1
|
Kong AHY, Wu AJ, Ho OKY, Leung MMK, Huang AS, Yu Y, Zhang G, Lyu A, Li M, Cheung KH. Exploring the Potential of Aptamers in Targeting Neuroinflammation and Neurodegenerative Disorders: Opportunities and Challenges. Int J Mol Sci 2023; 24:11780. [PMID: 37511539 PMCID: PMC10380291 DOI: 10.3390/ijms241411780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neuroinflammation is the precursor for several neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Targeting neuroinflammation has emerged as a promising strategy to address a wide range of CNS pathologies. These NDDs still present significant challenges in terms of limited and ineffective diagnosis and treatment options, driving the need to explore innovative and novel therapeutic alternatives. Aptamers are single-stranded nucleic acids that offer the potential for addressing these challenges through diagnostic and therapeutic applications. In this review, we summarize diagnostic and therapeutic aptamers for inflammatory biomolecules, as well as the inflammatory cells in NDDs. We also discussed the potential of short nucleotides for Aptamer-Based Targeted Brain Delivery through their unique features and modifications, as well as their ability to penetrate the blood-brain barrier. Moreover, the unprecedented opportunities and substantial challenges of using aptamers as therapeutic agents, such as drug efficacy, safety considerations, and pharmacokinetics, are also discussed. Taken together, this review assesses the potential of aptamers as a pioneering approach for target delivery to the CNS and the treatment of neuroinflammation and NDDs.
Collapse
Affiliation(s)
- Anna Hau-Yee Kong
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aston Jiaxi Wu
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Olivia Ka-Yi Ho
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Maggie Ming-Ki Leung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Alexis Shiying Huang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Min Li
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - King-Ho Cheung
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
2
|
Momeni M, Mashayekhi K, Navashenaq JG, Sankian M. Identification of G-quadruplex anti-Interleukin-2 aptamer with high specificity through SELEX stringency. Heliyon 2022; 8:e09721. [PMID: 35756119 PMCID: PMC9218155 DOI: 10.1016/j.heliyon.2022.e09721] [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: 10/22/2021] [Revised: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022] Open
Abstract
Aptamers are short single-stranded oligonucleotides capable of binding to various targets with high specificity and affinity. This study aimed to identify an aptamer against mouse interleukin-2 (mIL-2) as one of the most important cytokines in autoimmune diseases for diagnostic and therapeutic purposes. For this purpose, 14 SELEX rounds were performed on recombinant mIL-2 with high stringency. The dot blot and flow cytometry techniques were conducted to determine affinity, dissociation constant (Kd), specificity, and SELEX rounds screening. The stringency of rounds was considered based on aptamer/target incubation time, washing steps, and target proteins. Finally, the aptamer's structure was mapped and predicted by M-fold and QGRS Mapper web-based software. After 14 rounds, the flow cytometry analysis revealed that the 11th round was a proper round. The high-affinity aptamers M20 and M15 were chosen for their ability to bind mIL-2. According to DNA folding software, M20 and M15 aptamers had G-quadruplex and stem-loop structures, respectively. The M20 aptamer affinity was greater than M15, and its predicted Kd was 91 nM. A simple SELEX protocol with round stringency was explained to identify DNA aptamers against protein targets. The reported G-quadruplex aptamer might have potential diagnostic or therapeutic application in IL-2–related disorders.
