1
|
Meier RPH, Ben Nasr M, Fife BT, Finger EB, Fiorina P, Luo X, Bromberg JS. Best practices in islet transplantation in mice. Am J Transplant 2025:S1600-6135(25)00137-6. [PMID: 40089068 DOI: 10.1016/j.ajt.2025.03.008] [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: 10/20/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Islet transplantation in mice serves as a crucial preclinical model for understanding alloimmune and autoimmune mechanisms, optimizing immunosuppressive strategies, and developing novel therapies for diabetes. This review provides a comprehensive overview of best practices in murine islet transplantation, including diabetes induction models, technical aspects of islet transplantation, and criteria for transplant graft and rejection. We discuss the immunological challenges posed by major histocompatibility complex disparities, the impact of various transplantation sites, and the limitations of murine models in translating findings to clinical settings. Special emphasis is placed on emerging strategies such as stem cell-derived insulin-producing cells, immune tolerance induction, and alternative transplantation sites. Although mouse models have significantly advanced our understanding of diabetes and β-cell replacement, their inherent differences from human physiology necessitate careful interpretation of findings. The review also highlights novel imaging modalities, immunosuppressive protocols, and biomarkers for graft monitoring, underscoring the need for further refinement of these models to bridge the gap between experimental research and clinical application. By standardizing methodologies and addressing translational limitations, murine islet transplantation studies remain a key model in transplantation and can continue to shape the future of β-cell replacement therapies for insulin-dependent diabetes.
Collapse
Affiliation(s)
- Raphael P H Meier
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian T Fife
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
2
|
Wang Q, Ren J, Lin X, Zhang B, Li J, Weng Y. Inflammatory stimulus-responsive polymersomes reprogramming glucose metabolism mitigates rheumatoid arthritis. Biomaterials 2025; 312:122760. [PMID: 39163825 DOI: 10.1016/j.biomaterials.2024.122760] [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: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
Inflammation-resident cells within arthritic sites undergo a metabolic shift towards glycolysis, which greatly aggravates rheumatoid arthritis (RA). Reprogramming glucose metabolism can suppress abnormal proliferation and activation of inflammation-related cells without affecting normal cells, holding potential for RA therapy. Single 2-deoxy-d-glucose (2-DG, glycolysis inhibitor) treatment often cause elevated ROS, which is detrimental to RA remission. The rational combination of glycolysis inhibition with anti-inflammatory intervention might cooperatively achieve favorable RA therapy. To improve drug bioavailability and exert synergetic effect, stable co-encapsulation of drugs in long circulation and timely drug release in inflamed milieu is highly desirable. Herein, we designed a stimulus-responsive hyaluronic acid-triglycerol monostearate polymersomes (HTDD) co-delivering 2-DG and dexamethasone (Dex) to arthritic sites. After intravenous injection, HTDD polymersomes facilitated prolonged circulation and preferential distribution in inflamed sites, where overexpressed matrix metalloproteinases and acidic pH triggered drug release. Results indicated 2-DG can inhibit the excessive cell proliferation and activation, and improve Dex bioavailability by reducing Dex efflux. Dex can suppress inflammatory signaling and prevent 2-DG-induced oxidative stress. Thus, the combinational strategy ultimately mitigated RA by inhibiting glycolysis and hindering inflammatory signaling. Our study demonstrated the great potential in RA therapy by reprogramming glucose metabolism in arthritic sites.
Collapse
Affiliation(s)
- Qin Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Jianheng Ren
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xin Lin
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Bin Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jiao Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yajun Weng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
3
|
Rui X, Calderon FA, Wobma H, Gerdemann U, Albanese A, Cagnin L, McGuckin C, Michaelis KA, Naqvi K, Blazar BR, Tkachev V, Kean LS. Human OX40L-CAR-T regs target activated antigen-presenting cells and control T cell alloreactivity. Sci Transl Med 2024; 16:eadj9331. [PMID: 39413160 PMCID: PMC11789419 DOI: 10.1126/scitranslmed.adj9331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 07/09/2024] [Accepted: 09/23/2024] [Indexed: 10/18/2024]
Abstract
Regulatory T cells (Tregs) make major contributions to immune homeostasis. Because Treg dysfunction can lead to both allo- and autoimmunity, there is interest in correcting these disorders through Treg adoptive transfer. Two of the central challenges in clinically deploying Treg cellular therapies are ensuring phenotypic stability and maximizing potency. Here, we describe an approach to address both issues through the creation of OX40 ligand (OX40L)-specific chimeric antigen receptor (CAR)-Tregs under the control of a synthetic forkhead box P3 (FOXP3) promoter. The creation of these CAR-Tregs enabled selective Treg stimulation by engagement of OX40L, a key activation antigen in alloimmunity, including both graft-versus-host disease and solid organ transplant rejection, and autoimmunity, including rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus. We demonstrated that OX40L-CAR-Tregs were robustly activated in the presence of OX40L-expressing cells, leading to up-regulation of Treg suppressive proteins without induction of proinflammatory cytokine production. Compared with control Tregs, OX40L-CAR-Tregs more potently suppressed alloreactive T cell proliferation in vitro and were directly inhibitory toward activated monocyte-derived dendritic cells (DCs). We identified trogocytosis as one of the central mechanisms by which these CAR-Tregs effectively decrease extracellular display of OX40L, resulting in decreased DC stimulatory capacity. OX40L-CAR-Tregs demonstrated an enhanced ability to control xenogeneic graft-versus-host disease compared with control Tregs without abolishing the graft-versus-leukemia effect. These results suggest that OX40L-CAR-Tregs may have wide applicability as a potent cellular therapy to control both allo- and autoimmune diseases.
