1
|
Yao T, Rudak PT, Laumont CM, Michaud AR, Rashu R, Knier NN, Foster PJ, McWilliam HEG, Villadangos JA, Nelson BH, DiMattia GE, Shepherd TG, Haeryfar SMM. MAIT cells accumulate in ovarian cancer-elicited ascites where they retain their capacity to respond to MR1 ligands and cytokine cues. Cancer Immunol Immunother 2021; 71:1259-1273. [PMID: 34854949 DOI: 10.1007/s00262-021-03118-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022]
Abstract
The low mutational burden of epithelial ovarian cancer (EOC) is an impediment to immunotherapies that rely on conventional MHC-restricted, neoantigen-reactive T lymphocytes. Mucosa-associated invariant T (MAIT) cells are MR1-restricted T cells with remarkable immunomodulatory properties. We sought to characterize intratumoral and ascitic MAIT cells in EOC. Single-cell RNA sequencing of six primary human tumor specimens demonstrated that MAIT cells were present at low frequencies within several tumors. When detectable, these cells highly expressed CD69 and VSIR, but otherwise exhibited a transcriptomic signature inconsistent with overt cellular activation and/or exhaustion. Unlike mainstream CD8+ T cells, CD8+ MAIT cells harbored high transcript levels of TNF, PRF1, GZMM and GNLY, suggesting their arming and cytotoxic potentials. In a congenic, MAIT cell-sufficient mouse model of EOC, MAIT and invariant natural killer T cells amassed in the peritoneal cavity where they showed robust IL-17A and IFN-γ production capacities, respectively. However, they gradually lost these functions with tumor progression. In a cohort of 23 EOC patients, MAIT cells were readily detectable in all ascitic fluids examined. In a sub-cohort in which we interrogated ascitic MAIT cells for functional impairments, several exhaustion markers, most notably VISTA, were present on the surface. However, ascitic MAIT cells were capable of producing IFN-γ, TNF-α and granzyme B, but neither IL-17A nor IL-10, in response to an MR1 ligand, bacterial lysates containing MR1 ligands, or a combination of IL-12 and IL-18. In conclusion, ascitic MAIT cells in EOC possess inducible effector functions that may be modified in future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tony Yao
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Patrick T Rudak
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Céline M Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alex R Michaud
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Rasheduzzaman Rashu
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada
| | - Natasha N Knier
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON, Canada.,Robarts Research Institute, Western University, London, ON, Canada
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Gabriel E DiMattia
- Department of Biochemistry, Western University, London, ON, Canada.,The Mary & John Knight Translational Ovarian Cancer Research Unit, London, ON, Canada.,Department of Oncology, Western University, London, ON, Canada.,Department of Obstetrics and Gynaecology, Western University, London, ON, Canada.,London Regional Cancer Program, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London, ON, Canada.,Department of Oncology, Western University, London, ON, Canada.,Department of Obstetrics and Gynaecology, Western University, London, ON, Canada.,London Regional Cancer Program, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, 1151 Richmond Street, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada. .,Centre for Human Immunology, Western University, London, ON, Canada. .,Department of Surgery, Division of General Surgery, Western University, London, ON, Canada. .,Department of Medicine, Division of Clinical Immunology and Allergy, Western University, London, ON, Canada.
