1
|
Scapin G, Cagdas E, Grav LM, Lewis NE, Goletz S, Hafkenscheid L. Implications of glycosylation for the development of selected cytokines and their derivatives for medical use. Biotechnol Adv 2024; 77:108467. [PMID: 39447666 DOI: 10.1016/j.biotechadv.2024.108467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cytokines are important regulators of immune responses, making them attractive targets for autoimmune diseases and cancer therapeutics. Yet, the significance of cytokine glycosylation remains underestimated. Many cytokines carry N- and O-glycans and some even undergo C-mannosylation. Recombinant cytokines produced in heterologous host cells may lack glycans or exhibit a different glycosylation pattern such as varying levels of galactosylation, sialylation, fucosylation or xylose addition compared to their human counterparts, potentially impacting critical immune interactions. We focused on cytokines that are currently utilized or designed in advanced therapeutic formats, including immunocytokines, fusokines, engager cytokines, and genetically engineered 'supercytokines.' Despite the innovative designs of these cytokine derivatives, their glycosylation patterns have not been extensively studied. By examining the glycosylation of the human native cytokines, G-CSF and GM-CSF, interferons β and γ, TNF-α and interleukins-2, -3 -4, -6, -7, -9, -12, -13, -15, -17A, -21, and - 22, we aim to assess its potential impact on their therapeutic derivatives. Understanding the glycosylation of the native cytokines could provide critical insights into the safety, efficacy, and functionality of these next-generation cytokine therapies, affecting factors such as stability, bioactivity, antigenicity, and half-life. This knowledge can guide the choice of optimal expression hosts for production and advance the development of effective cytokine-based therapeutics and synthetic immunology drugs.
Collapse
Affiliation(s)
- Giulia Scapin
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Ece Cagdas
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| |
Collapse
|
2
|
Karaaslan BG, Rosain J, Bustamante J, Kıykım A. Interferon Gamma in Sickness Predisposing to Mycobacterial Infectious Diseases. Balkan Med J 2024; 41:326-332. [PMID: 39183693 PMCID: PMC11588913 DOI: 10.4274/balkanmedj.galenos.2024.2024-8-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
In recent decades, the prevalence of inborn errors of immunity has increased, necessitating the development of more effective treatment and care options for these highly morbid conditions. Due to these “experiments of nature,” the complicated nature of the immune system is being revealed. Based on the functional and molecular tests, targeted therapies are now being developed which offer a more effective approach and reduce damage. This study aimed to investigate a key cytokine of the cellular immune response, interferon‐gamma (IFN-γ), which is linked to Mendelian susceptibility to Mycobacterial disease, and its potential as a therapeutic option for IFN-γ deficiency.
Collapse
Affiliation(s)
- Betül Gemici Karaaslan
- Department of Pediatric Allergy and Immunology İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Türkiye
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases Necker Branch, Necker Hospital for Sick Children, Paris, France
- University of Paris Cité Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies Necker Hospital for Sick Children, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases Rockefeller Branch, Rockefeller University, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases Necker Branch, Necker Hospital for Sick Children, Paris, France
- University of Paris Cité Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies Necker Hospital for Sick Children, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases Rockefeller Branch, Rockefeller University, New York, USA
| | - Ayça Kıykım
- Department of Pediatric Allergy and Immunology İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Türkiye
| |
Collapse
|
3
|
Shao Z, Chen L, Zhang Z, Wu Y, Mou H, Jin X, Teng W, Wang F, Yang Y, Zhou H, Xue Y, Eloy Y, Yao M, Zhao S, Cui W, Yu X, Ye Z. KERS-Inspired Nanostructured Mineral Coatings Boost IFN-γ mRNA Therapeutic Index for Antitumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304296. [PMID: 37587307 DOI: 10.1002/adma.202304296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/01/2023] [Indexed: 08/18/2023]
Abstract
Tumor-associated macrophage (TAM) reprogramming is a promising therapeutic approach for cancer immunotherapy; however, its efficacy remains modest due to the low bioactivity of the recombinant cytokines used for TAM reprogramming. mRNA therapeutics are capable of generating fully functional proteins for various therapeutic purposes but accused for its poor sustainability. Inspired by kinetic energy recovery systems (KERS) in hybrid vehicles, a cytokine efficacy recovery system (CERS) is designed to substantially augment the therapeutic index of mRNA-based tumor immunotherapy via a "capture and stabilize" mechanism exerted by a nanostructured mineral coating carrying therapeutic cytokine mRNA. CERS remarkably recycles nearly 40% expressed cytokines by capturing them onto the mineral coating to extend its therapeutic timeframe, further polarizing the macrophages to strengthen their tumoricidal activity and activate adaptive immunity against tumors. Notably, interferon-γ (IFN-γ) produced by CERS exhibits ≈42-fold higher biological activity than recombinant IFN-γ, remarkably decreasing the required IFN-γ dosage for TAM reprogramming. In tumor-bearing mice, IFN-γ cmRNA@CERS effectively polarizes TAMs to inhibit osteosarcoma progression. When combined with the PD-L1 monoclonal antibody, IFN-γ cmRNA@CERS significantly boosts antitumor immune responses, and substantially prevents malignant lung metastases. Thus, CERS-mediated mRNA delivery represents a promising strategy to boost antitumor immunity for tumor treatment.
Collapse
Affiliation(s)
- Zhenxuan Shao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Liang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Zengjie Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Yan Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Xiaoqiang Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Wangsiyuan Teng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Yinxian Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hao Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Yinwang Eloy
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Minjun Yao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Shenzhi Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiaohua Yu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Orthopedics Research Institute of Zhejiang University, Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Clinical Research Center of Motor System Disease of Zhejiang Province, 88 Jiefang Road, Hangzhou City, Zhejiang Province, 310003, P. R. China
| |
Collapse
|
4
|
Dong S, Liu X, Bi Y, Wang Y, Antony A, Lee D, Huntoon K, Jeong S, Ma Y, Li X, Deng W, Schrank BR, Grippin AJ, Ha J, Kang M, Chang M, Zhao Y, Sun R, Sun X, Yang J, Chen J, Tang SK, Lee LJ, Lee AS, Teng L, Wang S, Teng L, Kim BYS, Yang Z, Jiang W. Adaptive design of mRNA-loaded extracellular vesicles for targeted immunotherapy of cancer. Nat Commun 2023; 14:6610. [PMID: 37857647 PMCID: PMC10587228 DOI: 10.1038/s41467-023-42365-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
The recent success of mRNA therapeutics against pathogenic infections has increased interest in their use for other human diseases including cancer. However, the precise delivery of the genetic cargo to cells and tissues of interest remains challenging. Here, we show an adaptive strategy that enables the docking of different targeting ligands onto the surface of mRNA-loaded small extracellular vesicles (sEVs). This is achieved by using a microfluidic electroporation approach in which a combination of nano- and milli-second pulses produces large amounts of IFN-γ mRNA-loaded sEVs with CD64 overexpressed on their surface. The CD64 molecule serves as an adaptor to dock targeting ligands, such as anti-CD71 and anti-programmed cell death-ligand 1 (PD-L1) antibodies. The resulting immunogenic sEVs (imsEV) preferentially target glioblastoma cells and generate potent antitumour activities in vivo, including against tumours intrinsically resistant to immunotherapy. Together, these results provide an adaptive approach to engineering mRNA-loaded sEVs with targeting functionality and pave the way for their adoption in cancer immunotherapy applications.
Collapse
Affiliation(s)
- Shiyan Dong
- School of Life Science, Jilin University, Changchun, 130012, China
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xuan Liu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Chemical Engineering, Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Ye Bi
- Practice Training Center, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Abin Antony
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Seongdong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yifan Ma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Xuefeng Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiye Deng
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Benjamin R Schrank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Adam J Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - JongHoon Ha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Minjeong Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengyu Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yarong Zhao
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Rongze Sun
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Xiangshi Sun
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Jie Yang
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Jiayi Chen
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Sarah K Tang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Spot Biosystems Ltd., Palo Alto, CA, 94305, USA
| | - Andrew S Lee
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518055, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Lirong Teng
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Shengnian Wang
- Chemical Engineering, Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA, 71272, USA.
| | - Lesheng Teng
- School of Life Science, Jilin University, Changchun, 130012, China.
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Zhaogang Yang
- School of Life Science, Jilin University, Changchun, 130012, China.
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Mertowska P, Smolak K, Mertowski S, Grywalska E. Immunomodulatory Role of Interferons in Viral and Bacterial Infections. Int J Mol Sci 2023; 24:10115. [PMID: 37373262 DOI: 10.3390/ijms241210115] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Interferons are a group of immunomodulatory substances produced by the human immune system in response to the presence of pathogens, especially during viral and bacterial infections. Their remarkably diverse mechanisms of action help the immune system fight infections by activating hundreds of genes involved in signal transduction pathways. In this review, we focus on discussing the interplay between the IFN system and seven medically important and challenging viruses (herpes simplex virus (HSV), influenza, hepatitis C virus (HCV), lymphocytic choriomeningitis virus (LCMV), human immunodeficiency virus (HIV), Epstein-Barr virus (EBV), and SARS-CoV coronavirus) to highlight the diversity of viral strategies. In addition, the available data also suggest that IFNs play an important role in the course of bacterial infections. Research is currently underway to identify and elucidate the exact role of specific genes and effector pathways in generating the antimicrobial response mediated by IFNs. Despite the numerous studies on the role of interferons in antimicrobial responses, many interdisciplinary studies are still needed to understand and optimize their use in personalized therapeutics.