Collapse
Affiliation(s)
- Xianliang Rui
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Francesca Alvarez Calderon
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Holly Wobma
- Harvard Medical School, Boston, MA 02115, USA
- Division of Immunology, Boston Children’s Hospital, Boston, MA 02215, USA
| | - Ulrike Gerdemann
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Albanese
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Kisa Naqvi
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Bruce R. Blazar
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leslie S. Kean
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Kim KM, D'Elia AM, Rodell CB. Hydrogel-based approaches to target hypersensitivity mechanisms underlying autoimmune disease. Adv Drug Deliv Rev 2024; 212:115395. [PMID: 39004347 DOI: 10.1016/j.addr.2024.115395] [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: 11/08/2023] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
A robust adaptive immune response is essential for combatting pathogens. In the wrong context such as due to genetic and environmental factors, however, the same mechanisms crucial for self-preservation can lead to a loss of self-tolerance. Resulting autoimmunity manifests in the development of a host of organ-specific or systemic autoimmune diseases, hallmarked by aberrant immune responses and tissue damage. The prevalence of autoimmune diseases is on the rise, medical management of which focuses primarily on pharmacological immunosuppression that places patients at a risk of side effects, including opportunistic infections and tumorigenesis. Biomaterial-based drug delivery systems confer many opportunities to address challenges associated with conventional disease management. Hydrogels, in particular, can protect encapsulated cargo (drug or cell therapeutics) from the host environment, afford their presentation in a controlled manner, and can be tailored to respond to disease conditions or support treatment via multiplexed functionality. Moreover, localized delivery to affected sites by these approaches has the potential to concentrate drug action at the site, reduce off-target exposure, and enhance patient compliance by reducing the need for frequent administration. Despite their many benefits for the management of autoimmune disease, such biomaterial-based approaches focus largely on the downstream effects of hypersensitivity mechanisms and have a limited capacity to eradicate the disease. In contrast, direct targeting of mechanisms of hypersensitivity reactions uniquely enables prophylaxis or the arrest of disease progression by mitigating the basis of autoimmunity. One promising approach is to induce self-antigen-specific tolerance, which specifically subdues damaging autoreactivity while otherwise retaining the normal immune responses. In this review, we will discuss hydrogel-based systems for the treatment of autoimmune disease, with a focus on those that target hypersensitivity mechanisms head-on. As the field continues to advance, it will expand the range of therapeutic choices for people coping with autoimmune diseases, providing fresh prospects for better clinical outcomes and improved quality of life.
Collapse
Affiliation(s)
- Kenneth M Kim
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| | - Christopher B Rodell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Yang Z, Zhang Z, Li L, Jing Z, Ma Y, Lan T, Li Y, Lin Z, Fang W, Zhang J, Zhang J, Liang X, Wu B, Zheng Y, Zhang X. Bioengineered Artificial Extracellular Vesicles Presenting PD-L1 and Gal-9 Ameliorate New-Onset Type 1 Diabetes. Diabetes 2024; 73:1325-1335. [PMID: 38771941 DOI: 10.2337/db23-0987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/05/2024] [Indexed: 05/23/2024]
Abstract
An important factor in the development of type 1 diabetes (T1D) is the deficiency of inhibitory immune checkpoint ligands, specifically programmed cell death ligand 1 (PD-L1) and galectin-9 (Gal-9), in β-cells. Therefore, modulation of pancreas-infiltrated T lymphocytes by exogenous PD-L1 or Gal-9 is an ideal approach for treating new-onset T1D. We genetically engineered macrophage cells to generate artificial extracellular vesicles (aEVs) overexpressing PD-L1 and Gal-9, which could restrict islet autoreactive T lymphocytes and protect β-cells from destruction. Intriguingly, overexpression of Gal-9 stimulated macrophage polarization to the M2 phenotype with immunosuppressive attributes. Alternatively, both PD-L1- and Gal-9-presenting aEVs (PD-L1-Gal-9 aEVs) favorably adhered to T cells via the interaction of programmed cell death protein 1/PD-L1 or T-cell immunoglobulin mucin 3/Gal-9. Moreover, PD-L1-Gal-9 aEVs prominently promoted effector T-cell apoptosis and splenic regulatory T (Treg) cell formation in vitro. Notably, PD-L1-Gal-9 aEVs efficaciously reversed new-onset hyperglycemia in NOD mice, prevented T1D progression, and decreased the proportion and activation of CD4+ and CD8+ T cells infiltrating the pancreas, which together contributed to the preservation of residual β-cell survival and mitigation of hyperglycemia. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Zhaoxin Yang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhirang Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Liyan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhangyan Jing