| |
Collapse
|
2
|
Adams SF, Grimm AJ, Chiang CLL, Mookerjee A, Flies D, Jean S, McCann GA, Michaux J, Pak H, Huber F, Neal C, Dangaj D, Bassani-Sternberg M, Rusakiewicz S, Facciabene A, Coukos G, Gimotty PA, Kandalaft LE. Rapid tumor vaccine using Toll-like receptor-activated ovarian cancer ascites monocytes. J Immunother Cancer 2020; 8:jitc-2020-000875. [PMID: 32817208 PMCID: PMC7430560 DOI: 10.1136/jitc-2020-000875] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Background Novel therapeutic strategies in ovarian cancer (OC) are needed as the survival rate remains dismally low. Although dendritic cell-based cancer vaccines are effective in eliciting therapeutic responses, their complex and costly manufacturing process hampers their full clinical utility outside specialized clinics. Here, we describe a novel approach of generating a rapid and effective cancer vaccine using ascites-derived monocytes for treating OC. Methods Using the ID8 mouse ovarian tumor model and OC patient samples, we isolated ascites monocytes and evaluated them with flow cytometry, Luminex cytokine and chemokine array analysis, ex vivo cocultures with T cells, in vivo tumor challenge and T cell transfer experiments, RNA-sequencing and mass spectrometry. Results We demonstrated the feasibility of isolating ascites monocytes and restoring their ability to function as bona fide antigen-presenting cells (APCs) with Toll-like receptor (TLR) 4 lipopolysaccharide and TLR9 CpG-oligonucleotides, and a blocking antibody to interleukin-10 receptor (IL-10R Ab) in the ID8 model. The ascites monocytes were laden with tumor antigens at a steady state in vivo. After a short 48 hours activation, they upregulated maturation markers (CD80, CD86 and MHC class I) and demonstrated strong ex vivo T cell stimulatory potential and effectively suppressed tumor and malignant ascites in vivo. They also induced protective long-term T cell memory responses. To evaluate the translational potential of this approach, we isolated ascites monocytes from stage III/IV chemotherapy-naïve OC patients. Similarly, the human ascites monocytes presented tumor-associated antigens (TAAs), including MUC1, ERBB2, mesothelin, MAGE, PRAME, GPC3, PMEL and TP53 at a steady state. After a 48-hour treatment with TLR4 and IL-10R Ab, they efficiently stimulated oligoclonal tumor-associated lymphocytes (TALs) with strong reactivity against TAAs. Importantly, the activated ascites monocytes retained their ability to activate TALs in the presence of ascitic fluid. Conclusions Ascites monocytes are naturally loaded with tumor antigen and can perform as potent APCs following short ex vivo activation. This novel ascites APC vaccine can be rapidly prepared in 48 hours with a straightforward and affordable manufacturing process, and would be an attractive therapeutic vaccine for OC.
Collapse
Affiliation(s)
- Sarah F Adams
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Alizée J Grimm
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Cheryl L-L Chiang
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Ananda Mookerjee
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dallas Flies
- Division of Gynecologic Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico, USA
| | - Stephanie Jean
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Georgia A McCann
- Division of Gynecologic Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Justine Michaux
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - HuiSong Pak
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Florian Huber
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Christopher Neal
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Denarda Dangaj
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Sylvie Rusakiewicz
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Andrea Facciabene
- Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lana E Kandalaft
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland .,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| |
Collapse
|
3
|
Birmpilis AI, Karachaliou CE, Samara P, Ioannou K, Selemenakis P, Kostopoulos IV, Kavrochorianou N, Kalbacher H, Livaniou E, Haralambous S, Kotsinas A, Farzaneh F, Trougakos IP, Voelter W, Dimopoulos MA, Bamias A, Tsitsilonis O. Antitumor Reactive T-Cell Responses Are Enhanced In Vivo by DAMP Prothymosin Alpha and Its C-Terminal Decapeptide. Cancers (Basel) 2019; 11:cancers11111764. [PMID: 31717548 PMCID: PMC6896021 DOI: 10.3390/cancers11111764] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Prothymosin α (proTα) and its C-terminal decapeptide proTα(100-109) were shown to pleiotropically enhance innate and adaptive immune responses. Their activities have been broadly studied in vitro, focusing primarily on the restoration of the deficient immunoreactivity of cancer patients' leukocytes. Previously, we showed that proTα and proTα(100-109) act as danger-associated molecular patterns (DAMPs), ligate Toll-like receptor-4, signal through TRIF- and MyD88-dependent pathways, promote the maturation of dendritic cells and elicit T-helper type 1 (Th1) immune responses in vitro, leading to the optimal priming of tumor antigen-reactive T-cell functions. Herein, we assessed their activity in a preclinical melanoma model. Immunocompetent mice bearing B16.F1 tumors were treated with two cycles of proTα or proTα(100-109) together with a B16.F1-derived peptide vaccine. Coadministration of proTα or proTα(100-109) and the peptide vaccine suppressed melanoma-cell proliferation, as evidenced by reduced tumor-growth rates. Higher melanoma infiltration by CD3+ T cells was observed, whereas ex vivo analysis of mouse total spleen cells verified the in vivo induction of melanoma-reactive cytotoxic responses. Additionally, increased levels of proinflammatory and Th1-type cytokines were detected in mouse serum. We propose that, in the presence of tumor antigens, DAMPs proTα and proTα(100-109) induce Th1-biased immune responses in vivo. Their adjuvant ability to orchestrate antitumor immunoreactivities can eventually be exploited therapeutically in humans.