Collapse
Affiliation(s)
- Mohsen Momeni
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Mojtaba Sankian
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Mollasalehi N, Francois-Moutal L, Porciani D, Burke DH, Khanna M. Aptamers Targeting Hallmark Proteins of Neurodegeneration. Nucleic Acid Ther 2022; 32:235-250. [PMID: 35452303 DOI: 10.1089/nat.2021.0091] [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: 11/13/2022] Open
Abstract
Neurodegeneration is a progressive deterioration of neural structures leading to cognitive or motor impairment of the affected patient. There is still no effective therapy for any of the most common neurodegenerative diseases (NDs) such as Alzheimer's or Parkinson's disease. Although NDs exhibit distinct clinical characteristics, many are characterized by the accumulation of misfolded proteins or peptide fragments in the brain and/or spinal cord. The presence of similar inclusion bodies in patients with diverse NDs provides a rationale for developing therapies directed at overlapping disease mechanisms. A novel targeting strategy involves the use of aptamers for therapeutic development. Aptamers are short nucleic acid ligands able to recognize molecular targets with high specificity and high affinity. Despite the fact that several academic groups have shown that aptamers have the potential to be used in therapeutic and diagnostic applications, their clinical translation is still limited. In this study, we describe aptamers that have been developed against proteins relevant to NDs, including prion protein and amyloid beta (Aβ), cell surface receptors and other cytoplasmic proteins. This review also describes advances in the application of these aptamers in imaging, protein detection, and protein quantification, and it provides insights about their accelerated clinical use for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Niloufar Mollasalehi
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona, USA.,Center for Innovation in Brain Science, Tucson, Arizona, USA
| | - Liberty Francois-Moutal
- Center for Innovation in Brain Science, Tucson, Arizona, USA.,Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - David Porciani
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, USA.,MU Bond Life Sciences Center, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Donald H Burke
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri-Columbia, Columbia, Missouri, USA.,MU Bond Life Sciences Center, University of Missouri-Columbia, Columbia, Missouri, USA
| | - May Khanna
- Center for Innovation in Brain Science, Tucson, Arizona, USA.,Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
4
|
Vachhani K, Prodeus A, Nakamura S, Rockel JS, Hopfgartner A, Kapoor M, Gariépy J, Whyne C, Nam D. Can CD200R1 Agonists Slow the Progression of Osteoarthritis Secondary to Injury? Front Immunol 2022; 13:836837. [PMID: 35359946 PMCID: PMC8964055 DOI: 10.3389/fimmu.2022.836837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic knee osteoarthritis is characterized by cartilage degeneration, subchondral bone remodeling, osteophyte formation, and synovial changes. Therapeutic targeting of inflammatory activity in the knee immediately post injury may alter the course of osteoarthritis development. This study aimed to determine whether CD200R1 agonists, namely the protein therapeutic CD200Fc or the synthetic DNA aptamer CCS13, both known to act as anti-inflammatory agents, are able to delay the pathogenesis of injury-associated knee osteoarthritis in a murine model. Ten week old male C57BL/6 mice were randomized and surgical destabilization of the medial meniscus (DMM) to induce knee arthritis or sham surgery as a control were performed. CCS13 was evaluated as a therapeutic treatment along with CD200Fc and a phosphate-buffered saline vehicle control. Oligonucleotides were injected intra-articularly beginning one week after surgery, with a total of six injections administered prior to sacrifice at 12 weeks post-surgery. Histopathological assessment was used as the primary outcome measure to assess cartilage and synovial changes, while µCT imaging was used to compare the changes to the subchondral bone between untreated and treated arthritic groups. We did not find any attenuation of cartilage degeneration or synovitis in DMM mice with CD200Fc or CCS13 at 12 weeks post-surgery, nor stereological differences in the properties of subchondral bone. The use of CD200R1 agonists to blunt the inflammatory response in the knee are insufficient to prevent disease progression in the mouse DMM model of OA without anatomical restoration of the normal joint biomechanics.