Collapse
Affiliation(s)
- Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
6
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
7
|
Santana H, González Y, Dorta D, Blanco R, Viltres Y, González J, Espinosa LA, Leyva A, Bello I. Stability-indicative and conformation-specific enzyme linked immunosorbent assay for analysis of recombinant human gamma interferon. J Pharm Biomed Anal 2023; 222:115070. [DOI: 10.1016/j.jpba.2022.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/06/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022]
|
8
|
Hemmati S, Rasekhi Kazerooni H. Polypharmacological Cell-Penetrating Peptides from Venomous Marine Animals Based on Immunomodulating, Antimicrobial, and Anticancer Properties. Mar Drugs 2022; 20:md20120763. [PMID: 36547910 PMCID: PMC9787916 DOI: 10.3390/md20120763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022] Open
Abstract
Complex pathological diseases, such as cancer, infection, and Alzheimer's, need to be targeted by multipronged curative. Various omics technologies, with a high rate of data generation, demand artificial intelligence to translate these data into druggable targets. In this study, 82 marine venomous animal species were retrieved, and 3505 cryptic cell-penetrating peptides (CPPs) were identified in their toxins. A total of 279 safe peptides were further analyzed for antimicrobial, anticancer, and immunomodulatory characteristics. Protease-resistant CPPs with endosomal-escape ability in Hydrophis hardwickii, nuclear-localizing peptides in Scorpaena plumieri, and mitochondrial-targeting peptides from Synanceia horrida were suitable for compartmental drug delivery. A broad-spectrum S. horrida-derived antimicrobial peptide with a high binding-affinity to bacterial membranes was an antigen-presenting cell (APC) stimulator that primes cytokine release and naïve T-cell maturation simultaneously. While antibiofilm and wound-healing peptides were detected in Synanceia verrucosa, APC epitopes as universal adjuvants for antiviral vaccination were in Pterois volitans and Conus monile. Conus pennaceus-derived anticancer peptides showed antiangiogenic and IL-2-inducing properties with moderate BBB-permeation and were defined to be a tumor-homing peptide (THP) with the ability to inhibit programmed death ligand-1 (PDL-1). Isoforms of RGD-containing peptides with innate antiangiogenic characteristics were in Conus tessulatus for tumor targeting. Inhibitors of neuropilin-1 in C. pennaceus are proposed for imaging probes or therapeutic delivery. A Conus betulinus cryptic peptide, with BBB-permeation, mitochondrial-targeting, and antioxidant capacity, was a stimulator of anti-inflammatory cytokines and non-inducer of proinflammation proposed for Alzheimer's. Conclusively, we have considered the dynamic interaction of cells, their microenvironment, and proportional-orchestrating-host- immune pathways by multi-target-directed CPPs resembling single-molecule polypharmacology. This strategy might fill the therapeutic gap in complex resistant disorders and increase the candidates' clinical-translation chance.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Correspondence: ; Tel.: +98-7132-424-128
| | | |
Collapse
|
9
|
Rojas-Peña M, Aceituno P, Salvador ME, Garcia-Ordoñez M, Teles M, Ortega-Villaizan MDM, Perez L, Roher N. How modular protein nanoparticles may expand the ability of subunit anti-viral vaccines: The spring viremia carp virus (SVCV) case. FISH & SHELLFISH IMMUNOLOGY 2022; 131:1051-1062. [PMID: 36371050 DOI: 10.1016/j.fsi.2022.10.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/08/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
Spring viremia of carp (SVC) remains as a vaccine orphan disease mostly affecting juvenile specimens. Young fish are especially difficult to vaccinate and oral administration of vaccine combined with food would be the election system to minimise stress and the vaccination costs associated to injection. However, administration of prophylactics with food pellets faces off several drawbacks mainly related with vaccine degradation and weak protection correlates of oral vaccines. Here we present a platform based on recombinant proteins (subunit vaccines) manufactured as highly resistant nanostructured materials, and providing excellent levels of protection against SVC virus in a preliminary i.p injection challenge. The G3 domain of SVCV glycoprotein G was overexpressed in E. coli together with IFNγ and the modular protein was purified from bacterial aggregates (inclusion bodies) as highly organised nanostructured biomaterial (nanopellets, NP). These SVCV-IFNNP were taken up by zebrafish cells leading to the enhanced expression of different antiviral and IFN markers (e.g vig1, mx, lmp2 or ifngr1 among others) in zebrafish liver cells (ZFL). To monitor if SVCVNP and SVCV-IFNNP can be taken up by intestinal epithelia and can induce antiviral response we performed experiments with SVCVNP and SVCV-IFNNP in 3 days post fertilization (dpf) zebrafish larvae. Both, SVCVNP and SVCV-IFNNP were taken up and accumulated in the intestine without signs of toxicity. The antiviral response in larvae showed a different induction pattern: SVCV-IFNNP did not induce an antiviral response while SVCVNP showed a good antiviral induction. Interestingly ZF4, an embryonic derived cell line, showed an antiviral response like ZFL cells, although the lmp2 and ifngr1 (markers of the IFNγ response) were not overexpressed. Experiments with adult zebrafish indicated an excellent level of protection against a SVCV model infection where SVCV-IFNNP vaccinated fish reached 20% cumulative mortality while control fish reached over 80% cumulative mortality.
Collapse
Affiliation(s)
- Mauricio Rojas-Peña
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Patricia Aceituno
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Maria E Salvador
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), Elche, Spain
| | - Marlid Garcia-Ordoñez
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Mariana Teles
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain; Department of Cell Biology, Animal Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
| | - Maria Del Mar Ortega-Villaizan
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), Elche, Spain
| | - Luis Perez
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández (UMH), Elche, Spain.
| | - Nerea Roher
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain; Department of Cell Biology, Animal Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
10
|
Kajiura H, Tatematsu KI, Nomura T, Miyazawa M, Usami A, Tamura T, Sezutsu H, Fujiyama K. Insights into the quality of recombinant proteins produced by two different Bombyx mori expression systems. Sci Rep 2022; 12:18502. [PMID: 36323753 PMCID: PMC9628610 DOI: 10.1038/s41598-022-22565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 10/17/2022] [Indexed: 11/26/2022] Open
Abstract
The silkworm, Bombyx mori, is an attractive host for recombinant protein production due to its high expression efficiency, quality, and quantity. Two expression systems have been widely used for recombinant protein production in B. mori: baculovirus/silkworm expression system and transgenic silkworm expression system. Both expression systems enable high protein production, but the qualities of the resulting recombinant proteins have not been well evaluated. In this study, we expressed bovine interferon γ (IFN-γ) using the two systems and examined the quality of the resulting proteins in terms of N-glycosylation and protein cleavage. Both expression systems successfully produced IFN-γ as an N-glycoprotein. Although the production in the baculovirus/silkworm expression system was much more efficient than that in the transgenic silkworm expression system, unexpected variants of IFN-γ were also produced in the former system due to the different N-glycosylation and C-terminal truncations. These results indicate that while high protein production could be achieved in the baculovirus/silkworm expression system, unintentional protein modification might occur, and therefore protein expression in the transgenic silkworm expression system is preferable from the point-of-view of N-glycosylation of the recombinant protein and evasion of unexpected attack by a protease in B. mori.
Collapse
Affiliation(s)
- Hiroyuki Kajiura
- grid.136593.b0000 0004 0373 3971International Center for Biotechnology, Osaka University, 2-1 Yamada-Oka, Suita-Shi, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-1 Yamada-Oka, Suita-Shi, Osaka, 565-0871 Japan
| | - Ken-ichiro Tatematsu
- grid.416835.d0000 0001 2222 0432Division of Silk-Producing Insect Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, 1-2 Owashi, Tsukuba, Ibaraki 305-8634 Japan
| | - Tsuyoshi Nomura
- grid.419812.70000 0004 1777 4627Sysmex Corporation, 1548 Ooaza Shimookudomi, Sayama, Saitama 350-1332 Japan
| | - Mitsuhiro Miyazawa
- grid.416835.d0000 0001 2222 0432Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, 1-2 Owashi, Tsukuba, Ibaraki 305-8634 Japan
| | - Akihiro Usami
- grid.419812.70000 0004 1777 4627Sysmex Corporation, 1548 Ooaza Shimookudomi, Sayama, Saitama 350-1332 Japan
| | - Toshiki Tamura
- grid.416629.e0000 0004 0377 2137Silk Science and Technology Research Institute, 1053, Iikura, Ami-Machi, Ibaraki, 300-0324 Japan
| | - Hideki Sezutsu
- grid.416835.d0000 0001 2222 0432Division of Silk-Producing Insect Biotechnology, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, 1-2 Owashi, Tsukuba, Ibaraki 305-8634 Japan
| | - Kazuhito Fujiyama
- grid.136593.b0000 0004 0373 3971International Center for Biotechnology, Osaka University, 2-1 Yamada-Oka, Suita-Shi, Osaka, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-1 Yamada-Oka, Suita-Shi, Osaka, 565-0871 Japan ,grid.10223.320000 0004 1937 0490Osaka University Cooperative Research Station in Southeast Asia (OU:CRS), Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
11
|
Kebeish R, Hamdy E, Al-Zoubi O, Habeeb T, Osailan R, El-Ayouty Y. A Biotechnological Approach for the Production of Pharmaceutically Active Human Interferon-α from Raphanus sativus L. Plants. Bioengineering (Basel) 2022; 9:bioengineering9080381. [PMID: 36004906 PMCID: PMC9404948 DOI: 10.3390/bioengineering9080381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Human interferon (IFN) is a type of cytokine that regulates the immune system’s response to viral and bacterial infections. Recombinant IFN-α has been approved for use in the treatment of a variety of viral infections as well as an anticancer medication for various forms of leukemia. The objective of the current study is to produce a functionally active recombinant human IFN-α2a from transgenic Raphanus sativus L. plants. Therefore, a binary plant expression construct containing the IFN-α2a gene coding sequence, under the regulation of the cauliflower mosaic virus 35SS promoter, was established. Agrobacterium-mediated floral dip transformation was used to introduce the IFN-α2a expression cassette into the nuclear genome of red and white rooted Raphanus sativus L. plants. From each genotype, three independent transgenic lines were established. The anticancer and antiviral activities of the partially purified recombinant IFN-α2a proteins were examined. The isolated IFN-α2a has been demonstrated to inhibit the spread of the Vesicular Stomatitis Virus (VSV). In addition, cytotoxicity and cell apoptosis assays against Hep-G2 cells (Human Hepatocellular Carcinoma) show the efficacy of the generated IFN-α2a as an anticancer agent. In comparison to bacterial, yeast, and animal cell culture systems, the overall observed results demonstrated the efficacy of using Raphanus sativus L. plants as a safe, cost-effective, and easy-to-use expression system for generating active human IFN-α2a.
Collapse
Affiliation(s)
- Rashad Kebeish
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Biology Department, Faculty of Science Yanbu, Taibah University, Yanbu El-Bahr 46423, Saudi Arabia
- Correspondence: or
| | - Emad Hamdy
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Omar Al-Zoubi
- Biology Department, Faculty of Science Yanbu, Taibah University, Yanbu El-Bahr 46423, Saudi Arabia
| | - Talaat Habeeb
- Biology Department, Faculty of Science Yanbu, Taibah University, Yanbu El-Bahr 46423, Saudi Arabia
| | - Raha Osailan
- Biology Department, Faculty of Science Yanbu, Taibah University, Yanbu El-Bahr 46423, Saudi Arabia
| | - Yassin El-Ayouty
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
12
|
Wu B, Song M, Dong Q, Xiang G, Li J, Ma X, Wei F. UBR5 promotes tumor immune evasion through enhancing IFN-γ-induced PDL1 transcription in triple negative breast cancer. Am J Cancer Res 2022; 12:5086-5102. [PMID: 35836797 PMCID: PMC9274738 DOI: 10.7150/thno.74989] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 01/12/2023] Open
Abstract
Background: The up-regulation of PD-L1 is recognized as an adaption of cancer cells to evade immune surveillance and attack. However, the intrinsic mechanisms of the induction of PD-L1 by interferon-γ (IFN-γ) in tumor microenvironment remain incompletely characterized. Ubiquitin ligase E3 component N-recognition protein 5 (UBR5) has a critical role in tumorigenesis of triple negative breast cancer (TNBC) by triggering specific immune responses to the tumor. Dual targeting of UBR5 and PD-L1 exhibited superior therapeutic benefits in a preclinical TNBC model in short term. Methods: The regulation of UBR5 to PD-L1 upon IFN-γ stimulation was evaluated through in UBR5 deficiency, reconstitution or overexpression cell line models by quantitative PCR, immunohistochemistry and RNA-seq. The effects of PD-L1 regulation by UBR5 and double blockade of both genes were evaluated in mouse TNBC model. Luciferase reporter assay, chromatin immunoprecipitation-qPCR and bioinformatics analysis were performed to explore the transcription factors involved in the regulation of UBR5 to PD-L1. Results: E3 ubiquitin ligase UBR5 plays a key role in IFN-γ-induced PDL1 transcription in TNBC in an E3 ubiquitination activity-independent manner. RNA-seq-based transcriptomic analyses reveal that UBR5 globally affects the genes in the IFN-γ-induced signaling pathway. Through its poly adenylate binding (PABC) domain, UBR5 enhances the transactivation of PDL1 by upregulating protein kinase RNA-activated (PKR), and PKR's downstream factors including signal transducers and activators of transcription 1 (STAT1) and interferon regulatory factor 1 (IRF1). Restoration of PD-L1 expression in UBR5-deficient tumor cells recoups their malignancy in vivo, whereas CRISPR/Cas9-mediated simultaneous abrogation of UBR5 and PD-L1 expression yields synergistic therapeutic benefits than either blockade alone, with a strong impact on the tumor microenvironment. Conclusions: This study identifies a novel regulator of PDL1 transcription, elucidates the underlying molecular mechanisms and provides a strong rationale for combination cancer immunotherapies targeting UBR5 and PD-L1.