Collapse
Affiliation(s)
- Anastasios I. Birmpilis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Pinelopi Samara
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Kyriaki Ioannou
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Platon Selemenakis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Ioannis V. Kostopoulos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Nadia Kavrochorianou
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, NCSR “Demokritos”, Agia Paraskevi, 15310 Athens, Greece; (C.-E.K.); (E.L.)
| | - Sylva Haralambous
- Inflammation Research Group, Transgenic Technology Laboratory, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.K.); (S.H.)
| | - Athanasios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, 11527 Athens, Greece; (P.S.); (A.K.)
| | - Farzin Farzaneh
- King’s College London, Rayne Institute, 123 Coldharbour Lane, SE5 9NU London, UK;
| | - Ioannis P. Trougakos
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen. Germany; (H.K.); (W.V.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.-A.D.); (A.B.)
| | - Ourania Tsitsilonis
- Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (A.I.B.); (P.S.); (K.I.); (I.V.K.); (I.P.T.)
- Correspondence: ; Tel.: +30-210-727-4215; Fax: +30-210-727-4635
| |
Collapse
|
4
|
Sandow JJ, Rainczuk A, Infusini G, Makanji M, Bilandzic M, Wilson AL, Fairweather N, Stanton PG, Garama D, Gough D, Jobling TW, Webb AI, Stephens AN. Discovery and Validation of Novel Protein Biomarkers in Ovarian Cancer Patient Urine. Proteomics Clin Appl 2018; 12:e1700135. [DOI: 10.1002/prca.201700135] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/16/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Jarrod J. Sandow
- Walter and Eliza Hall Institute, Department of Medical Biology; University of Melbourne; Parkville VIC Australia
| | - Adam Rainczuk
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | - Giuseppe Infusini
- Walter and Eliza Hall Institute, Department of Medical Biology; University of Melbourne; Parkville VIC Australia
| | - Ming Makanji
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | - Maree Bilandzic
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | - Amy L. Wilson
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | | | - Peter G. Stanton
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
| | - Daniel Garama
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | - Daniel Gough
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
| | - Thomas W. Jobling
- Obstetrics and Gynaecology; Monash Medical Centre; Clayton VIC Australia
| | - Andrew I. Webb
- Walter and Eliza Hall Institute, Department of Medical Biology; University of Melbourne; Parkville VIC Australia
| | - Andrew N. Stephens
- Department of Molecular and Translational Sciences; Monash University; VIC Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; VIC Australia
- Epworth Research Institute; Epworth HealthCare; Richmond VIC Australia
| |
Collapse
|
5
|
Karachaliou CE, Triantis C, Liolios C, Palamaris L, Zikos C, Tsitsilonis OE, Kalbacher H, Voelter W, Loudos G, Papadopoulos M, Pirmettis I, Livaniou E. In vivo biodistribution and imaging studies with a 99mTc-radiolabeled derivative of the C-terminus of prothymosin alpha in mice bearing experimentally-induced inflammation. Eur J Pharm Biopharm 2017; 113:188-197. [PMID: 28087377 DOI: 10.1016/j.ejpb.2016.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/15/2016] [Accepted: 12/14/2016] [Indexed: 10/20/2022]
Abstract
Prothymosin alpha (ProTα) is a highly conserved mammalian polypeptide (109 amino acids in man) exerting in vitro and in vivo immunoenhancing activities. Recently, our team has developed a 99mTc-radiolabeled derivative of the C-terminal bioactive decapeptide of ProTα ([99mTc]C1) and employed it in in vitro studies, the results of which support the existence of binding sites on human neutrophils that recognize [99mTc]C1, intact ProTα as well as the C-terminal decapeptide of ProTα and presumably involve Toll-like receptor 4. In the present work, [99mTc]C1 was administered to Swiss albino mice with experimentally-induced inflammation for in vivo biodistribution and imaging studies, in parallel with a suitable negative control, which differs from [99mTc]C1 only in bearing a scrambled version of the ProTα decapeptide. The biodistribution data obtained with [99mTc]C1 demonstrated fast clearance of radioactivity from blood, heart, lungs, normal muscle, and predominantly urinary excretion. Most importantly, slow clearance of radioactivity from the inflammation focus was observed, resulting in a high ratio of inflamed/normal muscle tissue (9.15 at 30min post injection, which remained practically stable up to 2h). The inflammation-targeting capacity of [99mTc]C1 was confirmed by imaging studies and might be attributed to neutrophils, which are recruited at the inflamed areas and bear binding sites for [99mTc]C1. In this respect, apart from being a valuable tool for further studies on ProTα in in vitro and in vivo systems, [99mTc]C1 merits further evaluation as a radiopharmaceutical for specific imaging of inflammation foci.