Collapse
Affiliation(s)
- Kathak Vachhani
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Aaron Prodeus
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Sayaka Nakamura
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Jason S. Rockel
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Mohit Kapoor
- Division of Orthopaedics, Osteoarthritis Research Program, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Jean Gariépy
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Cari Whyne
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
- Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Diane Nam
- Sunnybrook Research Institute, Toronto, ON, Canada
- Division of Orthopedic Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- *Correspondence: Diane Nam,
| |
Collapse
|
5
|
Su Y, Yamazaki S, Morisue R, Suzuki J, Yoshikawa T, Nakatsura T, Tsuboi M, Ochiai A, Ishii G. Tumor-Infiltrating T Cells Concurrently Overexpress CD200R with Immune Checkpoints PD-1, CTLA-4, and TIM-3 in Non-Small-Cell Lung Cancer. Pathobiology 2020; 88:218-227. [PMID: 33321503 DOI: 10.1159/000511557] [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: 08/19/2020] [Accepted: 09/13/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION CD200R has been reported to be the receptor for the immune checkpoint molecule CD200 and can transduce immune-suppressive signals. In this study, we mainly focused on the expression level of CD200R in T cells in pulmonary artery (PA) blood and non-small-cell lung cancer (NSCLC) tumor tissue. METHODS Immune cells were isolated from dissected tumor samples and PA blood of NSCLC patients and analyzed with multiparameter flow cytometry. The co-expression of CD200R with other immune checkpoints, including programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), was also investigated. RESULTS CD200R expression was observed on the surface of approximately 75% of T cells among tumor-infiltrating leukocytes (TILs). Compared to T cells extracted from TILs, only 55% of T cells extracted from PA blood exhibited CD200R expression. Moreover, with higher expression of CD200R, the expression of other immune checkpoints, including PD-1, CTLA-4, and TIM-3, was also increased in tumor-infiltrating T cells compared to T cells in PA blood. CONCLUSIONS Our results showed that those tumors were dominated by T cells expressing CD200R together with other checkpoints, which suggests a phenotypic change after T cell infiltration into the tumor, such as T cell exhaustion.
Collapse
Affiliation(s)
- Yinghan Su
- Laboratory of Cancer Biology, Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shota Yamazaki
- Laboratory of Cancer Biology, Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ryo Morisue
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Jun Suzuki
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Atsushi Ochiai
- Laboratory of Cancer Biology, Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan.,Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Genichiro Ishii
- Laboratory of Cancer Biology, Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan, .,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan, .,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan,
| |
Collapse
|
6
|
Yasmeen F, Seo H, Javaid N, Kim MS, Choi S. Therapeutic Interventions into Innate Immune Diseases by Means of Aptamers. Pharmaceutics 2020; 12:pharmaceutics12100955. [PMID: 33050544 PMCID: PMC7600108 DOI: 10.3390/pharmaceutics12100955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/03/2020] [Accepted: 10/04/2020] [Indexed: 12/25/2022] Open
Abstract
The immune system plays a crucial role in the body's defense system against various pathogens, such as bacteria, viruses, and parasites, as well as recognizes non-self- and self-molecules. The innate immune system is composed of special receptors known as pattern recognition receptors, which play a crucial role in the identification of pathogen-associated molecular patterns from diverse microorganisms. Any disequilibrium in the activation of a particular pattern recognition receptor leads to various inflammatory, autoimmune, or immunodeficiency diseases. Aptamers are short single-stranded deoxyribonucleic acid or ribonucleic acid molecules, also termed "chemical antibodies," which have tremendous specificity and affinity for their target molecules. Their features, such as stability, low immunogenicity, ease of manufacturing, and facile screening against a target, make them preferable as therapeutics. Immune-system-targeting aptamers have a great potential as a targeted therapeutic strategy against immune diseases. This review summarizes components of the innate immune system, aptamer production, pharmacokinetic characteristics of aptamers, and aptamers related to innate-immune-system diseases.