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yumeng Ma
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Tianyu Lan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yuan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Zhongda Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Wenli Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jinxie Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jinling Zhang
- Department of Gynaecology, Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Xin Liang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Key Laboratory of Stem Cell and Regenerative Tissue Engineering, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Benqing Wu
- Center for Medical Experiments (CME), Benqing Laboratory, Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Yi Zheng
- Center for Medical Experiments (CME), Benqing Laboratory, Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Nguyen TL, Phan NM, Kim J. Administration of ROS-Scavenging Cerium Oxide Nanoparticles Simply Mixed with Autoantigenic Peptides Induce Antigen-Specific Immune Tolerance against Autoimmune Encephalomyelitis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33106-33120. [PMID: 38906850 DOI: 10.1021/acsami.4c05428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
The scavenging ability of cerium oxide nanoparticles (CeNPs) for reactive oxygen species has been intensively studied in the field of catalysis. However, the immunological impact of these particles has not yet been thoroughly investigated, despite intensive research indicating that modulation of the reactive oxygen species could potentially regulate cell fate and adaptive immune responses. In this study, we examined the intrinsic capability of CeNPs to induce tolerogenic dendritic cells via their reactive oxygen species-scavenging effect when the autoantigenic peptides were simply mixed with CeNPs. CeNPs effectively reduced the intracellular reactive oxygen species levels in dendritic cells in vitro, leading to the suppression of costimulatory molecules as well as NLRP3 inflammasome activation, even in the presence of pro-inflammatory stimuli. Subcutaneously administrated PEGylated CeNPs were predominantly taken up by antigen-presenting cells in lymph nodes and to suppress cell maturation in vivo. The administration of a mixture of PEGylated CeNPs and myelin oligodendrocyte glycoprotein peptides, a well-identified autoantigen associated with antimyelin autoimmunity, resulted in the generation of antigen-specific Foxp3+ regulatory T cells in mouse spleens. The induced peripheral regulatory T cells actively inhibited the infiltration of autoreactive T cells and antigen-presenting cells into the central nervous system, ultimately protecting animals from experimental autoimmune encephalomyelitis when tested using a mouse model mimicking human multiple sclerosis. Overall, our findings reveal the potential of CeNPs for generating antigen-specific immune tolerance to prevent multiple sclerosis, opening an avenue to restore immune tolerance against specific antigens by simply mixing the well-identified autoantigens with the immunosuppressive CeNPs.
Collapse
Affiliation(s)
- Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Zhao Y, Chen X, He P, Wang X, Xu Y, Hu R, Ou Y, Zhang Z, Zhang Z, Du G, Sun X. Transdermal Microneedles Alleviated Rheumatoid Arthritis by Inducing Immune Tolerance via Skin-Resident Antigen Presenting Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307366. [PMID: 38039446 DOI: 10.1002/smll.202307366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/26/2023] [Indexed: 12/03/2023]
Abstract
Restoring immune tolerance is the ultimate goal for rheumatoid arthritis (RA) treatment. The most reported oral or intravenous injection routes for the immunization of autoantigens cause gastrointestinal side effects, low patient compliance, and unsatisfied immune tolerance induction. Herein, the use of a transdermal microneedle patch is for the first time investigated to codeliver CII peptide autoantigen and rapamycin for reversing immune disorders of RA. The immunized microneedles efficiently recruit antigen-presenting cells particularly Langerhans cells, and induce tolerogenic dendritic cells at the administration skin site. The tolerogenic dendritic cells further homing to lymph nodes to activate systemic Treg cell differentiation, which upregulates the expression of anti-inflammatory mediators while inhibiting the polarization of Th1/2 and Th17 T cell phenotypes and the expression of inflammatory profiles. As a result, the optimized microneedles nearly completely eliminate RA symptoms and inflammatory infiltrations. Furthermore, it is demonstrated that a low dose of rapamycin is crucial for the successful induction of immune tolerance. The results indicate that a rationally designed microneedle patch is a promising strategy for immune balance restoration with increased immune tolerance induction efficiency and patient compliance.
