Ren
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffery Liu
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guy Lahat
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keila Torres
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gonzalo Lopez
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexander J Lazar
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andrea Hayes-Jordan
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA
| | - Jim Bankson
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - John D. Hazle
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dina Lev
- The Sarcoma Research Center at the University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
40
|
Chan J, Prado-Lourenco L, Khachigian LM, Bennett MR, Di Bartolo BA, Kavurma MM. TRAIL promotes VSMC proliferation and neointima formation in a FGF-2-, Sp1 phosphorylation-, and NFkappaB-dependent manner. Circ Res 2010; 106:1061-71. [PMID: 20150555 DOI: 10.1161/circresaha.109.206029] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) is well reported as an inducer of apoptosis in tumor models; however, its role and function in vivo in atherosclerosis and vascular injury has not been established. OBJECTIVE We sought to study the function of TRAIL in cardiovascular pathology and its regulation in vivo. METHODS AND RESULTS Here, we show that TRAIL was upregulated in medial vascular smooth muscle cells (VSMCs) 24 hours following perivascular cuff placement around femoral arteries of mice. We also show that TRAIL mRNA and promoter activity was induced in VSMCs following in vitro mechanical injury. Intimal thickening 15 days after cuff placement was reduced 2- to 3-fold in TRAIL(-/-) compared to wild-type mice and was reversible by administration of recombinant TRAIL. Additionally, reduced VSMC proliferation was observed in injured arteries of TRAIL(-/-) mice. Fibroblast growth factor (FGF)-2, a potent growth factor released following vascular injury, was also reduced in arteries of TRAIL(-/-) mice, and VSMCs isolated from these animals did not respond to FGF-2 in vitro. Injury and FGF-2 regulated TRAIL transcriptional activity via 2 specificity protein (Sp)1 elements in the proximal TRAIL promoter, a binding site also shared by nuclear factor (NF)kappaB. Mutational studies confirmed a role for Sp1 in injury- and FGF-2-inducible TRAIL transcription. Furthermore, increased NFkappaB expression after injury transactivated the TRAIL promoter. Interestingly, following mechanical injury, Sp1 phosphorylation (Thr453) and an increase in the physical interaction of p-Sp1(Thr453) with NFkappaB was observed. CONCLUSIONS We conclude that TRAIL induction involves FGF-2, Sp1-phosphorylation and NFkappaB and that TRAIL promotes VSMC proliferation and neointima formation after arterial injury.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cell Proliferation
- Cells, Cultured
- Femoral Artery/injuries
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Fibroblast Growth Factor 2/metabolism
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Molecular Sequence Data
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/metabolism
- Phosphorylation
- Promoter Regions, Genetic
- RNA, Messenger/metabolism
- Rats
- Recombinant Proteins/metabolism
- Signal Transduction
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- TNF-Related Apoptosis-Inducing Ligand/deficiency
- TNF-Related Apoptosis-Inducing Ligand/genetics
- TNF-Related Apoptosis-Inducing Ligand/metabolism
- Transcriptional Activation
- Transfection
- Tunica Intima/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Jeffrey Chan
- Centre for Vascular Research, The University of New South Wales, Sydney NSW 2052, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Loreto C, Almeida LE, Migliore MR, Caltabiano M, Leonardi R. TRAIL, DR5 and caspase 3-dependent apoptosis in vessels of diseased human temporomandibular joint disc. An immunohistochemical study. Eur J Histochem 2010; 54:e40. [PMID: 20839416 PMCID: PMC3167309 DOI: 10.4081/ejh.2010.e40] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 08/05/2010] [Indexed: 01/16/2023] Open
Abstract
To evaluate the apoptosis involvement in the angiogenesis as a self-limiting process in patients with temporomandibular joint (TMJ) degenerated disc vessels, we assessed, by immunohistochemistry, the detection of TRAIL, its death receptor DR5 and caspase 3. TRAIL, its death receptor DR5 and caspase 3 expression were studied by immunohistochemistry in 15 TMJ discs displaced without reduction and in 4 unaffected discs. These apoptosis molecules were detected in the intima and media layers of newly formed vessels affected discs. In conclusion, vessels apoptosis activation in TMJ disc with ID could be regarded as a self-limiting process that try to leads to vessel regression; in this way an inhibition of angiogenic vessels may prove a key strategy in limiting pathological angiogenesis, by cutting off blood supply to tumors, or by reducing harmful inflammation.