Collapse
Affiliation(s)
- Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Charalampos Triantis
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Christos Liolios
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Lazaros Palamaris
- Department of Medical Instruments Technology, Technological Educational Institute, Athens 12243, Greece
| | - Christos Zikos
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Ourania E Tsitsilonis
- Division of Animal and Human Physiology, Department of Biology, University of Athens, Athens 15784, Greece
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - George Loudos
- Department of Medical Instruments Technology, Technological Educational Institute, Athens 12243, Greece
| | - Minas Papadopoulos
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety (INRASTES), National Center for Scientific Research "Demokritos" (NCSR "Demokritos"), Athens 15310, Greece.
| |
Collapse
|
6
|
Ueda H, Halder SK, Matsunaga H, Sasaki K, Maeda S. Neuroprotective impact of prothymosin alpha-derived hexapeptide against retinal ischemia-reperfusion. Neuroscience 2016; 318:206-18. [PMID: 26779836 DOI: 10.1016/j.neuroscience.2016.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/17/2015] [Accepted: 01/05/2016] [Indexed: 01/13/2023]
Abstract
Prothymosin alpha (ProTα) has robustness roles against brain and retinal ischemia or serum-starvation stress. In the ProTα sequence, the active core 30-amino acid peptide/P30 (a.a.49-78) is necessary for the original neuroprotective actions against ischemia. Moreover, the 9-amino acid peptide sequence/P9 (a.a.52-60) in P30 still shows neuroprotective activity against brain and retinal ischemia, though P9 is less potent than P30. As the previous structure-activity relationship study for ProTα may not be enough, the possibility still exists that any sequence smaller than P9 retains potent neuroprotective activity. When different P9- and P30-related peptides were intravitreally injected 24h after retinal ischemia in mice, the 6-amino acid peptide/P6 (NEVDEE, a.a.51-56) showed potent protective effects against ischemia-induced retinal functional deficits, which are equipotent to the level of P30 peptide in electroretinography (ERG) and histological damage in Hematoxylin and Eosin (HE) staining. Further studies using ERG and HE staining suggested that intravitreal or intravenous (i.v.) injection with modified P6 peptide/P6Q (NEVDQE) potently inhibited retinal ischemia-induced functional and histological damage. In an immunohistochemical analysis, the ischemia-induced loss of retinal ganglion, bipolar, amacrine and photoreceptor cells were inhibited by a systemic administration with P6Q peptide 24h after the ischemic stress. In addition, systemic post-treatment with P6Q peptide significantly inhibited retinal ischemia-induced microglia and astrocyte activation in terms of increased ionized calcium-binding adaptor molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP) intensity, respectively, as well as their morphological changes, increased number and migration. Thus, this study demonstrates the therapeutic significance of modified P6 peptide P6Q (NEVDQE) derived from 6-amino acid peptide (P6) in ProTα against ischemic damage.
Collapse
Affiliation(s)
- H Ueda
- Department of Pharmacology and Therapeutic Innovation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8521, Japan.
| | - S K Halder
- Department of Pharmacology and Therapeutic Innovation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - H Matsunaga
- Department of Pharmacology and Therapeutic Innovation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - K Sasaki
- Department of Pharmacology and Therapeutic Innovation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - S Maeda
- Department of Pharmacology and Therapeutic Innovation, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| |
Collapse
|
7
|
Prothymosin Alpha and Immune Responses: Are We Close to Potential Clinical Applications? VITAMINS AND HORMONES 2016; 102:179-207. [PMID: 27450735 PMCID: PMC7126549 DOI: 10.1016/bs.vh.2016.04.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The thymus gland produces soluble molecules, which mediate significant immune functions. The first biologically active thymic extract was thymosin fraction V, the fractionation of which led to the isolation of a series of immunoactive polypeptides, including prothymosin alpha (proTα). ProTα displays a dual role, intracellularly as a survival and proliferation mediator and extracellularly as a biological response modifier. Accordingly, inside the cell, proTα is implicated in crucial intracellular circuits and may serve as a surrogate tumor biomarker, but when found outside the cell, it could be used as a therapeutic agent for treating immune system deficiencies. In fact, proTα possesses pleiotropic adjuvant activity and a series of immunomodulatory effects (eg, anticancer, antiviral, neuroprotective, cardioprotective). Moreover, several reports suggest that the variable activity of proTα might be exerted through different parts of the molecule. We first reported that the main immunoactive region of proTα is the carboxy-terminal decapeptide proTα(100-109). In conjunction with data from others, we also revealed that proTα and proTα(100-109) signal through Toll-like receptor 4. Although their precise molecular mechanism of action is yet not fully elucidated, proTα and proTα(100-109) are viewed as candidate adjuvants for cancer immunotherapy. Here, we present a historical overview on the discovery and isolation of thymosins with emphasis on proTα and data on some immune-related new activities of the polypeptide and smaller immunostimulatory peptides thereof. Finally, we propose a compiled scenario on proTα's mode of action, which could eventually contribute to its clinical application.