Collapse
Affiliation(s)
- Yuanhao Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xiaoyan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xuanyu Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhihua Zhang
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Zhibing Zhang
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
8
|
Liu Q, Chen G, Liu X, Tao L, Fan Y, Xia T. Tolerogenic Nano-/Microparticle Vaccines for Immunotherapy. ACS NANO 2024. [PMID: 38323542 DOI: 10.1021/acsnano.3c11647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Autoimmune diseases, allergies, transplant rejections, generation of antidrug antibodies, and chronic inflammatory diseases have impacted a large group of people across the globe. Conventional treatments and therapies often use systemic or broad immunosuppression with serious efficacy and safety issues. Tolerogenic vaccines represent a concept that has been extended from their traditional immune-modulating function to induction of antigen-specific tolerance through the generation of regulatory T cells. Without impairing immune homeostasis, tolerogenic vaccines dampen inflammation and induce tolerogenic regulation. However, achieving the desired potency of tolerogenic vaccines as preventive and therapeutic modalities calls for precise manipulation of the immune microenvironment and control over the tolerogenic responses against the autoantigens, allergens, and/or alloantigens. Engineered nano-/microparticles possess desirable design features that can bolster targeted immune regulation and enhance the induction of antigen-specific tolerance. Thus, particle-based tolerogenic vaccines hold great promise in clinical translation for future treatment of aforementioned immune disorders. In this review, we highlight the main strategies to employ particles as exciting tolerogenic vaccines, with a focus on the particles' role in facilitating the induction of antigen-specific tolerance. We describe the particle design features that facilitate their usage and discuss the challenges and opportunities for designing next-generation particle-based tolerogenic vaccines with robust efficacy to promote antigen-specific tolerance for immunotherapy.
Collapse
Affiliation(s)
- Qi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Guoqiang Chen
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xingchi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Lu Tao
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Yubo Fan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Tian Xia
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
9
|
Phan NM, Nguyen TL, Shin H, Trinh TA, Kim J. ROS-Scavenging Lignin-Based Tolerogenic Nanoparticle Vaccine for Treatment of Multiple Sclerosis. ACS NANO 2023; 17:24696-24709. [PMID: 38051295 DOI: 10.1021/acsnano.3c04497] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease, in which the immune system attacks myelin. Although systemic immunosuppressive agents have been used to treat MS, long-term treatment with these drugs causes undesirable side effects such as altered glucose metabolism, insomnia, and hypertension. Herein, we propose a tolerogenic therapeutic vaccine to treat MS based on lignin nanoparticles (LNP) with intrinsic reactive oxygen species (ROS)-scavenging capacity derived from their phenolic moieties. The LNP loaded with autoantigens of MS allowed for inducing tolerogenic DCs with low-level expression of costimulatory molecules while presenting antigenic peptides. Intravenous injection of an LNP-based tolerogenic vaccine into an experimental autoimmune encephalomyelitis (EAE) model led to durable antigen-specific immune tolerance via inducing regulatory T cells (Tregs). Autoreactive T helper type 1 cells, T helper type 17 cells, and inflammatory antigen presentation cells (APCs) were suppressed in the central nervous system (CNS), ameliorating ongoing MS in early and late disease states. Additionally, the incorporation of dexamethasone into an LNP-based tolerogenic nanovaccine could further improve the recovery of EAE mice in the severe chronic stage. As lignin is the most abundant biomass and waste byproduct in the pulping industry, a lignin-based tolerogenic vaccine could be a novel, cost-effective, high-value vaccine platform with potent therapeutic efficiency in treating autoimmune diseases.
Collapse
Affiliation(s)
- Ngoc Man Phan
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Thanh Loc Nguyen
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Hyunsu Shin
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Thuy An Trinh
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
10
|
Schifferli A, Rüfer A, Rovo A, Nimmerjahn F, Cantoni N, Holbro A, Favre G, Dirks J, Wieland A, Faeth H, Pereira R, Kühne T. Immunomodulation with romiplostim as a second-line strategy in primary immune thrombocytopenia: The iROM study. Br J Haematol 2023; 203:119-130. [PMID: 37735543 DOI: 10.1111/bjh.19074] [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: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 09/23/2023]
Abstract
Thrombopoietin receptor agonists (TPO-RAs) stimulate platelet production, which might restore immunological tolerance in primary immune thrombocytopenia (ITP). The iROM study investigated romiplostim's immunomodulatory effects. Thirteen patients (median age, 31 years) who previously received first-line treatment received romiplostim for 22 weeks, followed by monitoring until week 52. In addition to immunological data, secondary end-points included the sustained remission off-treatment (SROT) rate at 1 year, romiplostim dose, platelet count and bleedings. Scheduled discontinuation of romiplostim and SROT were achieved in six patients with newly diagnosed ITP, whereas the remaining seven patients relapsed. Romiplostim dose titration was lower and platelet count response was stronger in patients with SROT than in relapsed patients. In all patients, regulatory T lymphocyte (Treg) counts increased until study completion and the counts were higher in patients with SROT. Interleukin (IL)-4, IL-9 and IL-17F levels decreased significantly in all patients. FOXP3 (Treg), GATA3 (Th2) mRNA expression and transforming growth factor-β levels increased in patients with SROT. Treatment with romiplostim modulates the immune system and possibly influences ITP prognosis. A rapid increase in platelet counts is likely important for inducing immune tolerance. Better outcomes might be achieved at an early stage of autoimmunity, but clinical studies are needed for confirmation.