Collapse
Affiliation(s)
- C Loreto
- Department of Anatomy, Diagnostic Pathology, Forensic Medicine, Hygiene and Public Health, University of Catania, via S. Sofia 87, Catania, Italy.
| | | | | | | | | |
Collapse
|
42
|
Evidence that tumor necrosis factor-related apoptosis inducing ligand (TRAIL) inhibits angiogenesis by inducing vascular endothelial cell apoptosis. Biochem Biophys Res Commun 2009; 391:936-41. [PMID: 19962958 DOI: 10.1016/j.bbrc.2009.11.168] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 11/26/2009] [Indexed: 01/18/2023]
Abstract
Tumor necrosis factor (TNF) and its related ligands TNF-related apoptosis inducing ligand (TRAIL) and Fas ligand (FasL) play roles in the regulation of vascular responses, but their effect on the formation of new blood vessels (angiogenesis) is unclear. Therefore, we have examined the effects of these ligands on angiogenesis modeled with primary cultures of human umbilical vein endothelial cells (HUVEC). To examine angiogenesis in the context of the central nervous system, we have also modeled cerebral angiogenesis with the human brain endothelial cell line hCMEC/D3. Parameters studied were bromodeoxyuridine (BrdU) incorporation and cell number (MTT) assay (to assess endothelial proliferation), scratch assay (migration) and networks on Matrigel (tube formation). In our hands, neither TRAIL nor FasL (1, 10, and 100 ng/ml) had an effect on parameters of angiogenesis in the HUVEC model. In hCMEC/D3 cells by contrast, TRAIL inhibited all parameters (10-100 ng/ml, 24h). This was due to apoptosis, since its action was blocked by the pan-caspase inhibitor zVADfmk (5 x 10(-5) mol/l) and TRAIL increased caspase-3 activity 1h after application. However FasL (100 ng/ml) increased BrdU uptake without other effects. We conclude that TRAIL has different effects on in vitro angiogenesis depending on which model is used, but that FasL is generally ineffective when applied in vitro. The data suggest that TRAIL primarily influences angiogenesis by the induction of vascular endothelial apoptosis, leading to vessel regression.
Collapse
|
43
|
Simoncini S, Njock MS, Robert S, Camoin-Jau L, Sampol J, Harlé JR, Nguyen C, Dignat-George F, Anfosso F. TRAIL/Apo2L Mediates the Release of Procoagulant Endothelial Microparticles Induced by Thrombin In Vitro. Circ Res 2009; 104:943-51. [DOI: 10.1161/circresaha.108.183285] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Microparticles are small vesicles playing a crucial role in cell communication by promoting prothrombotic and proinflammatory responses. However, the molecular mechanisms underlying their release are still elusive. We previously established that thrombin promoted the generation of endothelial microparticles (EMPs). In the present study, gene profiling identified
TRAIL/Apo2L
, a cytokine belonging to the tumor necrosis factor-α superfamily, as a target of thrombin. Thrombin increased the expression of cell-associated and soluble forms of TRAIL (sTRAIL) in HMEC-1 cells and human umbilical vein endothelial cells (HUVECs). Blocking TRAIL by specific antibodies or by small interfering RNA reduced both the number and the procoagulant activity of EMPs released by thrombin. Consistent with an involvement of sTRAIL in thrombin-induced EMP release, we showed that (1) exogenously added sTRAIL generated procoagulant EMPs; (2) supernatants from thrombin-stimulated endothelial cells induced EMP release by HMEC-1 cells and HUVECs, whereas those recovered from TRAIL knockdown endothelial cells displayed no effect. TRAIL/TRAIL-R2 complex mediated EMP release by initiating the recruitment of adaptor proteins and the activation of nuclear factor κB. Moreover, sTRAIL modulated intercellular adhesion molecule-1 and interleukin-8 expression induced by thrombin by a downstream pathway involving nuclear factor κB activation. Our data reveal a novel mechanism controlling EMP release and identify TRAIL as a key partner in the pathway linking coagulation and inflammation elicited by thrombin.