Collapse
|
8
|
Zhang BC, Sun L. Tongue sole (Cynoglossus semilaevis) prothymosin alpha: Cytokine-like activities associated with the intact protein and the C-terminal region that lead to antiviral immunity via Myd88-dependent and -independent pathways respectively. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 53:96-104. [PMID: 26162512 DOI: 10.1016/j.dci.2015.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 06/04/2023]
Abstract
Prothymosin alpha (ProTα) is a small protein that in mammals is known to participate in diverse biological processes including immunomodulation. In teleost, the immunological function of ProTα is unknown. In the current study, we investigated the expression and function of the ProTα (named CsProTα) from the teleost fish tongue sole (Cynoglossus semilaevis). We found that CsProTα expression was abundant in immune relevant tissues and upregulated by megalocytivirus infection. Immunoblot detected secretion of CsProTα by peripheral blood leukocytes. Recombinant CsProTα (rCsProTα) as well as the C-terminal 11-residue (Ct11) were able to bind head kidney monocytes (HKM) and induce immune gene expression; however, the induction patterns caused by rCsProTα and Ct11 differed considerably. When introduced in vivo, rCsProTα and Ct11 significantly reduced megalocytivirus infection in fish tissues, whereas rCsProTα antibody significantly promoted viral replication. Blocking of Myd88 activity abolished the virus-inhibitory effect of rCsProTα but not Ct11. Taken together, these results demonstrate for the first time that both the intact protein and the C-terminal segment of a teleost ProTα can act like cytokines and induce antiviral immunity via, however, distinct signaling pathways that differ in the requirement of Myd88.
Collapse
Affiliation(s)
- Bao-cun Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Li Sun
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| |
Collapse
|
9
|
Wang JJ, Liu YH, Li GC. Induction of protective and therapeutic anti-cancer immunity by using bispecific anti-idiotype antibody G22-I50 for nasopharyngeal carcinoma. Int Immunopharmacol 2015; 28:1026-33. [PMID: 26303768 DOI: 10.1016/j.intimp.2015.07.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 06/30/2015] [Accepted: 07/20/2015] [Indexed: 11/16/2022]
Abstract
Increasing evidence has suggested that bispecific and multivalent antibodies which have more antigen binding sites will improve their immunogenicity. The bispecific anti-idiotype antibody vaccine G22-I50 was obtained through genetic engineering to enhance the immunogenicity of anti-idiotype antibody vaccines G22 and I50. G22-I50 vaccination could induce anti-tumor immunity in the Balb/c mouse model. The protective and therapeutic efficacy of G22-I50 was also evaluated using the hu-PBL-SCID mouse model injected three times with G22-I50, G22, or I50 mixed with Freund's adjuvant. Results demonstrated that the protective anti-tumor effect of G22-I50 could be relevant with the production of Ab3 antibody and activation of CD8(+) cytotoxic T-lymphocytes. In preventive and therapeutic experiments, G22-I50 could reduce tumor size and prolong the survival time of HNE2-bearing mice (p<0.05). Human CD8(+) T lymphocytes infiltrated the tumor sites, and high levels of human IFN-γ, TNF-α, and caspase-3 were also detected in the tumors from G22-I50-vaccinated and -treated mice. Therefore, the bispecific anti-idiotype antibody vaccine G22-I50 can induce strong humoral and cell-mediated immune responses. This vaccine can be potentially applied to prevent and treat nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Jia-Jia Wang
- Clinical Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China; Tumor Immunobiology Laboratory of Cancer Research Institution, Central South University, Changsha 410078, Hunan Province, China.