Collapse
|
7
|
Mashayekhi K, Ganji A, Sankian M. Designing a new dimerized anti human TNF-α aptamer with blocking activity. Biotechnol Prog 2020; 36:e2969. [PMID: 31989789 DOI: 10.1002/btpr.2969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/10/2023]
Abstract
The human tumor necrosis factor α (hTNF-α) is an important pro-inflammatory cytokine which plays critical roles in inflammatory diseases such as rheumatoid arthritis (RA). The anti-TNF-α proteins can reduce symptoms of RA. Due to limitations of protein-based therapies, it is necessary to find new anti-TNF-α agents instead of common anti-TNF-α proteins. Therefore, the aim of the current study was to identify a new DNA aptamer with anti-hTNF-α activity. The protein systematic evolution of ligands by exponential enrichment (SELEX) process was used for identifying DNA aptamers. Anti-hTNF-α aptamers were selected using dot blot, real-time PCR, and in vitro inhibitory assay. The selected aptamers were truncated in two steps, and finally, a dimer aptamer was constructed from different selected truncates to improve their inhibitory effect. Also, Etanercept was used as a positive control to inhibit TNF-α, in comparison to the designed aptamers. After 11 rounds, four aptamers with anti-hTNF-α inhibitory effect were identified. The truncation and dimerization strategy revealed a new dimer aptamer with 67 nM Kd , which has 40% inhibitory effect compared with Etanercept (60%). Overall, the dimerization and truncation aptamers could improve its activity. With regard to the several limitations of anti-TNF-α proteins therapies including immunogenicity, side effects, and cost-intensive, a new designed anti-hTNF-α dimer aptamer could be considered as a potential therapeutic and/or diagnostic agent for hTNF-α-related disorders.
Collapse
Affiliation(s)
- Kazem Mashayekhi
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Ganji
- Department of Microbiology and Immunology, School of Medicine, Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Mojtaba Sankian
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Erin N, Dilmaç S, Curry A, Duymuş Ö, Tanriover G, Prodeus A, Gariepy J, Gorczynski RM. CD200 mimetic aptamer PEG-M49 markedly increases the therapeutic effects of pegylated liposomal doxorubicin in a mouse model of metastatic breast carcinoma: an effect independent of CD200 receptor 1. Cancer Immunol Immunother 2019; 69:103-114. [PMID: 31811336 DOI: 10.1007/s00262-019-02444-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022]
Abstract
We previously reported that CD200 overexpression in the host decreases progression and metastasis of the highly aggressive metastatic 4THM breast carcinoma. We have explored a possible synergistic interaction between the CD200 mimetic PEG-M49 and pegylated liposomal doxorubicin (Peg-Dox) in wild-type CD200 knockout (CD200-/-) and CD200 Receptor 1 knockout (CD200R1-/-) mice for the first time. A 4THM breast carcinoma model and three groups of BALB/c mice (wild type, CD200-/- and CD200R1-/-) were used. Five days after injection of tumor cells, mice were injected with Peg-Dox (ip, once a week) and PEG-M49 or a control aptamer (iv, every 3 days). Necropsies were performed either 12 (mid-point) or 24 (endpoint) days after injection and the extent of tumor growth, visceral metastasis and changes in the tumor-directed immune response were evaluated. PEG-M49 and Peg-Dox co-treatment induced complete tumor regression and loss of macroscopic lung metastasis in four out of seven WT mice. This synergistic anti-tumoral effect is thought to be due to Peg-M49-induced inhibition of Gr1 + CD11b + cells and Peg-Dox-induced increases in tumor-infiltrating CD8 + and CD8CD4 double-positive cells. Similar changes were observed in CD200R1-/- mice indicating that the primary effects of Peg-M49 are mediated by non-CD200R1 receptors. We also demonstrated for the first time that tumor growth, metastasis, and tumor infiltrating GR1 + CD11b + cells were markedly increased in CD200R1-/- mice, indicating an anti-inflammatory and protective role of CD200. CD200 mimetics might be a safe and effective immunomodulatory treatment in conjunction with classical chemotherapeutics for therapy of aggressive metastatic breast carcinoma.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, Immunopharmacology and Immunooncology Research unit, Akdeniz University School of Medicine, B-Blok Kat 1 Immunoloji, 07070, Antalya, Turkey.