Collapse
Affiliation(s)
- Bingbing Wu
- Sheng Yushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Mei Song
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York
| | - Qun Dong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Gang Xiang
- Sheng Yushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York.,✉ Corresponding author: Fang Wei, 800 Dongchuan Road, Minghang, Shanghai 200240, China. Phone: 86-21-34205287; Fax: 86-21-34205287; E-mail: ; Xiaojing Ma,
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,✉ Corresponding author: Fang Wei, 800 Dongchuan Road, Minghang, Shanghai 200240, China. Phone: 86-21-34205287; Fax: 86-21-34205287; E-mail: ; Xiaojing Ma,
| |
Collapse
|
13
|
Li Z, Ma R, Ma S, Tian L, Lu T, Zhang J, Mundy-Bosse BL, Zhang B, Marcucci G, Caligiuri MA, Yu J. ILC1s control leukemia stem cell fate and limit development of AML. Nat Immunol 2022; 23:718-730. [PMID: 35487987 PMCID: PMC9106917 DOI: 10.1038/s41590-022-01198-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022]
Abstract
Type I innate lymphoid cells (ILC1s) are critical regulators of inflammation and immunity in mammalian tissues. However, their function in cancer is mostly undefined. Here, we show that a high density of ILC1s induces leukemia stem cell (LSC) apoptosis in mice. At a lower density, ILC1s prevent LSCs from differentiating into leukemia progenitors and promote their differentiation into non-leukemic cells, thus blocking the production of terminal myeloid blasts. All of these effects, which require ILC1s to produce interferon-γ after cell-cell contact with LSCs, converge to suppress leukemogenesis in vivo. Conversely, the antileukemia potential of ILC1s wanes when JAK-STAT or PI3K-AKT signaling is inhibited. The relevant antileukemic properties of ILC1s are also functional in healthy individuals and impaired in individuals with acute myeloid leukemia (AML). Collectively, these findings identify ILC1s as anticancer immune cells that might be suitable for AML immunotherapy and provide a potential strategy to treat AML and prevent relapse of the disease.
Collapse
Affiliation(s)
- Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Rui Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Shoubao Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Ting Lu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bin Zhang
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Shahin K, Zhang L, Mehraban MH, Collard JM, Hedayatkhah A, Mansoorianfar M, Soleimani-Delfan A, Wang R. Clinical and experimental bacteriophage studies: Recommendations for possible approaches for standing against SARS-CoV-2. Microb Pathog 2022; 164:105442. [PMID: 35151823 PMCID: PMC8830156 DOI: 10.1016/j.micpath.2022.105442] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/23/2022]
Abstract
In 2019, the world faced a serious health challenge, the rapid spreading of a life-threatening viral pneumonia, coronavirus disease 2019 (COVID-19) caused by a betacoronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As of January 2022 WHO statistics shows more than 5.6 million death and about 350 million infection by SARS-CoV-2. One of the life threatening aspects of COVID-19 is secondary infections and reduced efficacy of antibiotics against them. Since the beginning of COVID-19 many researches have been done on identification, treatment, and vaccine development. Bacterial viruses (bacteriophages) could offer novel approaches to detect, treat and control COVID-19. Phage therapy and in particular using phage cocktails can be used to control or eliminate the bacterial pathogen as an alternative or complementary therapeutic agent. At the same time, phage interaction with the host immune system can regulate the inflammatory response. In addition, phage display and engineered synthetic phages can be utilized to develop new vaccines and antibodies, stimulate the immune system, and elicit a rapid and well-appropriate defense response. The emergence of SARS-CoV-2 new variants like delta and omicron has proved the urgent need for precise, efficient and novel approaches for vaccine development and virus detection techniques in which bacteriophages may be one of the plausible solutions. Therefore, phages with similar morphology and/or genetic content to that of coronaviruses can be used for ecological and epidemiological modeling of SARS-CoV-2 behavior and future generations of coronavirus, and in general new viral pathogens. This article is a comprehensive review/perspective of potential applications of bacteriophages in the fight against the present pandemic and the post-COVID era.
Collapse
Affiliation(s)
- Khashayar Shahin
- Center for Microbes, Development, and Health (CMDH), Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200025, China; Key Laboratory of Phage Research, International Phage Research Center, Jiangsu Academy of Agricultural, China; Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
| | - Lili Zhang
- Key Laboratory of Phage Research, International Phage Research Center, Jiangsu Academy of Agricultural, China; Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Mohammad Hossein Mehraban
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jean-Marc Collard
- Center for Microbes, Development, and Health (CMDH), Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200025, China
| | | | | | - Abbas Soleimani-Delfan
- Key Laboratory of Phage Research, International Phage Research Center, Jiangsu Academy of Agricultural, China; Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Ran Wang
- Key Laboratory of Phage Research, International Phage Research Center, Jiangsu Academy of Agricultural, China; Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China.
| |
Collapse
|
15
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
16
|
Cao L, Zhang L, Zhang X, Liu J, Jia MA, Zhang J, Liu J, Wang F. Types of Interferons and Their Expression in Plant Systems. J Interferon Cytokine Res 2022; 42:62-71. [PMID: 35171703 DOI: 10.1089/jir.2021.0148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interferons (IFNs) are divided into 3 types (type I, type II, and type III) on the basis of sequence homology and functional properties. Recombinant IFNs have been approved by regulatory agencies in many countries for clinical treatment of hepatitis B, hepatitis C, and other diseases; these IFNs are mainly produced in microorganisms and mammalian cell systems. However, there are serious obstacles to the production of recombinant IFNs in microorganism systems; for example, the recombinant IFN may have different glycosylation patterns from the native protein, be present in insoluble inclusion bodies, be contaminated with impurities such as endotoxins and nucleic acids, have a short half-life in human blood, and incur high production costs. Some medicinal proteins have been successfully expressed in plants and used in clinical applications, suggesting that plants may also be a good system for IFN expression. However, there are still many technical problems that need to be addressed before the clinical application of plant-expressed IFNs, such as increasing the amount of recombinant protein expression and ensuring that the IFN is modified with the correct type of glycosylation. In this article, we review the classification of IFNs, their roles in antiviral signal transduction pathways, their clinical applications, and their expression in plant systems.
Collapse
Affiliation(s)
- Linggai Cao
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| | - Lili Zhang
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| | - Xiaolian Zhang
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| | - Jia Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Meng-Ao Jia
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| | - Jishun Zhang
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| | - Jiemin Liu
- Guizhou Provincial People's Hospital, Guiyang, China
| | - Feng Wang
- Key Laboratory of Molecular Genetics, China National Tobacco Corporation, Guizhou Academy of Tobacco Science, Guiyang, China
| |
Collapse
|
17
|
Vesicular IFN-γ as a cooperative attacker to enhance anti-cancer effect of 5-fluorouracil via thymidine phosphorylase upregulation and tumor microenvironment normalization. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 40:102501. [PMID: 34843983 DOI: 10.1016/j.nano.2021.102501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/15/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022]
Abstract
On the basis of immuno-modulating effect and upregulating the activity of thymidine phosphorylase (TP), interferon-γ (IFN-γ) as a cooperative attacker was explored to enhance the anticancer activity of 5-fluorouracil (5-FU). We designed and prepared a self-assembled nano-vesicular system IFN-γ-EDP formulated by amphiphilic poly((polyethylene glycol)(dodecylphosphoethanolamine)phosphazene) (EDP) to entrap IFN-γ in the hydrophilic cavity. The IFN-γ-EDP vesicles allowed IFN-γ to accumulate at the tumor site and be taken up by tumor cells, resulting in significantly upregulated expression level of TP, distinct inhibition of cell growth, more cellular apoptosis and more serious cell cycle arrest when administrated combined with 5-FU. Moreover, IFN-γ-EDP could normalize the tumor microenvironment by enhancing the CD4+ and CD8+ T cell populations, promoting the IL-12 secretion and suppressing the IL-10 secretion in tumor. As a consequence, the combination therapy of IFN-γ-EDP with 5-FU achieved remarkably enhanced tumor inhibition rate of 56.9% against CT26 colorectal cancer.
Collapse
|
18
|
Karki R, Rimal S, Rieth MD. Predicted N-terminal N-linked glycosylation sites may underlie membrane protein expression patterns in Saccharomyces cerevisiae. Yeast 2021; 38:497-506. [PMID: 34182612 DOI: 10.1002/yea.3657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
N-linked glycosylation is one type of posttranslational modification that proteins undergo during expression. The following describes the effects of N-linked glycosylation on high-level membrane protein expression in yeast with an emphasis on Saccharomyces cerevisiae. N-linked glycosylation is highlighted here as an important consideration when preparing membrane protein gene constructs for expression in S. cerevisiae, which continues to be used as a workhorse in both research and industrial applications. Non-native N-linked glycosylation commonly occurs during the heterologous expression of mammalian proteins in many yeast species which can have important immunological consequences when used in the production of biotherapeutic proteins or peptides. Further, non-native N-linked glycosylation can lead to improper protein folding and premature degradation, which can impede high-level expression yields and hinder downstream analysis. Multiple strategies are presented in this article, which suggest different methods that can be implemented to circumvent the unwanted consequences of N-linked glycosylation during the expression process. These considerations may have long-term benefits for high-level protein production in S. cerevisiae across a broad spectrum of expression targets with special emphasis placed on G-protein coupled receptors, one of the largest families of membrane proteins.