Collapse
Affiliation(s)
- Stéphanie Simoncini
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Makon-Sébastien Njock
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Stéphane Robert
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Laurence Camoin-Jau
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - José Sampol
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Jean-Robert Harlé
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Catherine Nguyen
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Françoise Dignat-George
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| | - Francine Anfosso
- From the Institut National de la Santé et de la Recherche Médicale UMR608 (S.S., M.-S.N., S.R., L.C.-J., J.S., F.D.-G., F.A.), Physiopathologie de l'Endothelium, Universite Mediterranee, Faculte de Pharmacie Marseille, France; Hopital de la Conception (L.C.-J., J.-R.H., F.D.-G.), Assistance Publique-Hopitaux de Marseille, France; and Institut National de la Santé et de la Recherche Médicale U928 (C.N.), Technologies Avancées pour le Génome et la Clinique, Marseille, France
| |
Collapse
|
44
|
Abstract
Death receptors are members of the tumor necrosis factor receptor superfamily characterized by a cytoplasmic region known as the "death domain" that enables the receptors to initiate cytotoxic signals when engaged by cognate ligands. Binding to the ligand results in receptor aggregation and recruitment of adaptor proteins, which, in turn, initiates a proteolytic cascade by recruiting and activating initiator caspases 8 and 10. Death receptors were once thought to primarily induce cytotoxic signaling cascades. However, recent data indicate that they initiate multiple signaling pathways, unveiling a number of nonapoptosis-related functions, including regulation of cell proliferation and differentiation, chemokine production, inflammatory responses, and tumor-promoting activities. These noncytotoxic cascades are not simply a manifestation of inhibiting proapoptotic pathways but are intrinsically regulated by adaptor protein and receptor internalization processes. Insights into these various death receptor signaling pathways provide new therapeutic strategies targeting these receptors in pathophysiological processes.
Collapse
Affiliation(s)
- Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN 55905, USA
| | | |
Collapse
|
45
|
Osteoprotegerin and RANKL differentially regulate angiogenesis and endothelial cell function. Angiogenesis 2008; 12:35-46. [DOI: 10.1007/s10456-008-9127-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Accepted: 12/04/2008] [Indexed: 10/21/2022]
|
46
|
Kavurma MM, Tan NY, Bennett MR. Death receptors and their ligands in atherosclerosis. Arterioscler Thromb Vasc Biol 2008; 28:1694-702. [PMID: 18669890 DOI: 10.1161/atvbaha.107.155143] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is characterized by the accumulation of a fibro-fatty plaque consisting of immune cells, vascular smooth muscle cells (VSMCs), vascular endothelial cells (ECs), and extracellular matrix, surrounding a lipid-rich core. The complexity of atherosclerosis is highlighted by the multifaceted effects that apoptosis and proliferation of specific cell types can have on vessels at different stages of the disease. Death receptors are membrane-bound protein complexes that on binding their cognate ligand, activate an intracellular signaling cascade that results in apoptosis. More recently, signaling from these receptors has been shown to activate multiple other processes, including cell proliferation. This review summarizes our current understanding of signaling events after death receptor activation and the role of death receptors and their ligands in atherosclerosis.
Collapse
Affiliation(s)
- Mary M Kavurma
- Centre for Vascular Research, The University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | |
Collapse
|
47
|
Levina V, Marrangoni AM, DeMarco R, Gorelik E, Lokshin AE. Multiple effects of TRAIL in human carcinoma cells: induction of apoptosis, senescence, proliferation, and cytokine production. Exp Cell Res 2008; 314:1605-16. [PMID: 18313665 PMCID: PMC2386090 DOI: 10.1016/j.yexcr.2007.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 12/23/2007] [Accepted: 12/26/2007] [Indexed: 01/08/2023]
Abstract
TRAIL is a death ligand that induces apoptosis in malignant but not normal cells. Recently the ability of TRAIL to induce proliferation in apoptosis-resistant normal and malignant cells was reported. In this study, we analyzed TRAIL effects in apoptosis sensitive MCF7, OVCAR3 and H460 human tumor cell lines. TRAIL at low concentrations preferentially induced cell proliferation. At 100 ng/ml, apoptotic death was readily observed, however surviving cells acquired higher proliferative capacity. TRAIL-stimulated production of several cytokines, IL-8, RANTES, MCP-1 and bFGF, and activation of caspases 1 and 8 was essential for this effect. Antibodies to IL-8, RANTES, and bFGF blocked TRAIL-induced cell proliferation and further stimulated apoptosis. For the first time, we report that high TRAIL concentrations induced cell senescence as determined by the altered morphology and expression of several senescence markers: SA-beta-gal, p21Waf1/Cip1, p16INK4a, and HMGA. Caspase 9 inhibition protected TRAIL-treated cells from senescence, whereas inhibition of caspases 1 and 8 increased the yield of SLP cells. In conclusion, in cultured human carcinoma cells, TRAIL therapy results in three functional outcomes, apoptosis, proliferation and senescence. TRAIL-induced proapoptotic and prosurvival responses correlate with the strength of signaling. TRAIL-induced cytokine production is responsible for its proliferative and prosurvival effects.