| | - Sayra Dilmaç
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Anna Curry
- University Health Network, Toronto General Hospital, Toronto, Canada
| | - Özlem Duymuş
- Department of Medical Pharmacology, Immunopharmacology and Immunooncology Research unit, Akdeniz University School of Medicine, B-Blok Kat 1 Immunoloji, 07070, Antalya, Turkey
| | - Gamze Tanriover
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Aaron Prodeus
- Sunnybrook Cancer Centre, Toronto General Hospital, Toronto, Canada
| | - Jean Gariepy
- Sunnybrook Cancer Centre, Toronto General Hospital, Toronto, Canada
| | | |
Collapse
|
9
|
Wang LQ, Yu XW, Zhang YA, Chu J, Zhou XL, Zhang HM, Cao GF, Teng L. Decreased expression of CD200 and CD200R1 by human decidual tissues in spontaneous early abortion. J Matern Fetal Neonatal Med 2019; 33:3399-3408. [PMID: 30897989 DOI: 10.1080/14767058.2019.1572741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objective: To examine the cellular distribution and the expression of CD200 and its receptor 1 (CD200R1) in human deciduas in first-trimester pregnant women with spontaneous early abortion (SEA) and normal pregnancy, and to explore their role in the etiology of SEA.Subjects and methods: Thirty-five women at 6-10-week gestation with SA and 30 women of similar gestational age with a healthy pregnancy were recruited. Expression of CD200 and CD200R1 in the deciduas was determined using immunohistochemistry, confocal laser scanning microscope, Western blot, and real-time PCR (RT-PCR).Results: The decidual stromal cells, glandular epithelial cells, and vessel endothelial cells during the first trimester of pregnancy express both CD200 and CD200R1 proteins. During this period, the expression of CD200 in glandular epithelial cells and vessel endothelial cells is significantly higher in normal pregnancy than that in women with SEA (0.3079 ± 0.0674 versus 0.2735 ± 0.0515; 0.4077 ± 0.1366 versus 0.3249 ± 0.0993); the expression of CD200R1 in stromal cells, decidual stromal cells, glandular epithelial cells is significantly higher during normal pregnancy than SEA (0.2574 ± 0.0588 versus 0.2292 ± 0.0415; 0.3617 ± 0.1046 versus 0.2804 ± 0.0640). Western blot analysis showed an approximately 44% decrease in CD200R1expression in decidua in the SEA versus the controls. Finally, in decidua, the expression of both CD200 protein and CD200R1 transcript are significantly higher in healthy first-trimester pregnancy than in SEA (CD200: 2.2089 ± 1.2754 versus 0.7241 ± 0.2143; CD200R1: 15.7843 ± 10.7085 versus 7.3381 ± 5.8529).Conclusions: Women with SEA have a lower level of CD200 and CD200R1 expression in deciduas compared with normal pregnant women suggesting that under physiological conditions, CD200 and CD200R1 expression by deciduas is important to prevent fetal loss ensure a successful pregnancy.
Collapse
Affiliation(s)
- Li-Qin Wang
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Xue-Wen Yu
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yong-Ai Zhang
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Jing Chu
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Xiao-Lan Zhou
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Hai-Miao Zhang
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Guo-Fen Cao
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| | - Li Teng
- Nursing Department and Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Xi'an Medical College, Xi'an, P. R. China
| |
Collapse
|
10
|
Paluch C, Santos AM, Anzilotti C, Cornall RJ, Davis SJ. Immune Checkpoints as Therapeutic Targets in Autoimmunity. Front Immunol 2018; 9:2306. [PMID: 30349540 PMCID: PMC6186808 DOI: 10.3389/fimmu.2018.02306] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022] Open
Abstract
Antibodies that block the immune checkpoint receptors PD1 and CTLA4 have revolutionized the treatment of melanoma and several other cancers, but in the process, a new class of drug side effect has emerged—immune related adverse events. The observation that therapeutic blockade of these inhibitory receptors is sufficient to break self-tolerance, highlights their crucial role in the physiological modulation of immune responses. Here, we discuss the rationale for targeting immune checkpoint receptors with agonistic agents in autoimmunity, to restore tolerance when it is lost. We review progress that has been made to date, using Fc-fusion proteins, monoclonal antibodies or other novel constructs to induce immunosuppressive signaling through these pathways. Finally, we explore potential mechanisms by which these receptors trigger and modulate immune cell function, and how understanding these processes might shape the design of more effective therapeutic agents in future.