Collapse
Affiliation(s)
- Rashmi Karki
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Swechha Rimal
- Department of Chemistry, Southern Illinois University Edwardsville, Edwardsville, Illinois, USA
| | - Monica D Rieth
- Department of Chemistry, Southern Illinois University Edwardsville, Edwardsville, Illinois, USA
| |
Collapse
|
19
|
Santibañez A, Paine D, Parra M, Muñoz C, Valdes N, Zapata C, Vargas R, Gonzalez A, Tello M. Oral Administration of Lactococcus lactis Producing Interferon Type II, Enhances the Immune Response Against Bacterial Pathogens in Rainbow Trout. Front Immunol 2021; 12:696803. [PMID: 34248997 PMCID: PMC8268009 DOI: 10.3389/fimmu.2021.696803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/08/2021] [Indexed: 11/13/2022] Open
Abstract
Lactic acid bacteria are a powerful vehicle for releasing of cytokines and immunostimulant peptides at the gastrointestinal level after oral administration. However, its therapeutic application against pathogens that affect rainbow trout and Atlantic salmon has been little explored. Type II interferon in Atlantic salmon activates the antiviral response, protecting against viral infection, but its role against bacterial infection has not been tested in vivo. In this work, through the design of a recombinant lactic acid bacterium capable of producing Interferon gamma from Atlantic salmon, we explore its role against bacterial infection and the ability to stimulate systemic immune response after oral administration of the recombinant probiotic. Recombinant interferon was active in vitro, mainly stimulating IL-6 expression in SHK-1 cells. In vivo, oral administration of the recombinant probiotic produced an increase in IL-6, IFNγ and IL-12 in the spleen and kidney, in addition to stimulating the activity of lysozyme in serum. The challenge trials indicated that the administration of the IFNγ-producing probiotic doubled the survival in fish infected with F. psychrophilum. In conclusion, our results showed that the oral administration of lactic acid bacteria producing IFNγ managed to stimulate the immune response at a systemic level, conferring protection against pathogens, showing a biotechnological potential for its application in aquaculture.
Collapse
Affiliation(s)
- Alvaro Santibañez
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Diego Paine
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Mick Parra
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Carlos Muñoz
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Natalia Valdes
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Claudia Zapata
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
| | - Rodrigo Vargas
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Alex Gonzalez
- Laboratorio de Microbiología Ambiental y Extremófilos, Departamento de Ciencias Biológicas, Universidad de los Lagos, Osorno, Chile
| | - Mario Tello
- Departamento de Biología, Laboratorio de Metagenómica Bacteriana, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Consorcio Tecnológico de Sanidad Acuícola, Ictio Biotechnologies S.A., Santiago, Chile
- IctioBiotic SpA, Santiago, Chile
| |
Collapse
|
20
|
Gocher AM, Workman CJ, Vignali DAA. Interferon-γ: teammate or opponent in the tumour microenvironment? Nat Rev Immunol 2021; 22:158-172. [PMID: 34155388 DOI: 10.1038/s41577-021-00566-3] [Citation(s) in RCA: 283] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy offers substantive benefit to patients with various tumour types, in some cases leading to complete tumour clearance. However, many patients do not respond to immunotherapy, galvanizing the field to define the mechanisms of pre-existing and acquired resistance. Interferon-γ (IFNγ) is a cytokine that has both protumour and antitumour activities, suggesting that it may serve as a nexus for responsiveness to immunotherapy. Many cancer immunotherapies and chemotherapies induce IFNγ production by various cell types, including activated T cells and natural killer cells. Patients resistant to these therapies commonly have molecular aberrations in the IFNγ signalling pathway or express resistance molecules driven by IFNγ. Given that all nucleated cells can respond to IFNγ, the functional consequences of IFNγ production need to be carefully dissected on a cell-by-cell basis. Here, we review the cells that produce IFNγ and the different effects of IFNγ in the tumour microenvironment, highlighting the pleiotropic nature of this multifunctional and abundant cytokine.
Collapse
Affiliation(s)
- Angela M Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Dickel S, Grimm C, Amschler K, Schnitzler SU, Schanz J, Moerer O, Payen D, Tampe B, Winkler MS. Case Report: Interferon-γ Restores Monocytic Human Leukocyte Antigen Receptor (mHLA-DR) in Severe COVID-19 With Acquired Immunosuppression Syndrome. Front Immunol 2021; 12:645124. [PMID: 33897692 PMCID: PMC8058468 DOI: 10.3389/fimmu.2021.645124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background The major histocompatibility complex (MHC) class II characterized by monocytes CD14+ expression of human leukocyte antigen receptors (HLA-DR), is essential for the synapse between innate and adaptive immune response in infectious disease. Its reduced expression is associated with a high risk of secondary infections in septic patients and can be safely corrected by Interferon-y (IFNy) injection. Coronavirus disease (COVID-19) induces an alteration of Interferon (IFN) genes expression potentially responsible for the observed low HLA-DR expression in circulating monocytes (mHLA-DR). Methods We report a case of one-time INFy injection (100 mcg s.c.) in a superinfected 61-year-old man with COVID-19–associated acute respiratory distress syndrome (ARDS), with monitoring of mHLA-DR expression and clinical tolerance. Observations Low mHLA-DR pretreatment expression (26.7%) was observed. IFNy therapy leading to a rapid increase in mHLA-DR expression (83.1%). Conclusions Severe ARDS in a COVID-19 patient has a deep reduction in mHLA-DR expression concomitantly with secondary infections. The unique IFNy injection was safe and led to a sharp increase in the expression of mHLA-DR. Based on immune and infection monitoring, more cases of severe COVID-19 patients with low mHLA-DR should be treated by IFNy to test the clinical effectiveness.
Collapse
Affiliation(s)
- Steffen Dickel
- Department of Anesthesiology and Intensive Care Medicine, University Medical, Center Goettingen, Goettingen, Germany
| | - Clemens Grimm
- Department of Anesthesiology and Intensive Care Medicine, University Medical, Center Goettingen, Goettingen, Germany
| | - Katharina Amschler
- Department of Dermatology, University Medical Center Goettingen, Goettingen, Germany
| | - Sebastian Uwe Schnitzler
- Department of Anesthesiology and Intensive Care Medicine, University Medical, Center Goettingen, Goettingen, Germany
| | - Julie Schanz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Goettingen, Goettingen, Germany
| | - Onnen Moerer
- Department of Anesthesiology and Intensive Care Medicine, University Medical, Center Goettingen, Goettingen, Germany
| | - Didier Payen
- Universite Paris 7 Cite Sorbonne, UMR INSERM 1160, Paris, France
| | - Bjoern Tampe
- Department of Nephrology and Rheumatology, University Medical Center, Goettingen, Goettingen, Germany
| | - Martin Sebastian Winkler
- Department of Anesthesiology and Intensive Care Medicine, University Medical, Center Goettingen, Goettingen, Germany
| |
Collapse
|
22
|
Niccolai E, Russo E, Baldi S, Ricci F, Nannini G, Pedone M, Stingo FC, Taddei A, Ringressi MN, Bechi P, Mengoni A, Fani R, Bacci G, Fagorzi C, Chiellini C, Prisco D, Ramazzotti M, Amedei A. Significant and Conflicting Correlation of IL-9 With Prevotella and Bacteroides in Human Colorectal Cancer. Front Immunol 2021; 11:573158. [PMID: 33488574 PMCID: PMC7820867 DOI: 10.3389/fimmu.2020.573158] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background and aim Gut microbiota (GM) can support colorectal cancer (CRC) progression by modulating immune responses through the production of both immunostimulatory and/or immunosuppressive cytokines. The role of IL-9 is paradigmatic because it can either promote tumor progression in hematological malignancies or inhibit tumorigenesis in solid cancers. Therefore, we investigate the microbiota–immunity axis in healthy and tumor mucosa, focusing on the correlation between cytokine profile and GM signature. Methods In this observational study, we collected tumor (CRC) and healthy (CRC-S) mucosa samples from 45 CRC patients, who were undergoing surgery in 2018 at the Careggi University Hospital (Florence, Italy). First, we characterized the tissue infiltrating lymphocyte subset profile and the GM composition. Subsequently, we evaluated the CRC and CRC-S molecular inflammatory response and correlated this profile with GM composition, using Dirichlet multinomial regression. Results CRC samples displayed higher percentages of Th17, Th2, and Tregs. Moreover, CRC tissues showed significantly higher levels of MIP-1α, IL-1α, IL-1β, IL-2, IP-10, IL-6, IL-8, IL-17A, IFN-γ, TNF-α, MCP-1, P-selectin, and IL-9. Compared to CRC-S, CRC samples also showed significantly higher levels of the following genera: Fusobacteria, Proteobacteria, Fusobacterium, Ruminococcus2, and Ruminococcus. Finally, the abundance of Prevotella spp. in CRC samples negatively correlated with IL-17A and positively with IL-9. On the contrary, Bacteroides spp. presence negatively correlated with IL-9. Conclusions Our data consolidate antitumor immunity impairment and the presence of a distinct microbiota profile in the tumor microenvironment compared with the healthy mucosa counterpart. Relating the CRC cytokine profile with GM composition, we confirm the presence of bidirectional crosstalk between the immune response and the host’s commensal microorganisms. Indeed, we document, for the first time, that Prevotella spp. and Bacteroides spp. are, respectively, positively and negatively correlated with IL-9, whose role in CRC development is still under debate.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Federica Ricci
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" University of Florence, Florence, Italy
| | - Giulia Nannini
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications "G. Parenti", Florence, Italy
| | | | - Antonio Taddei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Paolo Bechi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Alessio Mengoni
- Department of Biology, University of Florence, Florence, Italy
| | - Renato Fani
- Department of Biology, University of Florence, Florence, Italy
| | - Giovanni Bacci
- Department of Biology, University of Florence, Florence, Italy
| | - Camilla Fagorzi
- Department of Biology, University of Florence, Florence, Italy
| | | | - Domenico Prisco
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy.,SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy.,SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| |
Collapse
|
23
|
Brilhante Bezerra FS, Silva Rezende ADF, Oliveira Silva MTD, Sena-Lopes Â, Roesch-Ely M, Pêgas Henriques JA, Padilha FF, Carvalho Azevedo VA, Dias Portela RW, Seixas FK, Collares TV, Savegnago L, Borsuk S. The combination of Brazilian red propolis and recombinant protein rCP01850 in the immunoprophylaxis of Corynebacterium pseudotuberculosis infection in mice. Microb Pathog 2020; 149:104354. [DOI: 10.1016/j.micpath.2020.104354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/30/2022]
|
24
|
Feng W, Wang Y, Chen S, Zhu X. Intra-tumoral heterogeneity and immune responses predicts prognosis of gastric cancer. Aging (Albany NY) 2020; 12:24333-24344. [PMID: 33259333 PMCID: PMC7762511 DOI: 10.18632/aging.202238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
Chemotherapy resistance eventually develops in patients with gastric cancer (GC). Intra-tumoral heterogeneity (ITH) refers to the intercellular genetic variations and phenotypic diversity that affect responses to drug therapy. We measured ITH using mutant-allele tumor heterogeneity (MATH) derived from whole-exome sequencing data of patients with GC in The Cancer Genome Atlas (TCGA) database. The study included 385 patients from the TCGA database with available data regarding gastrectomy, survival, and whole-exome sequencing. Further analysis was performed in 171 GC patients with available data regarding adjuvant chemotherapy. Multiple factor analysis showed that MATH was an independent predictor of OS (hazard ratio [HR], 1.432; 95% confidence interval [CI], 1.073-1.913; P = 0.015) in patients with GC. Moreover, MATH was also an independent predictor of OS among the 171 GC patients who received adjuvant chemotherapy (HR, 2.016; 95% CI, 1.236-3.289; P = 0.005). Pathway enrichment and immune cell analyses revealed significantly higher infiltration by 20 types of immune cells in the low/intermediate group, compared to the group with high MATH scores. In conclusion, low/intermediate MATH scores predicted longer OS, when compared to those with high MATH scores. The immune response was obviously upregulated in patients with GC and low/intermediate MATH scores.