Collapse
Affiliation(s)
- Alexandra Schifferli
- Department of Hematology/Oncology, University Children's Hospital Basel, Basel, Switzerland
| | - Axel Rüfer
- Department of Hematology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - Alicia Rovo
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nathan Cantoni
- Department of Hematology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Andreas Holbro
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Geneviève Favre
- Department of Hematology, Cantonal Hospital Liestal, Switzerland
| | - Jan Dirks
- Department of Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Anna Wieland
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Heike Faeth
- Medical University of Basel, Basel, Switzerland
| | | | - Thomas Kühne
- Department of Hematology/Oncology, University Children's Hospital Basel, Basel, Switzerland
| |
Collapse
|
11
|
Chen DP, Wen YH, Lin WT, Hsu FP, Yu KH. Exploration of the association between the single-nucleotide polymorphism of co-stimulatory system and rheumatoid arthritis. Front Immunol 2023; 14:1123832. [PMID: 37457686 PMCID: PMC10344454 DOI: 10.3389/fimmu.2023.1123832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction The human leukocyte antigen (HLA) has been linked to the majority of autoimmune diseases (ADs). However, non-HLA genes may be risk factors for ADs. A number of genes encoding proteins involved in regulating T-cell and B-cell function have been identified as rheumatoid arthritis (RA) susceptibility genes. Methods In this study, we investigated the association between RA and single-nucleotide polymorphisms (SNPs) of co-stimulatory or co-inhibitory molecules in 124 RA cases and 100 healthy controls without immune-related diseases [including tumor necrosis factor superfamily member 4 (TNFSF4), CD28, cytotoxic T-lymphocyte-associated protein 4 (CTLA4), and programmed cell death protein 1 (PDCD1)]. Results The results showed that there were 13 SNPs associated with RA, including rs181758110 of TNFSF4 (CC vs. CT, p = 0.038); rs3181096 of CD28 (TT vs. CC + CT, p = 0.035; CC vs. TT, p = 0.047); rs11571315 (TT vs. CT, p = 0.045), rs733618 (CC vs. TT + CT, p = 0.043), rs4553808 (AA vs. AG vs. GG, p = 0.035), rs11571316 (GG vs. AG vs. AA, p = 0.048; GG vs. AG + AA, p = 0.026; GG vs. AG, p = 0.014), rs16840252 (CC vs. CT vs. TT, p = 0.007; CC vs. CT, p = 0.011), rs5742909 (CC vs. CT vs. TT, p = 0.040), and rs11571319 of CTLA4 (GG vs. AG vs. AA, p < 0.001; GG vs. AG + AA, p = 0.048; AA vs. GG + AG, p = 0.001; GG vs. AA, p = 0.008; GG vs. AG, p ≤ 0.001); and rs10204525 (TT vs. CT + CC, p = 0.024; TT vs. CT, p = 0.021), rs2227982 (AA vs. GG, p = 0.047), rs36084323 (TT vs. CT vs. CC, p = 0.022; TT vs. CT + CC, p = 0.013; CC vs. TT + CT, p = 0.048; TT vs. CC, p = 0.008), and rs5839828 of PDCD1 (DEL vs. DEL/G vs. GG, p = 0.014; DEL vs. DEL/G + GG, p = 0.014; GG vs. DEL + DEL/G, p = 0.025; DEL vs. GG, p = 0.007). Discussion Consequently, these SNPs may play an important role in immune regulation, and further research into the role of these SNPs of immune regulatory genes in the pathogenesis of RA is required.
Collapse
Affiliation(s)
- Ding-Ping Chen
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Hao Wen
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Tzu Lin
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Fang-Ping Hsu
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuang-Hui Yu
- Division of Rheumatology, Allergy, and Immunology, Linkou Chang Gung University and Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
12
|
Haghighitalab A, Dominici M, Matin MM, Shekari F, Ebrahimi Warkiani M, Lim R, Ahmadiankia N, Mirahmadi M, Bahrami AR, Bidkhori HR. Extracellular vesicles and their cells of origin: Open issues in autoimmune diseases. Front Immunol 2023; 14:1090416. [PMID: 36969255 PMCID: PMC10031021 DOI: 10.3389/fimmu.2023.1090416] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
The conventional therapeutic approaches to treat autoimmune diseases through suppressing the immune system, such as steroidal and non-steroidal anti-inflammatory drugs, are not adequately practical. Moreover, these regimens are associated with considerable complications. Designing tolerogenic therapeutic strategies based on stem cells, immune cells, and their extracellular vesicles (EVs) seems to open a promising path to managing autoimmune diseases' vast burden. Mesenchymal stem/stromal cells (MSCs), dendritic cells, and regulatory T cells (Tregs) are the main cell types applied to restore a tolerogenic immune status; MSCs play a more beneficial role due to their amenable properties and extensive cross-talks with different immune cells. With existing concerns about the employment of cells, new cell-free therapeutic paradigms, such as EV-based therapies, are gaining attention in this field. Additionally, EVs' unique properties have made them to be known as smart immunomodulators and are considered as a potential substitute for cell therapy. This review provides an overview of the advantages and disadvantages of cell-based and EV-based methods for treating autoimmune diseases. The study also presents an outlook on the future of EVs to be implemented in clinics for autoimmune patients.