Collapse
Affiliation(s)
- Christopher Paluch
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Consuelo Anzilotti
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Richard J Cornall
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J Davis
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
A Synthetic Cross-Species CD200R1 Agonist Suppresses Inflammatory Immune Responses In Vivo. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:350-358. [PMID: 30195773 PMCID: PMC6037911 DOI: 10.1016/j.omtn.2018.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 05/25/2018] [Accepted: 05/26/2018] [Indexed: 01/10/2023]
Abstract
Functional aptamers displaying agonistic or antagonistic properties are showing great promise as modulators of immune responses. Here, we report the development of a polyethylene glycol-modified (PEGylated) DNA aptamer as a cross-species (murine and human) CD200R1 agonist that modulates inflammatory responses in vivo. Specifically, DNA aptamers were discovered by performing independent SELEX searches on recombinant murine and human CD200R1. Aptamer motifs identified by next generation sequencing (NGS) were subsequently compared, leading to the discovery of motifs common to both targets. The CD200R1 DNA aptamer CCS13 displayed the highest agonistic activity toward CD200R1 in terms of suppressing the induction of cytotoxic T-lymphocytes (CTLs) in both human and murine allogeneic-mixed lymphocyte cultures (allo-MLCs). A 20-kDa polyethylene glycol (PEG) chain was covalently attached to the 5′ end of this aptamer, and the resulting conjugate was shown to block inflammatory responses in murine models of skin graft rejection and house-dust-mite-induced allergic airway inflammation. Importantly, this agonistic aptamer does not suppress CTL induction in 5-day allo-MLCs with responder cells derived from CD200R1−/− mice, indicating that its mode of action is directly linked to CD200R1 activation. This study suggests that one can derive agonistic DNA aptamers that can be verified as immuno-modulators in murine models with outcomes potentially translatable to the treatment of human conditions.
Collapse
|
12
|
Liu Y, Xie X, Xia LP, Lv H, Lou F, Ren Y, He ZY, Luo XG. Peripheral immune tolerance alleviates the intracranial lipopolysaccharide injection-induced neuroinflammation and protects the dopaminergic neurons from neuroinflammation-related neurotoxicity. J Neuroinflammation 2017; 14:223. [PMID: 29145874 PMCID: PMC5693474 DOI: 10.1186/s12974-017-0994-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroinflammation plays a critical role in the onset and development of neurodegeneration disorders such as Parkinson's disease. The immune activities of the central nervous system are profoundly affected by peripheral immune activities. Immune tolerance refers to the unresponsiveness of the immune system to continuous or repeated stimulation to avoid excessive inflammation and unnecessary by-stander injury in the face of continuous antigen threat. It has been proved that the immune tolerance could suppress the development of various peripheral inflammation-related diseases. However, the role of immune tolerance in neuroinflammation and neurodegenerative diseases was not clear. METHODS Rats were injected with repeated low-dose lipopolysaccharide (LPS, 0.3 mg/kg) intraperitoneally for 4 days to induce peripheral immune tolerance. Neuroinflammation was produced using intracranial LPS (15 μg) injection. Inflammation cytokines were measured using enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). Microglial activation were measured using immunostaining of Iba-1 and ED-1. Dopaminergic neuronal damage was evaluated using immunochemistry staining and stereological counting of TH-positive neurons. Behavioral impairment was evaluated using amphetamine-induced rotational behavioral assessment. RESULTS Compared with the non-immune tolerated animals, pre-treatment of peripheral immune tolerance significantly decreased the production of inflammatory cytokines, suppressed the microglial activation, and increased the number of dopaminergic neuronal survival in the substantia nigra. CONCLUSIONS Our results indicated that peripheral immune tolerance attenuated neuroinflammation and inhibited neuroinflammation-induced dopaminergic neuronal death.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xin Xie