Collapse
Affiliation(s)
- Wanjing Feng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People’s Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People’s Republic of China
| | - Yue Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, People’s Republic of China
| | - Siyuan Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People’s Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People’s Republic of China
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, People’s Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People’s Republic of China
| |
Collapse
|
25
|
Jiang MC, Hu CC, Hsu WL, Hsu TL, Lin NS, Hsu YH. Fusion of a Novel Native Signal Peptide Enhanced the Secretion and Solubility of Bioactive Human Interferon Gamma Glycoproteins in Nicotiana benthamiana Using the Bamboo Mosaic Virus-Based Expression System. FRONTIERS IN PLANT SCIENCE 2020; 11:594758. [PMID: 33281853 PMCID: PMC7688984 DOI: 10.3389/fpls.2020.594758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/23/2020] [Indexed: 05/31/2023]
Abstract
Plant viruses may serve as expression vectors for the efficient production of pharmaceutical proteins in plants. However, the downstream processing and post-translational modifications of the target proteins remain the major challenges. We have previously developed an expression system derived from Bamboo mosaic virus (BaMV), designated pKB19, and demonstrated its applicability for the production of human mature interferon gamma (mIFNγ) in Nicotiana benthamiana. In this study, we aimed to enhance the yields of soluble and secreted mIFNγ through the incorporation of various plant-derived signal peptides. Furthermore, we analyzed the glycosylation patterns and the biological activity of the mIFNγ expressed by the improved pKB19 expression system in N. benthamiana. The results revealed that the fusion of a native N. benthamiana extensin secretory signal (SSExt) to the N-terminal of mIFNγ (designated SSExt mIFNγ) led to the highest accumulation level of protein in intracellular (IC) or apoplast washing fluid (AWF) fractions of N. benthamiana leaf tissues. The addition of 10 units of 'Ser-Pro' motifs of hydroxyproline-O-glycosylated peptides (HypGPs) at the C-terminal end of SSExt mIFNγ (designated SSExt mIFNγ(SP)10) increased the solubility to nearly 2.7- and 1.5-fold higher than those of mIFNγ and SSExt mIFNγ, respectively. The purified soluble SSExt mIFNγ(SP)10 protein was glycosylated with abundant complex-type N-glycan attached to residues N56 and N128, and exhibited biological activity against Sindbis virus and Influenza virus replication in human cell culture systems. In addition, suspension cell cultures were established from transgenic N. benthamiana, which produced secreted SSExt mIFNγ(SP)10 protein feasible for downstream processing. These results demonstrate the applicability of the BaMV-based vector systems as a useful alternative for the production of therapeutic proteins, through the incorporation of appropriate fusion tags.
Collapse
Affiliation(s)
- Min-Chao Jiang
- Ph.D. Program in Microbial Genomics, National Chung Hsing University and Academia Sinica, Taichung, Taiwan
| | - Chung-Chi Hu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Na-Sheng Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Yau-Heiu Hsu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
26
|
Quintana ME, Cardoso NP, Pereyra R, Barone LJ, Barrionuevo FM, Mansilla FC, Turco CS, Capozzo AV. Interferon lambda protects cattle against bovine viral diarrhea virus infection. Vet Immunol Immunopathol 2020; 230:110145. [PMID: 33160262 DOI: 10.1016/j.vetimm.2020.110145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 01/15/2023]
Abstract
Interferon lambda (IFN-λ) plays an important role in inducing an antiviral state in mucosal surfaces and has been used as an effective biotherapeutic against several viral diseases. Here we performed a proof of concept study on the activity of a biologically active recombinant bovine IFN-λ (rIFN-λ) produced in eukaryotic cells against Bovine Viral Diarrhea Virus (BVDV) in cattle. We first confirmed the lack of toxicity of different concentrations of rIFN-λ in bovine peripheral blood cells and the safety of its subcutaneous application in calves in doses up to 12 IU/kg. The antiviral activity of the rIFN-λ against BVDV was assessed in calves that were inoculated with 6 IU/kg of rIFN-λ (n = 4) or mock-treated (n = 2) two days before and after challenge with a BVDV type-2 non-cytopathic strain. Mock-treated animals developed respiratory disease, shedded the virus from 4 to 7 days post-infection (dpi) and had viremia between 4 and 14 dpi. Conversely, calves treated with rIFN-λ did not develop clinical symptoms. The virus was not found in nasal secretions or sera. Only one animal had a positive viral RNA detection in serum at 7 dpi. All infected animals treated with rIFN-λ increased systemic type-I IFNs levels at 4 dpi. The antiviral treatment induced an earlier onset of the anti-BVDV neutralizing antibodies. Altogether, these results constitute the proof-of-principle of bovine IFN-λ as an antiviral biotherapeutic to protect cattle against the clinical disease caused by BVDV.
Collapse
Affiliation(s)
- María Eugenia Quintana
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Nancy Patricia Cardoso
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Rodrigo Pereyra
- Área de Investigación en Sanidad Animal IIACS-CIAP- Estación Experimental Agropecuaria EEA, INTA Cerrillos, Salta, Argentina
| | - Lucas José Barone
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Florencia Mariel Barrionuevo
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Florencia Celeste Mansilla
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Cecilia Soledad Turco
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina
| | - Alejandra Victoria Capozzo
- Instituto de Virología e Innovaciones Tecnológicas. IVIT, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Instituto Nacional de Tecnología Agropecuaria (INTA). Nicolás Repetto y Los Reseros s/n. Hurlingham, Buenos Aires, Argentina.
| |
Collapse
|
27
|
Wang X, Ashhurst AS, Dowman LJ, Watson EE, Li HY, Fairbanks AJ, Larance M, Kwan A, Payne RJ. Total Synthesis of Glycosylated Human Interferon-γ. Org Lett 2020; 22:6863-6867. [PMID: 32830985 DOI: 10.1021/acs.orglett.0c02401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Interferon-γ (IFN-γ) is a glycoprotein that is responsible for orchestrating numerous critical immune induction and modulation processes and is used clinically for the treatment of a number of diseases. Herein, we describe the total chemical synthesis of homogeneously glycosylated variants of human IFN-γ using a tandem diselenide-selenoester ligation-deselenization strategy in the C- to N-terminal direction. The synthetic glycoproteins were successfully folded, and the structures and antiviral functions were assessed.
Collapse
Affiliation(s)
- Xiaoyi Wang
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Anneliese S Ashhurst
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Luke J Dowman
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Emma E Watson
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Henry Y Li
- School of Physical and Chemical Sciences, The University of Canterbury, Christchurch 8140, New Zealand
| | - Antony J Fairbanks
- School of Physical and Chemical Sciences, The University of Canterbury, Christchurch 8140, New Zealand
| | - Mark Larance
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ann Kwan
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, https://cipps.org.au/
| |
Collapse
|
28
|
Elyasi A, Voloshyna I, Ahmed S, Kasselman LJ, Behbodikhah J, De Leon J, Reiss AB. The role of interferon-γ in cardiovascular disease: an update. Inflamm Res 2020; 69:975-988. [PMID: 32699989 DOI: 10.1007/s00011-020-01382-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is the leading cause of death, globally, and its prevalence is only expected to rise due to the increasing incidence of co-morbidities such as obesity and diabetes. Medical treatment of CVD is directed primarily at slowing or reversing the underlying atherosclerotic process by managing circulating lipids with an emphasis on control of low-density lipoprotein (LDL) cholesterol. However, over the past several decades, there has been increasing recognition that chronic inflammation and immune system activation are important contributors to atherosclerosis. This shift in focus has led to the elucidation of the complex interplay between cholesterol and cellular secretion of cytokines involved in CVD pathogenesis. Of the vast array of cytokine promoting atherosclerosis, interferon (IFN)-γ is highly implicated and, therefore, of great interest. METHODS Literature review was performed to further understand the effect of IFN-γ on the development of atherosclerotic CVD. RESULTS IFN-γ, the sole member of the type II IFN family, is produced by T cells and macrophages, and has been found to induce production of other cytokines and to have multiple effects on all stages of atherogenesis. IFN-γ activates a variety of signaling pathways, most commonly the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, to induce oxidative stress, promote foam cell accumulation, stimulate smooth muscle cell proliferation and migration into the arterial intima, enhance platelet-derived growth factor expression, and destabilize plaque. These are just a few of the contributions of IFN-γ to the initiation and progression of atherosclerotic CVD. CONCLUSION Given the pivotal role of IFN-γ in the advancement of CVD, activation of its signaling pathways is being explored as a driver of atherosclerosis. Manipulation of this key cytokine may lead to novel therapeutic avenues for CVD prevention and treatment. A number of therapies are being explored with IFN-γ as the potential target.
Collapse
Affiliation(s)
- Ailin Elyasi
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Iryna Voloshyna
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Saba Ahmed
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Lora J Kasselman
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Jennifer Behbodikhah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Allison B Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| |
Collapse
|
29
|
Towards a new avenue for producing therapeutic proteins: Microalgae as a tempting green biofactory. Biotechnol Adv 2020; 40:107499. [DOI: 10.1016/j.biotechadv.2019.107499] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/02/2019] [Accepted: 12/17/2019] [Indexed: 02/08/2023]
|
30
|
Yang X, Arslan M, Liu X, Song H, Du M, Li Y, Zhang Z. IFN-γ establishes interferon-stimulated gene-mediated antiviral state against Newcastle disease virus in chicken fibroblasts. Acta Biochim Biophys Sin (Shanghai) 2020; 52:268-280. [PMID: 32047904 PMCID: PMC7109688 DOI: 10.1093/abbs/gmz158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/24/2022] Open
Abstract
Newcastle disease virus (NDV) causes severe economic losses through severe morbidity and mortality and poses a significant threat to the global poultry industry. Significant efforts have been made to develop novel vaccines and therapeutics; however, the interaction of NDV with the host is not yet fully understood. Interferons (IFNs), an integral component of innate immune signaling, act as the first line of defense against invading viruses. Compared with the mammalian repertoire of IFNs, limited information is available on the antiviral potential of IFNs in chickens. Here, we expressed chicken IFN-γ (chIFN-γ) using a baculovirus expression vector system, characterized its antiviral potential against NDV, and determined its antiviral potential. Priming of chicken embryo fibroblasts with chIFN-γ elicited an antiviral environment in primary cells, which was mainly due to interferon-stimulated genes (ISGs). A genome-wide transcriptomics approach was used to elucidate the possible signaling pathways associated with IFN-γ-induced immune responses. RNA-sequencing (RNA-seq) data revealed significant induction of ISG-associated pathways, activated temporal expression of ISGs, antiviral mediators, and transcriptional regulators in a cascade of antiviral responses. Collectively, we found that IFN-γ significantly elicited an antiviral response against NDV infection. These data provide a foundation for chIFN-γ-mediated antiviral responses and underpin functional annotation of these important chIFN-γ-induced antiviral influencers.