Collapse
Affiliation(s)
- Azadeh Haghighitalab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton VIC, Australia
| | - Naghmeh Ahmadiankia
- Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahdi Mirahmadi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| |
Collapse
|
13
|
Yousefpour P, Ni K, Irvine DJ. Targeted modulation of immune cells and tissues using engineered biomaterials. NATURE REVIEWS BIOENGINEERING 2023; 1:107-124. [PMID: 37772035 PMCID: PMC10538251 DOI: 10.1038/s44222-022-00016-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 09/30/2023]
Abstract
Therapies modulating the immune system offer the prospect of treating a wide range of conditions including infectious diseases, cancer and autoimmunity. Biomaterials can promote specific targeting of immune cell subsets in peripheral or lymphoid tissues and modulate the dosage, timing and location of stimulation, thereby improving safety and efficacy of vaccines and immunotherapies. Here we review recent advances in biomaterials-based strategies, focusing on targeting of lymphoid tissues, circulating leukocytes, tissue-resident immune cells and immune cells at disease sites. These approaches can improve the potency and efficacy of immunotherapies by promoting immunity or tolerance against different diseases.
Collapse
Affiliation(s)
- Parisa Yousefpour
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
14
|
Casey LM, Decker JT, Podojil JR, Rad L, Hughes KR, Rose JA, Pearson RM, Miller SD, Shea LD. Nanoparticle dose and antigen loading attenuate antigen-specific T-cell responses. Biotechnol Bioeng 2023; 120:284-296. [PMID: 36221192 PMCID: PMC9999438 DOI: 10.1002/bit.28252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022]
Abstract
Immune-mediated hypersensitivities such as autoimmunity, allergy, and allogeneic graft rejection are treated with therapeutics that suppress the immune system, and the lack of specificity is associated with significant side effects. The delivery of disease-relevant antigens (Ags) by carrier systems such as poly(lactide-co-glycolide) nanoparticles (PLG-Ag) and carbodiimide (ECDI)-fixed splenocytes (SP-Ag) has demonstrated Ag-specific tolerance induction in model systems of these diseases. Despite therapeutic outcomes by both platforms, tolerance is conferred with different efficacy. This investigation evaluated Ag loading and total particle dose of PLG-Ag on Ag presentation in a coculture system of dendritic cells (DCs) and Ag-restricted T cells, with SP-Ag employed as a control. CD25 expression was observed in nearly all T cells even at low concentrations of PLG-Ag, indicating efficient presentation of Ag by dendritic cells. However, the secretion of IL-2, Th1, and Th2 cytokines (IFNγ and IL-4, respectively) varied depending on PLG-Ag concentration and Ag loading. Concentration escalation of soluble Ag resulted in an increase in IL-2 and IFNγ and a decrease in IL-4. Treatment with PLG-Ag followed a similar trend but with lower levels of IL-2 and IFNγ secreted. Transcriptional Activity CEll ARrays (TRACER) were employed to measure the real-time transcription factor (TF) activity in Ag-presenting DCs. The kinetics and magnitude of TF activity was dependent on the Ag delivery method, concentration, and Ag loading. Ag positively regulated IRF1 activity and, as carriers, NPs and ECDI-treated SP negatively regulated this signaling. The effect of Ag loading and dose on tolerance induction were corroborated in vivo using the delayed-type hypersensitivity (DTH) and experimental autoimmune encephalomyelitis (EAE) mouse models where a threshold of 8 μg/mg Ag loading and 0.5 mg PLG-Ag dose were required for tolerance. Together, the effect of Ag loading and dosing on in vitro and in vivo immune regulation provide useful insights for translating Ag-carrier systems for the clinical treatment of immune disorders.