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Li-Ping Xia
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hong Lv
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Fan Lou
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Yan Ren
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhi-Yi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xiao-Guang Luo
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
13
|
Prodeus A, Abdul-Wahid A, Sparkes A, Fischer NW, Cydzik M, Chiang N, Alwash M, Ferzoco A, Vacaresse N, Julius M, Gorczysnki RM, Gariépy J. VISTA.COMP - an engineered checkpoint receptor agonist that potently suppresses T cell-mediated immune responses. JCI Insight 2017; 2:94308. [PMID: 28931757 DOI: 10.1172/jci.insight.94308] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/10/2017] [Indexed: 01/25/2023] Open
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) is a recently discovered immune checkpoint ligand that functions to suppress T cell activity. The therapeutic potential of activating this immune checkpoint pathway to reduce inflammatory responses remains untapped, largely due to the inability to derive agonists targeting its unknown receptor. A dimeric construct of the IgV domain of VISTA (VISTA-Fc) was shown to suppress the activation of T cells in vitro. However, this effect required its immobilization on a solid surface, suggesting that VISTA-Fc may display limited efficacy as a VISTA-receptor agonist in vivo. Herein, we have designed a stable pentameric VISTA construct (VISTA.COMP) by genetically fusing its IgV domain to the pentamerization domain from the cartilage oligomeric matrix protein (COMP). In contrast to VISTA-Fc, VISTA.COMP does not require immobilization to inhibit the proliferation of CD4+ T cells undergoing polyclonal activation. Furthermore, we show that VISTA.COMP, but not VISTA-Fc, functions as an immunosuppressive agonist in vivo capable of prolonging the survival of skin allografts in a mouse transplant model as well as rescuing mice from acute concanavalin-A-induced hepatitis. Collectively, we believe our data demonstrate that VISTA.COMP is a checkpoint receptor agonist and the first agent to our knowledge targeting the putative VISTA-receptor to suppress T cell-mediated immune responses.
Collapse
Affiliation(s)
- Aaron Prodeus
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | | | - Nicholas W Fischer
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | - Nicholas Chiang
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Pharmaceutical Sciences and
| | - Mays Alwash
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Pharmaceutical Sciences and
| | - Alessandra Ferzoco
- Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Michael Julius
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Reginald M Gorczysnki
- Department of Pharmaceutical Sciences and.,Transplant Research Division, University Health Network, Toronto, Ontario, Canada
| | - Jean Gariépy
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Pharmaceutical Sciences and
| |
Collapse
|
14
|
Poolsup S, Kim CY. Therapeutic applications of synthetic nucleic acid aptamers. Curr Opin Biotechnol 2017; 48:180-186. [PMID: 28582756 DOI: 10.1016/j.copbio.2017.05.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/20/2017] [Accepted: 05/08/2017] [Indexed: 12/25/2022]
Abstract
It is possible to generate oligonucleotide aptamers for a wide variety of target molecules using a process known as Systematic Evolution of Ligands by Exponential Enrichment. Researchers have successfully generated aptamers which recognize specific metal ions, small chemical compounds, peptides, proteins, saccharides, and even whole cells. Aptamers show much promise as future therapeutics and as drug targeting agents. A particularly active area of aptamer research in the past two years was development of aptamer based cancer therapeutics and development of aptamer based cancer drug delivery systems. Aptamers were also used to address inflammatory diseases, infectious diseases, cardiovascular diseases, and eye diseases.