Collapse
Affiliation(s)
- Xin Yang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Mehboob Arslan
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Haozhi Song
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Mengtan Du
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
31
|
Development of reporter gene assays to determine the bioactivity of biopharmaceuticals. Biotechnol Adv 2020; 39:107466. [DOI: 10.1016/j.biotechadv.2019.107466] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/02/2019] [Accepted: 11/02/2019] [Indexed: 02/06/2023]
|
32
|
Donde R, Gupta MK, Gouda G, Dash SK, Behera L, Vadde R. Immune Cell Therapy Against Gastrointestinal Tract Cancers. IMMUNOTHERAPY FOR GASTROINTESTINAL MALIGNANCIES 2020:61-77. [DOI: 10.1007/978-981-15-6487-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
33
|
Cui F, Qu D, Sun R, Zhang M, Nan K. NK cell-produced IFN-γ regulates cell growth and apoptosis of colorectal cancer by regulating IL-15. Exp Ther Med 2019; 19:1400-1406. [PMID: 32010315 DOI: 10.3892/etm.2019.8343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/13/2019] [Indexed: 12/24/2022] Open
Abstract
Globally, colorectal cancer (CC) is the third leading cause of mortality associated with cancer. Natural killer (NK) cells are a major class of cells that are responsible for eliminating tumor cells and cytokine production. NK cell-mediated production of interferon gamma (IFN-γ) has antiviral, immunoregulatory and anti-tumor properties. IL-15 is important in linking inflammation with cancer. For instance, IL-15 promotes humoral and cell-mediated immune responses to inhibit tumor growth. IL-15 inhibits colitis-associated colon carcinogenesis by inducing antitumor immunity. However, the effect of NK cell-mediated IFN-γ on IL-15 expression in CC progression remains unknown. mRNA and protein level were detected using reverse transcription-quantitative PCR and western blotting, respectively. IFN-γ concentrations were detected using ELISAs. The cytotoxicity of NK-92 cells on SW480 cells was detected using cytoTox 96® non-radioactive cytotoxicity assays. Cell apoptosis and cell proliferation was detected using flow cytometry and CCK-8 assays, respectively. IL-2 was used for NK-92 stimulation, IL-15 antibodies were used to neutralize IL-15 bioactivity. For the present study, 21 patients with CC and 21 healthy volunteers were enrolled at the First Affiliated Hospital of Xi'an Jiaotong University. IL-15 mRNA and protein expression were significantly lower in NK cells isolated from the CC group compared with healthy volunteer group. IL-2 enhanced the production/secretion of IFN-γ in addition to enhancing NK-92 cell-mediated killing of SW480 cells. Compared with the control group, NK-92 cells treated with IL-2 alone significantly increased cell apoptosis, BAX expression levels as well as phosphorylated (p)-Janus kinase 2 and p-STAT1 protein levels, whilst reducing cell viability and Bcl-2 protein levels in SW480 cells. These observations were not made when treated with IL-2 and polyclonal antibody (pAb) targeting IL-15. Taken together, NK cell-mediated IFN-γ served a pivotal role in CC by regulating IL-15. The effects of IL-2 induced IFN-γ were abolished by pAb IL-15 treatment. The mechanisms of action behind how IFN-γ regulates IL-2 is unclear, and is a promising area for future research.
Collapse
Affiliation(s)
- Feng Cui
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China.,Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| | - Di Qu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ruya Sun
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Mingming Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Kejun Nan
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shannxi 710061, P.R. China
| |
Collapse
|
34
|
Prabhu AA, Kumar JP, Mandal BB, Veeranki VD. Glucose-methanol-based fed-batch fermentation for the production of recombinant human interferon gamma (rhIFN-γ) and evaluation of its antitumor potential. Biotechnol Appl Biochem 2019; 67:973-982. [PMID: 31811672 DOI: 10.1002/bab.1868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/02/2019] [Indexed: 11/09/2022]
Abstract
Squamous cell carcinoma (SCC) is nonmelanoma skin cancer, which is very common in patients having T-cell immunosuppressant drugs. Anticancerous agents such as cytokines showed effective response on SCC. Human interferon-gamma (hIFN-γ), a type II cytokines, are having potent antiproliferative and immunomodulatory effects. In the current study, the fed-batch cultivation of recombinant Pichia pastoris was carried out, and its effect on cell biomass production, recombinant human interferon-gamma (rhIFN-γ) production, and the overflow metabolites was estimated. P. pastoris GS115 strain coexpressed with 6-phosphogluconolactonase (SOL3) and ribulose-phosphate 3-epimerase (RPE1) gene (GS115/rhIFN-γ/SR) resulted in 60 mg L-1 of rhIFN-γ production, which was twofold higher as compared with the production from GS115/rhIFN-γ strain. The antiproliferative potential of rhIFN-γ was examined on the human squamous carcinoma (A431) cell lines. Cells treated with 80 ng mL-1 of rhIFN-γ exhibited 50% growth inhibition by enhancing the production of intracellular reactive oxygen species levels and disrupting membrane integrity. Our findings highlight a state of art process development strategy for the high-level production of rhIFN-γ and its potential application as a therapeutic drug in SCC therapy.
Collapse
Affiliation(s)
- Ashish A Prabhu
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| | - Jadi Praveen Kumar
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| | - Biman B Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Venkata Dasu Veeranki
- Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, India
| |
Collapse
|
35
|
Payen D, Faivre V, Miatello J, Leentjens J, Brumpt C, Tissières P, Dupuis C, Pickkers P, Lukaszewicz AC. Multicentric experience with interferon gamma therapy in sepsis induced immunosuppression. A case series. BMC Infect Dis 2019; 19:931. [PMID: 31690258 PMCID: PMC6833157 DOI: 10.1186/s12879-019-4526-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
Background The sepsis-induced immunodepression contributes to impaired clinical outcomes of various stress conditions. This syndrome is well documented and characterized by attenuated function of innate and adaptive immune cells. Several pharmacological interventions aimed to restore the immune response are emerging of which interferon-gamma (IFNγ) is one. It is of paramount relevance to obtain clinical information on optimal timing of the IFNγ-treatment, −tolerance, −effectiveness and outcome before performing a RCT. We describe the effects of IFNγ in a cohort of 18 adult and 2 pediatric sepsis patients. Methods In this open-label prospective multi-center case-series, IFNγ treatment was initiated in patients selected on clinical and immunological criteria early (< 4 days) or late (> 7 days) following the onset of sepsis. The data collected in 18 adults and 2 liver transplanted pediatric patients were: clinical scores, monocyte expression of HLA-DR (flow cytometry), lymphocyte immune-phenotyping (flow cytometry), IL-6 and IL-10 plasma levels (ELISA), bacterial cultures, disease severity, and mortality. Results In 15 out of 18 patients IFNγ treatment was associated with an increase of median HLA-DR expression from 2666 [IQ 1547; 4991] to 12,451 [IQ 4166; 19,707], while the absolute number of lymphocyte subpopulations were not affected, except for the decrease number of NK cells 94.5 [23; 136] to 32.5 [13; 90.8] (0.0625)]. Plasma levels of IL-6 464 [201–770] to 108 (89–140) ng/mL (p = 0.04) and IL-10 from IL-10 from 29 [12–59] to 9 [1–15] pg/mL decreased significantly. Three patients who received IFNγ early after ICU admission (<4 days) died. The other patients had a rapid clinical improvement assessed by the SOFA score and bacterial cultures that were repeatedly positive became negative. The 2 pediatric cases improved rapidly, but 1 died for hemorrhagic complication. Conclusion Guided by clinical and immunological monitoring, adjunctive immunotherapy with IFNγ appears well-tolerated in our cases and improves immune host defense in sepsis induced immuno suppression. Randomized clinical studies to assess its potential clinical benefit are warranted.
Collapse
Affiliation(s)
- Didier Payen
- Groupe Hospitalier Saint-Louis Lariboisière, AP-HP, Université Paris 7 Denis Diderot, 2 rue Ambroise Paré, 75010, Paris, France. .,UMR INSERM 1160 University Paris 7 Denis Diderot, Paris, France.
| | - Valerie Faivre
- Groupe Hospitalier Saint-Louis Lariboisière, AP-HP, Université Paris 7 Denis Diderot, 2 rue Ambroise Paré, 75010, Paris, France.,UMR INSERM 1160 University Paris 7 Denis Diderot, Paris, France
| | - Jordi Miatello
- Pediatric Intensive Care and Neonatal Medicine, Bicêtre Hospital, AP-HP, Le Kremlin-Bicêtre, France.,Institute of Integrative Biology of Cell, CNRS, CEA, Univ. Paris Sud, Paris Saclay University, Gif sur Yvette, France
| | - Jenneke Leentjens
- Departments of intensive care and internal medicine, Radboud university medical center Nijmegen, PO box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Caren Brumpt
- Service d'Hématologie Biologique, Pôle B2P, Hôpital Lariboisière, APHP, Paris, France
| | - Pierre Tissières
- Pediatric Intensive Care and Neonatal Medicine, Bicêtre Hospital, AP-HP, Le Kremlin-Bicêtre, France.,Institute of Integrative Biology of Cell, CNRS, CEA, Univ. Paris Sud, Paris Saclay University, Gif sur Yvette, France
| | - Claire Dupuis
- Groupe Hospitalier Saint-Louis Lariboisière, AP-HP, Université Paris 7 Denis Diderot, 2 rue Ambroise Paré, 75010, Paris, France
| | - Peter Pickkers
- Department Intensive Care Medicine, Radboud university medical center Nijmegen, PO box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Anne Claire Lukaszewicz
- Groupe Hospitalier Saint-Louis Lariboisière, AP-HP, Université Paris 7 Denis Diderot, 2 rue Ambroise Paré, 75010, Paris, France.,UMR INSERM 1160 University Paris 7 Denis Diderot, Paris, France
| |
Collapse
|
36
|
Negative regulators of cell death pathways in cancer: perspective on biomarkers and targeted therapies. Apoptosis 2019; 23:93-112. [PMID: 29322476 DOI: 10.1007/s10495-018-1440-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cancer is a primary cause of human fatality and conventional cancer therapies, e.g., chemotherapy, are often associated with adverse side-effects, tumor drug-resistance, and recurrence. Molecularly targeted therapy, composed of small-molecule inhibitors and immunotherapy (e.g., monoclonal antibody and cancer vaccines), is a less harmful alternative being more effective against cancer cells whilst preserving healthy tissues. Drug-resistance, however, caused by negative regulation of cell death signaling pathways, is still a challenge. Circumvention of negative regulators of cell death pathways or development of predictive and response biomarkers is, therefore, quintessential. This review critically discusses the current state of knowledge on targeting negative regulators of cell death signaling pathways including apoptosis, ferroptosis, necroptosis, autophagy, and anoikis and evaluates the recent advances in clinical and preclinical research on biomarkers of negative regulators. It aims to provide a comprehensive platform for designing efficacious polytherapies including novel agents for restoring cell death signaling pathways or targeting alternative resistance pathways to improve the chances for antitumor responses. Overall, it is concluded that nonapoptotic cell death pathways are a potential research arena for drug discovery, development of novel biomarkers and targeted therapies.
Collapse
|
37
|
Abstract
Three decades of research in hematopoietic stem cell transplantation and HIV/AIDS fields have shaped a picture of immune restoration disorders. This manuscript overviews the molecular biology of interferon networks, the molecular pathogenesis of immune reconstitution inflammatory syndrome, and post-hematopoietic stem cell transplantation immune restoration disorders (IRD). It also summarizes the effects of thymic involution on T cell diversity, and the results of the assessment of diagnostic biomarkers of IRD, and tested targeted immunomodulatory treatments.