Collapse
Affiliation(s)
- Liam M. Casey
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Joseph T. Decker
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Joseph R. Podojil
- Department of Microbiology‐Immunology, Feinberg School of MedicineNorthwestern UniversityChicagollinoisUSA
| | - Laila Rad
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin R. Hughes
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Justin A. Rose
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ryan M. Pearson
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Stephen D. Miller
- Department of Microbiology‐Immunology, Feinberg School of MedicineNorthwestern UniversityChicagollinoisUSA
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology Center, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Lonnie D. Shea
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
15
|
Titus HE, Xu H, Robinson AP, Patel PA, Chen Y, Fantini D, Eaton V, Karl M, Garrison ED, Rose IVL, Chiang MY, Podojil JR, Balabanov R, Liddelow SA, Miller RH, Popko B, Miller SD. Repurposing the cardiac glycoside digoxin to stimulate myelin regeneration in chemically-induced and immune-mediated mouse models of multiple sclerosis. Glia 2022; 70:1950-1970. [PMID: 35809238 PMCID: PMC9378523 DOI: 10.1002/glia.24231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 06/07/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022]
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) autoimmune disease characterized by inflammation, demyelination, and neurodegeneration. The ideal MS therapy would both specifically inhibit the underlying autoimmune response and promote repair/regeneration of myelin as well as maintenance of axonal integrity. Currently approved MS therapies consist of non-specific immunosuppressive molecules/antibodies which block activation or CNS homing of autoreactive T cells, but there are no approved therapies for stimulation of remyelination nor maintenance of axonal integrity. In an effort to repurpose an FDA-approved medication for myelin repair, we chose to examine the effectiveness of digoxin, a cardiac glycoside (Na+ /K+ ATPase inhibitor), originally identified as pro-myelinating in an in vitro screen. We found that digoxin regulated multiple genes in oligodendrocyte progenitor cells (OPCs) essential for oligodendrocyte (OL) differentiation in vitro, promoted OL differentiation both in vitro and in vivo in female naïve C57BL/6J (B6) mice, and stimulated recovery of myelinated axons in B6 mice following demyelination in the corpus callosum induced by cuprizone and spinal cord demyelination induced by lysophosphatidylcholine (LPC), respectively. More relevant to treatment of MS, we show that digoxin treatment of mice with established MOG35-55 -induced Th1/Th17-mediated chronic EAE combined with tolerance induced by the i.v. infusion of biodegradable poly(lactide-co-glycolide) nanoparticles coupled with MOG35-55 (PLG-MOG35-55 ) completely ameliorated clinical disease symptoms and stimulated recovery of OL lineage cell numbers. These findings provide critical pre-clinical evidence supporting future clinical trials of myelin-specific tolerance with myelin repair/regeneration drugs, such as digoxin, in MS patients.
Collapse
Affiliation(s)
- Haley E. Titus
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Huan Xu
- NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Andrew P. Robinson
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Priyam A. Patel
- Quantitative Data Science Core Center for Genetic MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Yanan Chen
- NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Damiano Fantini
- UrologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Valerie Eaton
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Molly Karl
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Eric D. Garrison
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Indigo V. L. Rose
- Neuroscience Institute and Departments of Neuroscience, & Physiology, and OphthalmologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Ming Yi Chiang
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Joseph R. Podojil
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Cour Pharmaceutical Development CompanyNorthbrookIllinoisUSA
| | - Roumen Balabanov
- NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Shane A. Liddelow
- Neuroscience Institute and Departments of Neuroscience, & Physiology, and OphthalmologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Robert H. Miller
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Brian Popko
- NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Stephen D. Miller
- Department of Microbiology‐Immunology and the Interdepartmental Immunobiology CenterNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
16
|
Rahiman N, Mohammadi M, Alavizadeh SH, Arabi L, Badiee A, Jaafari MR. Recent advancements in nanoparticle-mediated approaches for restoration of multiple sclerosis. J Control Release 2022; 343:620-644. [PMID: 35176392 DOI: 10.1016/j.jconrel.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
Multiple Sclerosis (MS) is an autoimmune disease with complicated immunopathology which necessitates considering multifactorial aspects for its management. Nano-sized pharmaceutical carriers named nanoparticles (NPs) can support impressive management of disease not only in early detection and prognosis level but also in a therapeutic manner. The most prominent initiator of MS is the domination of cellular immunity to humoral immunity and increment of inflammatory cytokines. The administration of several platforms of NPs for MS management holds great promise so far. The efforts for MS management through in vitro and in vivo (experimental animal models) evaluations, pave a new way to a highly efficient therapeutic means and aiding its translation to the clinic in the near future.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of pharmaceutics, School of pharmacy, Mashhad University of Medical sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
New Insights into Artesunate as a Pleiotropic Regulator of Innate and Adaptive Immune Cells. J Immunol Res 2022; 2022:9591544. [PMID: 35178460 PMCID: PMC8844150 DOI: 10.1155/2022/9591544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/05/2022] [Accepted: 01/25/2022] [Indexed: 02/03/2023] Open
Abstract
Artesunate, one of the derivatives of artemisinin (“qinghaosu” in Chinese), is known as an antimalarial drug with high efficiency and low toxicity. Of interest, emerging evidences suggest that artesunate also possesses an immunomodulatory effect during innate and adaptive immune responses in cell types and context-dependent manner. Although it shows promising application in many diseases, such as inflammatory diseases, hypersensitivity, autoimmune diseases, and cancers, little is known about underlying molecular. In this review, we summarize recent advances of how artesunate regulates innate and adaptive immune cells. In addition, its potential application in immune-related diseases is also highlighted.