Collapse
Affiliation(s)
- Vera Levina
- Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Adele M. Marrangoni
- Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Richard DeMarco
- Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Elieser Gorelik
- Department of Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Immunology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Anna E. Lokshin
- Department of Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
48
|
Kavurma MM, Schoppet M, Bobryshev YV, Khachigian LM, Bennett MR. TRAIL stimulates proliferation of vascular smooth muscle cells via activation of NF-kappaB and induction of insulin-like growth factor-1 receptor. J Biol Chem 2008; 283:7754-62. [PMID: 18178561 DOI: 10.1074/jbc.m706927200] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TRAIL/Apo2L (tumor necrosis factor-related apoptosis-inducing ligand) is a multifunctional protein regulating homeostasis of the immune system, infection, autoimmune diseases, and apoptosis. However, its function in normal, nontransformed tissues is not clear. Here we show that TRAIL increases vascular smooth muscle cell (VSMC) proliferation in vitro, effects that can be blocked with neutralizing antibodies to TRAIL receptors DR4 and DcR1. In aortocoronary saphenous vein bypass grafts in vivo, TRAIL co-localizes with VSMC, proliferating cell nuclear antigen, and insulin-like growth factor type 1 receptor (IGF1R) expression but not active caspase-3. TRAIL is required for serum-inducible IGF1R expression, and antisense IGF1R inhibits TRAIL-induced VSMC proliferation. At 1 ng/ml, TRAIL stimulates IGF1R mRNA expression greater than insulin-like growth factor-1 and also activates the IGF1R promoter 7-fold. TRAIL-inducible IGF1R expression requires NF-kappaB activation. Consistent with this, ammonium pyrrolidine dithiocarbamate, a pharmacological inhibitor of NF-kappaB, blocks TRAIL-induced IGF1R expression, and p65 overexpression increases IGF1R protein levels. In addition, NF-kappaB binds a novel TRAIL-responsive element on the IGF1R promoter. Our findings suggest that the biological functions of TRAIL in VSMC extend beyond its role in promoting apoptosis. Thus, TRAIL may play an important role in atherosclerosis by regulating IGF1R expression in VSMC in an NF-kappaB-dependent manner.
Collapse
Affiliation(s)
- Mary M Kavurma
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | | | | | | | | |
Collapse
|
49
|
Kavurma MM, Bennett MR. Expression, regulation and function of trail in atherosclerosis. Biochem Pharmacol 2007; 75:1441-50. [PMID: 18061141 DOI: 10.1016/j.bcp.2007.10.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 10/03/2007] [Accepted: 10/11/2007] [Indexed: 01/22/2023]
Abstract
Atherosclerosis is a condition where vascular smooth muscle cells (VSMCs), inflammatory cells, lipids, cholesterol and cellular waste accumulate in the inner lining of an artery, producing a fibro-fatty plaque and resulting in the thickening of the arterial wall. The tumor necrosis factor (TNF) family of cytokines plays a major role in the progression of atherosclerosis. Recently, TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, has been implicated in the development of atherosclerosis since it has been detected in normal and diseased atherosclerotic tissue. Not only is TRAIL involved in apoptosis and immune regulation, recent studies have provided a new function of TRAIL on vascular cells, such that TRAIL can promote endothelial cell (EC) and VSMCs migration and proliferation. In addition, TRAIL is implicated in regulating vascular tone. This review discusses our current understanding of TRAIL expression, regulation and function, and summarises the recent data implicating a role for TRAIL in atherosclerosis.
Collapse
Affiliation(s)
- Mary M Kavurma
- Centre for Vascular Research, The University of New South Wales, Kensington, Sydney, NSW 2052, Australia.
| | | |
Collapse
|
50
|
|