Collapse
Affiliation(s)
- Suttinee Poolsup
- Department of Chemistry, The University of Texas at El Paso, El Paso, TX 79912, United States
| | - Chu-Young Kim
- Department of Chemistry, The University of Texas at El Paso, El Paso, TX 79912, United States; School of Pharmacy, The University of Texas at El Paso, El Paso, TX 79902, United States.
| |
Collapse
|
15
|
Xie X, Luo X, Liu N, Li X, Lou F, Zheng Y, Ren Y. Monocytes, microglia, and CD200-CD200R1 signaling are essential in the transmission of inflammation from the periphery to the central nervous system. J Neurochem 2017; 141:222-235. [PMID: 28164283 DOI: 10.1111/jnc.13972] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 01/05/2023]
Abstract
Peripheral inflammation is known to trigger neuroinflammation and neurodegenerative disease. However, the key components during the propagation of inflammation from the periphery to the central nervous system (CNS) remain unclear. Lipopolysaccharide (LPS) was administered to Sprague-Dawley rats to induce peripheral inflammation. An intravenous injection and an intranigral injection of clodronate liposomes were given to deplete monocytes and microglia, respectively. Recombinant CD200 fusion protein (CD200Fc) or an anti-CD200R1 antibody was injected into the substantia nigra to manipulate the involvement of CD200 and CD200R1. Immunohistochemistry and immunofluorescence staining were used to measure microglial activation and dopaminergic neuronal loss. The expression of brain pro-inflammatory cytokines (i.e., tumor necrosis factor alpha, IL-1β) and CD200-CD200R1 signaling were measured by quantitative RT-PCR. Our data showed that the peripheral LPS injection activated the microglia and induced an increase in the levels of pro-inflammatory cytokines (i.e., tumor necrosis factor alpha, IL-1β). The depletion of either monocytes or microglia suppressed these inflammatory effects that were induced by peripheral LPS administration. The peripheral LPS injection increased the expression of CD200 and CD200R1 in the substantia nigra. Dopaminergic neuronal loss induced by the peripheral LPS injection was accelerated by the blockade of CD200-CD200R1 signaling with an anti-CD200R1 antibody and attenuated by intensifying the signaling with CD200Fc. These results highlight the importance of monocytes, microglia, and CD200-CD200R1 signaling in the transmission of inflammation from the periphery to the CNS.
Collapse
Affiliation(s)
- Xin Xie
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaoguang Luo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Na Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaohong Li
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fan Lou
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yumin Zheng
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Ren
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Targeting the PD-1/PD-L1 Immune Evasion Axis With DNA Aptamers as a Novel Therapeutic Strategy for the Treatment of Disseminated Cancers. MOLECULAR THERAPY-NUCLEIC ACIDS 2015; 4:e237. [PMID: 25919090 PMCID: PMC4417124 DOI: 10.1038/mtna.2015.11] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/18/2015] [Indexed: 12/15/2022]
Abstract
Blocking the immunoinhibitory PD-1:PD-L1 pathway using monoclonal antibodies has led to dramatic clinical responses by reversing tumor immune evasion and provoking robust and durable antitumor responses. Anti-PD-1 antibodies have now been approved for the treatment of melanoma, and are being clinically tested in a number of other tumor types as both a monotherapy and as part of combination regimens. Here, we report the development of DNA aptamers as synthetic, nonimmunogenic antibody mimics, which bind specifically to the murine extracellular domain of PD-1 and block the PD-1:PD-L1 interaction. One such aptamer, MP7, functionally inhibits the PD-L1-mediated suppression of IL-2 secretion in primary T-cells. A PEGylated form of MP7 retains the ability to block the PD-1:PD-L1 interaction, and significantly suppresses the growth of PD-L1+ colon carcinoma cells in vivo with a potency equivalent to an antagonistic anti-PD-1 antibody. Importantly, the anti-PD-1 DNA aptamer treatment was not associated with off-target TLR-9-related immune responses. Due to the inherent advantages of aptamers including their lack of immunogenicity, low cost, long shelf life, and ease of synthesis, PD-1 antagonistic aptamers may represent an attractive alternative over antibody-based anti PD-1 therapeutics.
Collapse
|