Collapse
Affiliation(s)
- Hesham Mohei
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Usha Kellampalli
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
38
|
Castro F, Pinto ML, Almeida R, Pereira F, Silva AM, Pereira CL, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Chitosan/poly(γ-glutamic acid) nanoparticles incorporating IFN-γ for immune response modulation in the context of colorectal cancer. Biomater Sci 2019; 7:3386-3403. [PMID: 31233057 DOI: 10.1039/c9bm00393b] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IFN-γ therapy has been approved by the Food and Drug Administration (FDA) for the treatment of chronic granulomatous disease and severe malignant osteopetrosis. Despite the promising IFN-γ-based therapeutic applications, its limited success in clinical trials is related with limitations inherent to its molecular properties and with the difficulties to deliver it locally or with adequate periodicity to achieve a therapeutic effect. We have previously shown that chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) are immunostimulatory, impairing colorectal cancer cell invasion. Ch is a biocompatible cationic polysaccharide extensively studied and already approved for biomedical applications while γ-PGA is a poly(amino acid), biodegradable and negatively charged. Here, we evaluated the potential of Ch/γ-PGA NPs as vehicles for IFN-γ and their ability to modulate immune cells' phenotype. In this study, Ch/IFN-γ/γ-PGA nanoparticles (IFN-γ-NPs) prepared by a co-acervation method, presenting a size of approximately 180 nm and a low polydispersity index, were tested for their immunomodulatory activity. These IFN-γ-NPs induced an immunostimulatory profile on dendritic cells (DCs) with increased cell surface costimulatory molecules and secretion of pro-inflammatory cytokines, including IL-6, IL-12p40 and TNF-α. IFN-γ-NPs also modulated the IL-10-stimulated macrophage profile, increasing their ability to secrete the pro-inflammatory cytokines IL-6, IL-12p40 and TNF-α. Concomitantly, these phenotypic alterations enhanced T cell proliferation. In addition, the ability of DCs and macrophages to induce colorectal cancer cell invasion was hampered in the presence of IFN-γ-NPs. Although the major observations were mediated by Ch/γ-PGA NPs, the incorporation of IFN-γ into NPs potentiated the expression of CD40 and CD86, and the impairment of colorectal cancer cell invasion. This work bridges the previously reported immunostimulatory capacity of Ch/γ-PGA NPs with their potential as carriers for immunomodulatory molecules, like IFN-γ, opening new avenues for their use in clinical settings.
Collapse
Affiliation(s)
- Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Marta L Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CNC - Centro de Neurociências e Biologia Celular, Universidade de Coimbra, Portugal
| | - Rui Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Flávia Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and CBMA - Centro de Biologia Molecular e Ambiental, Universidade do Minho, Braga, Portugal
| | - Andreia M Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina L Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. and Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Jiang MC, Hu CC, Lin NS, Hsu YH. Production of Human IFNγ Protein in Nicotiana benthamiana Plant through an Enhanced Expression System Based on Bamboo mosaic Virus. Viruses 2019; 11:E509. [PMID: 31163694 PMCID: PMC6630494 DOI: 10.3390/v11060509] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 02/02/2023] Open
Abstract
Plant-based systems are safe alternatives to the current platforms for the production of biologically active therapeutic proteins. However, plant-based expression systems face certain major challenges, including the relatively low productivity and the generation of target proteins in biologically active forms. The use of plant virus-based expression systems has been shown to enhance yields, but further improvement is still required to lower the production cost. In this study, various strategies were employed to increase the yields of an important therapeutic protein, human interferon gamma (IFNγ), in Nicotiana benthamiana through modifications of expression vectors based on potexviruses. Among these, the vector based on a coat protein (CP)-deficient Bamboo mosaic virus (BaMV), pKB△CHis, was shown to exhibit the highest expression level for the unmodified IFNγ. Truncation of the N-terminal signal peptide of IFN (designated mIFNγ) resulted in a nearly seven-fold increase in yield. Co-expression of a silencing suppressor protein by replacing the coding sequence of BaMV movement protein with that of P19 led to a 40% increase in mIFNγ accumulation. The fusion of endoplasmic reticulum (ER) retention signal with mIFNγ significantly enhanced the accumulation ratio of biologically active dimeric mIFNγ to 87% relative to the non-active monomeric form. The construct pKB19mIFNγER, employing the combination of all the above enhancement strategies, gave the highest level of protein accumulation, up to 119 ± 0.8 μg/g fresh weight, accounting for 2.5% of total soluble protein (TSP) content. These findings advocate the application of the modified BaMV-based vector as a platform for high-level expression of therapeutic protein in N. benthamiana.
Collapse
Affiliation(s)
- Min-Chao Jiang
- Ph.D Program in Microbial Genomic, National Chung Hsing University and Academia Sinica, Taichung 40227, Taiwan.
| | - Chung-Chi Hu
- Graduate Institute of Biotechnology, Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Na-Sheng Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan.
| | - Yau-Heiu Hsu
- Graduate Institute of Biotechnology, Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
40
|
Morales J, Pawle RH, Akkilic N, Luo Y, Xavierselvan M, Albokhari R, Calderon IAC, Selfridge S, Minns R, Takiff L, Mallidi S, Clark HA. DNA-Based Photoacoustic Nanosensor for Interferon Gamma Detection. ACS Sens 2019; 4:1313-1322. [PMID: 30973005 DOI: 10.1021/acssensors.9b00209] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tracking protein levels in the body is vital in both research and medicine, where understanding their physiological roles provides insight into their regulation in homeostasis and diseases. In medicine, protein levels are actively sampled since they continuously fluctuate, reflecting the status of biological systems and provide insight into patient health. One such protein is interferon gamma, a clinically relevant protein with immunoregulatory functions that play critical roles against infection. New tools for continuously monitoring protein levels in vivo are invaluable in monitoring real-time conditions of patients to allow better care. Here, we developed a DNA-based nanosensor for the photoacoustic detection of interferon gamma. This work demonstrates how we transformed a simple DNA motif, receptors, and a novel phthalocyanine dye into a proof-of-concept photoacoustic nanosensor for protein detection. Surface plasmon resonance kinetic analysis demonstrated that the nanosensor is responsive and reversible to interferon gamma with an affinity in the nanomolar range, KD1 = 167 nM and KD2 = 316 nM. As a reporter, our design includes a novel phthalocyanine-based photoacoustic dye that stacks in a J-aggregate, causing a 22.5% increase in signal. Upon receptor binding, the DNA structure bends to induce phthalocyanine dye stacking, resulting in a 55% increase in photoacoustic signal in the presence of 10 μM interferon gamma. This proof-of-concept nanosensor is a novel approach to the development of a photoacoustic sensor and may be adapted for other proteins of interest in the future for in vivo tracking.
Collapse
Affiliation(s)
- Jennifer Morales
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States,
| | - Robert H. Pawle
- Akita Innovations LLC, Billerica, Massachusetts 01862, United States,
| | - Namik Akkilic
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States,
| | - Yi Luo
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States,
| | - Marvin Xavierselvan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States,
| | - Rayan Albokhari
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States,
| | - Isen Andrew C. Calderon
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States,
| | - Scott Selfridge
- Akita Innovations LLC, Billerica, Massachusetts 01862, United States,
| | - Richard Minns
- Akita Innovations LLC, Billerica, Massachusetts 01862, United States,
| | - Larry Takiff
- Akita Innovations LLC, Billerica, Massachusetts 01862, United States,
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States,
| | - Heather A. Clark
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, United States,
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
41
|
Interferon gamma induces inflammatory responses through the interaction of CEACAM1 and PI3K in airway epithelial cells. J Transl Med 2019; 17:147. [PMID: 31072323 PMCID: PMC6507156 DOI: 10.1186/s12967-019-1894-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/25/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Interferon gamma (IFNγ) plays an important role in the development of chronic lung diseases via the production of inflammatory mediators, although the exact mechanism remains unclear. The present study aimed at investigating the potential mechanisms by which IFNγ induced over-production of interleukins through the interaction between carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) pathway. METHODS IFN-γ induced over-production of interleukin (IL) 6 and IL8, and RNA expression of CEACAM1 and its subtypes or PI3K and its subtypes in human bronchial epithelial cells (HBE). The production of IL6 and IL8 or cell proliferation and movement were also evaluated in cellCEACAM1- or cellCEACAM1+ after the induction of IFN-γ. Roles of PI3K subtype proteins, e.g. PI3Kp110α/δ, Akt, p110α/γ/δ/β/mTOR, PI3Kp110α/δ/β, PI3Kp110δ, or pan-PI3K in IFN-γ-induced CEACAM1 subtype alterations were furthermore validated using those proteins of PI3K subtypes. RESULTS CEACAM1, especially CEACAM1-S isoforms, was significantly up-regulated in HBE cells after treatment with IFN-γ. CEACAM1 played roles in expression of IL-6 and IL-8, and facilitated cellular proliferation and migration. IFN-γ up-regulated the expression of CEACAM1 in airway epithelial cells, especially CEACAM1-S isoforms, promoting cellular proliferation, migration, and the production of inflammatory factors. PI3K (p110δ)/Akt/mTOR pathway was involved in the process of IFN-γ-upregulated CEACAM1, especially CEACAM1-S. On the other hand, CEACAM1 could promote the activation of PI3K/Akt/mTOR pathway. CONCLUSION IFN-γ could induce inflammatory responses, cellular growth and proliferation through the interaction of CEACAM1 (especially CEACAM1-S isoforms) and PI3K(p110δ)/Akt/mTOR in airway epithelial cells, which might be new alternative of future therapies against epithelial transition from inflammation to cancer.
Collapse
|
42
|
Luo Q, Zhang L, Luo C, Jiang M. Emerging strategies in cancer therapy combining chemotherapy with immunotherapy. Cancer Lett 2019; 454:191-203. [PMID: 30998963 DOI: 10.1016/j.canlet.2019.04.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy holds great potential to battle cancer by exerting a durable immunity effect. However, this process might be limited by various constraints existing in the tumor microenvironment (TME), such as the lack of available neoantigen, insufficient T cells from the naive repertoire, or immunosuppressive networks in which immunogenic tissue is protected from immune attacks. Certain chemotherapeutic drugs could elicit immune-potentiating effects by either inducing immunogenicity or relieving tumor-induced immunosuppression. Some also leave tumors directly susceptible to cytotoxic T cell attacks. Mounting evidence accumulated from preclinical and clinical studies suggests that these two treatment modalities might be mutually reinforcing as an effective "chemo-immunotherapy" strategy. Herein, we reviewed the latest advances in cancer immunotherapy and related mechanisms involved in chemotherapeutic-mediated immune activation. The emerging combination strategies and synergistic effects in response to chemo-immunotherapy are highlighted. We also discuss the challenges and critical considerations in its future development.