| | - Yan-Hong Liu
- Tumor Immunobiology Laboratory of Cancer Research Institution, Central South University, Changsha 410078, Hunan Province, China
| | - Guan-Cheng Li
- Tumor Immunobiology Laboratory of Cancer Research Institution, Central South University, Changsha 410078, Hunan Province, China
| |
Collapse
|
10
|
Karachaliou CE, Liolios C, Triantis C, Zikos C, Samara P, Tsitsilonis OE, Kalbacher H, Voelter W, Papadopoulos M, Pirmettis I, Livaniou E. Specific in vitro binding of a new (99m)Tc-radiolabeled derivative of the C-terminal decapeptide of prothymosin alpha on human neutrophils. Int J Pharm 2015; 486:1-12. [PMID: 25796124 DOI: 10.1016/j.ijpharm.2015.03.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 11/28/2022]
Abstract
Prothymosin alpha (ProTα) is a conserved mammalian polypeptide with intracellular functions associated with cell proliferation and apoptosis and an extracellular role associated with immunopotentiation. The N-terminal fragment [1-28], which is identical with the immunostimulating peptide thymosin α1 (Tα1), was earlier considered as the immunoactive region of the polypeptide; however, recent data suggest that ProTα may exert a discrete immunomodulating action through its central or C-terminal region, via targeting Toll-like receptor- 4 (TLR4). In this work, a derivative of the C-terminal fragment ProTα[100-109] (ProTα-D1) that can be radiolabeled with (99m)Tc was developed. The biological activity of the non-radioactive (185/187)rhenium-complex of this derivative ([(185/187)Re]ProTα-D1, structurally similar with [(99m)Tc]ProTα-D1) was verified through suitable in vitro bioassays on human neutrophils. Subsequent cell-binding studies revealed specific, time-dependent and saturable binding of [(99m)Tc]ProTα-D1 on neutrophils, which was inhibited by intact ProTα and ProTα[100-109], as well as by a "prototype" TLR4-ligand (lipopolysaccharide from Escherichia coli). Overall, our results support the existence of ProTα-binding sites on human neutrophils, recognizing [(99m)Tc]ProTα-D1, which might involve TLR4. [(99m)Tc]ProTα-D1 may be a useful tool for conducting further in vitro and in vivo studies, aiming to elucidate the extracellular mode of action of ProTα and, eventually, develop ProTα-based immunotherapeutics.
Collapse
Affiliation(s)
- Chrysoula-Evangelia Karachaliou
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Christos Liolios
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Charalampos Triantis
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Christos Zikos
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Pinelopi Samara
- Faculty of Biology, University of Athens, Athens 15784, Greece
| | | | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - Wolfgang Voelter
- Interfaculty Institute of Biochemistry, University of Tuebingen, Tuebingen 72076, Germany
| | - Minas Papadopoulos
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Ioannis Pirmettis
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece
| | - Evangelia Livaniou
- Institute of Nuclear and Radiological Sciences & Technology, Energy and Safety (INRaSTES), National Center for Scientific Research "Demokritos", Athens 15310, Greece.
| |
Collapse
|
11
|
Development of an ELISA for the quantification of the C-terminal decapeptide prothymosin α(100-109) in sera of mice infected with bacteria. J Immunol Methods 2013; 395:54-62. [PMID: 23831611 DOI: 10.1016/j.jim.2013.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 11/20/2022]
Abstract
Apoptosis is characterized by a series of discrete biochemical events, among which is the truncation of the nuclear polypeptide prothymosin alpha (proTα) by activated caspase-3. This early apoptotic event results in the generation of a carboxy-terminal fragment of proTα, the immunoactive decapeptide proTα(100-109). We hypothesized that the detection of increased levels of proTα(100-109) in serum can be directly correlated with the induction of massive cell apoptosis, resulting from a severe bacterial infection. Thus, using high-affinity-purified polyclonal antibodies (Abs), raised in rabbits and a prototype antibody-capture system, we developed a highly sensitive and specific competitive ELISA for proTα(100-109). The sensitivity of the ELISA (0.1ng/mL to 10μg/mL) is acceptable for the quantification of the decapeptide in serum samples. To assess our initial hypothesis, we determined the concentration of proTα(100-109) in the sera of mice infected with the bacterium Streptococcus pyogenes over the course of the infection. We show that serum concentration of proTα(100-109) was marginal to undetectable before infection, increased over time and peaked at 72h postinfection. In silico analysis suggests that the Abs generated are unlikely to cross-react with any other unrelated mouse or bacterial protein. Further validation of our ELISA using serum samples from humans, infected with bacteria, may provide a useful tool to differentiate the causative agent of a potentially lethal septic infection.
Collapse
|