Collapse
Affiliation(s)
- Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, 155 Nanjing South Street, Shenyang, Liaoning Province, 110016, PR China; Department of Pharmacy, China Medical University, 155 Nanjing South Street, Shenyang, Liaoning Province, 110016, PR China.
| | - Ling Zhang
- Department of Biotherapy, Cancer Research Institute, The First Affiliated Hospital of China Medical University, 155 Nanjing South Street, Shenyang, Liaoning Province, 110016, PR China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, PR China
| | - Mingyan Jiang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, 155 Nanjing South Street, Shenyang, Liaoning Province, 110016, PR China; Department of Pharmacy, China Medical University, 155 Nanjing South Street, Shenyang, Liaoning Province, 110016, PR China
| |
Collapse
|
43
|
Unni S, Prabhu AA, Pandey R, Hande R, Veeranki VD. Artificial neural network‐genetic algorithm (ANN‐GA) based medium optimization for the production of human interferon gamma (hIFN‐γ) inKluyveromyces lactiscell factory. CAN J CHEM ENG 2019. [DOI: 10.1002/cjce.23350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Silpa Unni
- Biochemical Engineering LaboratoryDepartment of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahati 781039AssamIndia
| | - Ashish A. Prabhu
- Biochemical Engineering LaboratoryDepartment of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahati 781039AssamIndia
| | - Rajat Pandey
- Biochemical Engineering LaboratoryDepartment of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahati 781039AssamIndia
| | - Rohit Hande
- Biochemical Engineering LaboratoryDepartment of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahati 781039AssamIndia
| | - Venkata Dasu Veeranki
- Biochemical Engineering LaboratoryDepartment of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahati 781039AssamIndia
| |
Collapse
|
44
|
Alqahtani SA, Sulkowski MS. The Role of Interferon for the Treatment of Chronic Hepatitis C Virus Infection. TOPICS IN MEDICINAL CHEMISTRY 2019:97-113. [DOI: 10.1007/7355_2018_59] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
45
|
Kontoravdi C, Jimenez del Val I. Computational tools for predicting and controlling the glycosylation of biopharmaceuticals. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.08.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Castilho A, Beihammer G, Pfeiffer C, Göritzer K, Montero‐Morales L, Vavra U, Maresch D, Grünwald‐Gruber C, Altmann F, Steinkellner H, Strasser R. An oligosaccharyltransferase from Leishmania major increases the N-glycan occupancy on recombinant glycoproteins produced in Nicotiana benthamiana. PLANT BIOTECHNOLOGY JOURNAL 2018; 16:1700-1709. [PMID: 29479800 PMCID: PMC6131413 DOI: 10.1111/pbi.12906] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/15/2017] [Accepted: 02/06/2018] [Indexed: 05/19/2023]
Abstract
N-glycosylation is critical for recombinant glycoprotein production as it influences the heterogeneity of products and affects their biological function. In most eukaryotes, the oligosaccharyltransferase is the central-protein complex facilitating the N-glycosylation of proteins in the lumen of the endoplasmic reticulum (ER). Not all potential N-glycosylation sites are recognized in vivo and the site occupancy can vary in different expression systems, resulting in underglycosylation of recombinant glycoproteins. To overcome this limitation in plants, we expressed LmSTT3D, a single-subunit oligosaccharyltransferase from the protozoan Leishmania major transiently in Nicotiana benthamiana, a well-established production platform for recombinant proteins. A fluorescent protein-tagged LmSTT3D variant was predominately found in the ER and co-located with plant oligosaccharyltransferase subunits. Co-expression of LmSTT3D with immunoglobulins and other recombinant human glycoproteins resulted in a substantially increased N-glycosylation site occupancy on all N-glycosylation sites except those that were already more than 90% occupied. Our results show that the heterologous expression of LmSTT3D is a versatile tool to increase N-glycosylation efficiency in plants.
Collapse
Affiliation(s)
- Alexandra Castilho
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Gernot Beihammer
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Christina Pfeiffer
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Kathrin Göritzer
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Laura Montero‐Morales
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Ulrike Vavra
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Daniel Maresch
- Department of ChemistryUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Friedrich Altmann
- Department of ChemistryUniversity of Natural Resources and Life SciencesViennaAustria
| | - Herta Steinkellner
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Richard Strasser
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
47
|
Dang W, Xu L, Yin Y, Chen S, Wang W, Hakim MS, Chang KO, Peppelenbosch MP, Pan Q. IRF-1, RIG-I and MDA5 display potent antiviral activities against norovirus coordinately induced by different types of interferons. Antiviral Res 2018; 155:48-59. [PMID: 29753657 DOI: 10.1016/j.antiviral.2018.05.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/06/2018] [Accepted: 05/08/2018] [Indexed: 11/19/2022]
Abstract
Norovirus represents the main cause of acute nonbacterial gastroenteritis worldwide. In immunocompromised patients, it bears high risk of causing chronic infection with significant morbidity and mortality. The lack of specific treatment prompts the development of anti-norovirus agents. In this study, we have investigated the role of interferon (IFN) response and evaluated antiviral activities of different IFNs against human norovirus (HuNoV) replication using a HuNoV replicon model. We found that HuNoV RNA replication was sensitive to all types of IFNs, including IFNα (type I), IFNγ (type II), IFNλ1 and 3 (type III). IFNs canonically induce interferon-stimulated genes (ISGs) to exert their antiviral activities. By profiling a subset of important human ISGs using an overexpression approach, we have identified RTP4 and HPSE as moderate anti-norovirus ISGs, whereas IRF-1, RIG-I (also known as DDX58) and MDA5 (also known as IFIH1) were identified as potent anti-norovirus effectors. Interestingly, type I and III IFNs coordinately induced IRF-1, RIG-I and MDA5; whereas type II IFN predominantly induced IRF-1 to exhibit their anti-norovirus activities. Combination of different IFNs revealed that IFNγ worked cooperatively with type I or type III IFNs to induce ISGs and subsequently inhibit HuNoV replication. Of note, replication of HuNoV did not interfere with antiviral IFN response. In summary, we showed the potent anti-norovirus activities of different types of IFNs and identified the key anti-norovirus effectors. These findings are important for understanding norovirus-host interactions and developing antiviral therapies.
Collapse
Affiliation(s)
- Wen Dang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Lei Xu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Yuebang Yin
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Sunrui Chen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Mohamad S Hakim
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, USA
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
48
|
Kumar N, Pandey R, Prabhu AA, Venkata Dasu V. Genetic and substrate-level modulation of Bacillus subtilis physiology for enhanced extracellular human interferon gamma production. Prep Biochem Biotechnol 2018; 48:391-401. [PMID: 29688129 DOI: 10.1080/10826068.2018.1446157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Human interferon-gamma (hIFNG) production is limited by various gene-level bottlenecks including translation, protein folding, and secretion which depends upon the physiological state of the organism. In this study gene-level and substrate-level modulations have been used to control Bacillus subtilis physiology for >15 fold extracellular soluble hIFNG production. Two variants of the native human interferon-gamma gene (hifng) were designed and synthesized, namely, cohifnghis and cohifng having codon adaptation index 25.33 and 26.89% higher than the native gene, respectively. BScoIFNG and BScoIFNGhis with ΔG of -100.0 and -113.7 kcal mol-1 resulted in 30 and 6.5% higher hIFNG compared to the native gene in complex medium. BScoIFNG produced 1.53 fold higher hIFNG using glucose-based defined medium as compared to the complex medium by modulating the physiological parameter growth rate from 0.35 to 0.26 hr-1. Further modulatory effect of various phosphotransferase transport system (PTS) and no-PTS sugars, sugar alcohols, and organic acids was quantified on the physiology of B. subtilis WB800N for extracellular hIFNG production. Sorbitol and glycerol emerged as the best hIFNG producers with lowest growth and substrate consumption rates. BScoIFNG produced maximum 3.15 mg L-1 hIFNG at 50 g L-1 glycerol with highest hIFNG yield (Yp/x = 0.136) and lowest substrate uptake rate (qs = 0.26).
Collapse
Affiliation(s)
- Nitin Kumar
- a Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Guwahati , Assam , India
| | - Rajat Pandey
- a Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Guwahati , Assam , India
| | - Ashish Anand Prabhu
- a Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Guwahati , Assam , India
| | - Veeranki Venkata Dasu
- a Biochemical Engineering Laboratory, Department of Biosciences and Bioengineering , Indian Institute of Technology Guwahati , Guwahati , Assam , India
| |
Collapse
|
49
|
Nikolic A. Pathophysiology and Genetics of Bronchiectasis Unrelated to Cystic Fibrosis. Lung 2018; 196:383-392. [PMID: 29754320 DOI: 10.1007/s00408-018-0121-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/04/2018] [Indexed: 10/16/2022]
Abstract
Bronchiectasis is characterized by deregulated inflammatory response and recurrent bacterial infection resulting in progressive lung damage and an irreversible dilatation of bronchi and bronchioles. Generally accepted model of the development of bronchiectasis is the "vicious cycle hypothesis" that proposes compromising of the mucociliary clearance by an initial event, which leads to the infection of the respiratory tract followed by further impairment of mucociliary function, bacterial proliferation, and more inflammation. Bronchiectasis is a very common symptom in patients with cystic fibrosis (CF), while bronchiectasis unrelated to CF is heterogeneous pathology of unknown cause with a large number of potential contributory factors and poorly understood pathogenesis. It is presumed that bronchiectasis unrelated to CF is a multifactorial condition predisposed by genetic factors. Different molecules have been implicated in the onset and development of idiopathic bronchiectasis, as well as modulation of the disease severity and response to therapy. Most of these molecules are involved in the processes that contribute to the homeostasis of the lung tissue, especially mucociliary clearance, protease-antiprotease balance, and immunomodulation. Evaluation of the studies performed towards investigation of the role these molecules play in bronchiectasis identifies genetic variants that may be of potential importance for clinical management of the disease, and also of interest for future research efforts. This review focuses on the molecules with major roles in lung homeostasis and their involvement in bronchiectasis unrelated to CF.
Collapse
Affiliation(s)
- Aleksandra Nikolic
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444A, PO Box 23, 11010, Belgrade, Serbia.
| |
Collapse
|
50
|
Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front Immunol 2018; 9:847. [PMID: 29780381 PMCID: PMC5945880 DOI: 10.3389/fimmu.2018.00847] [Citation(s) in RCA: 797] [Impact Index Per Article: 113.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
Interferon-gamma (IFN-γ) is a pleiotropic molecule with associated antiproliferative, pro-apoptotic and antitumor mechanisms. This effector cytokine, often considered as a major effector of immunity, has been used in the treatment of several diseases, despite its adverse effects. Although broad evidence implicating IFN-γ in tumor immune surveillance, IFN-γ-based therapies undergoing clinical trials have been of limited success. In fact, recent reports suggested that it may also play a protumorigenic role, namely, through IFN-γ signaling insensitivity, downregulation of major histocompatibility complexes, and upregulation of indoleamine 2,3-dioxygenase and of checkpoint inhibitors, as programmed cell-death ligand 1. However, the IFN-γ-mediated responses are still positively associated with patient's survival in several cancers. Consequently, major research efforts are required to understand the immune contexture in which IFN-γ induces its intricate and highly regulated effects in the tumor microenvironment. This review discusses the current knowledge on the pro- and antitumorigenic effects of IFN-γ as part of the complex immune response to cancer, highlighting the relevance to identify IFN-γ responsive patients for the improvement of therapies that exploit associated signaling pathways.
Collapse
Affiliation(s)
- Flávia Castro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Patrícia Cardoso
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Raquel Madeira Gonçalves
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Karine Serre
- IMM – Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria José Oliveira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|