Collapse
|
18
|
Docampo MJ, Lutterotti A, Sospedra M, Martin R. Mechanistic and Biomarker Studies to Demonstrate Immune Tolerance in Multiple Sclerosis. Front Immunol 2022; 12:787498. [PMID: 35069562 PMCID: PMC8766750 DOI: 10.3389/fimmu.2021.787498] [Citation(s) in RCA: 4] [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: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
The induction of specific immunological tolerance represents an important therapeutic goal for multiple sclerosis and other autoimmune diseases. Sound knowledge of the target antigens, the underlying pathomechanisms of the disease and the presumed mechanisms of action of the respective tolerance-inducing approach are essential for successful translation. Furthermore, suitable tools and assays to evaluate the induction of immune tolerance are key aspects for the development of such treatments. However, investigation of the mechanisms of action underlying tolerance induction poses several challenges. The optimization of sensitive, robust methods which allow the assessment of low frequency autoreactive T cells and the long-term reduction or change of their responses, the detection of regulatory cell populations and their immune mediators, as well as the validation of specific biomarkers indicating reduction of inflammation and damage, are needed to develop tolerance-inducing approaches successfully to patients. This short review focuses on how to demonstrate mechanistic proof-of-concept in antigen-specific tolerance-inducing therapies in MS.
Collapse
Affiliation(s)
| | | | | | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Neurology Clinic, University Hospital Zurich & University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Wang H, Shang J, He Z, Zheng M, Jia H, Zhang Y, Yang W, Gao X, Gao F. Dual peptide nanoparticles platform for enhanced antigen-specific immune tolerance for treatment of experimental autoimmune encephalomyelitis. Biomater Sci 2022; 10:3878-3891. [DOI: 10.1039/d2bm00444e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current therapeutic strategies for autoimmune diseases including multiple sclerosis (MS) are directed toward nonspecific immunosuppression which has severe side effects. The induction of antigen-specific tolerance becomes an ideal therapy for...
Collapse
|
20
|
Valdes AZ. Immunological tolerance and autoimmunity. TRANSLATIONAL AUTOIMMUNITY 2022:325-345. [DOI: 10.1016/b978-0-12-822564-6.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Au KM, Tisch R, Wang AZ. In Vivo Bioengineering of Beta Cells with Immune Checkpoint Ligand as a Treatment for Early-Onset Type 1 Diabetes Mellitus. ACS NANO 2021; 15:19990-20002. [PMID: 34843648 DOI: 10.1021/acsnano.1c07538] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by autoreactive T cells targeting the insulin-producing beta (β) cells. Despite advances in insulin therapy, T1DM still leads to high morbidity and mortality in patients. A key focus of T1DM research has been to identify strategies that re-establish self-tolerance and suppress ongoing autoimmunity. Here, we describe a strategy that utilizes pretargeting and glycochemistry to bioengineer β cells in situ to induce β-cell-specific tolerance. We hypothesized that β-cell-targeted Ac4ManNAz-encapsulated nanoparticles deliver and establish β cells with high levels of surface reactive azide groups. We further theorized that administration of a dibenzylcyclooctyne (DBCO)-functionalized programmed death-ligand 1 immunoglobulin fusion protein (PD-L1-Ig) can be readily conjugated to the surface of native β cells. Using nonobese diabetic (NOD) mice, we demonstrated that our strategy effectively and selectively conjugates PD-L1 onto β cells through bioorthogonal stain-promoted azide-alkyne cycloaddition. We also showed that the in vivo functionalized β cells simultaneously present islet-specific antigen and PD-L1 to the engaged T cells, reversing early onset T1DM by reducing IFN-gamma expressing cytotoxic toxic T cells and inducing antigen-specific tolerance.
Collapse
Affiliation(s)
- Kin Man Au
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75230, United States
| | - Roland Tisch
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75230, United States
| |
Collapse
|
22
|
Sadanandan P, Payne NL, Sun G, Ashokan A, Gowd SG, Lal A, Satheesh KMK, Pulakkat S, Nair SV, Menon KN, Bernard CCA, Koyakutty M. Exploiting the preferential phagocytic uptake of nanoparticle-antigen conjugates for the effective treatment of autoimmunity. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102481. [PMID: 34748963 DOI: 10.1016/j.nano.2021.102481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/09/2021] [Accepted: 10/23/2021] [Indexed: 10/19/2022]
Abstract
Tolerance induction is central to the suppression of autoimmunity. Here, we engineered the preferential uptake of nano-conjugated autoantigens by spleen-resident macrophages to re-introduce self-tolerance and suppress autoimmunity. The brain autoantigen, myelin oligodendrocyte glycoprotein (MOG), was conjugated to 200 or 500 nm silica nanoparticles (SNP) and delivered to the spleen and liver-resident macrophages of experimental autoimmune encephalomyelitis (EAE) mice model of multiple sclerosis. MOG-SNP conjugates significantly reduced signs of EAE at a very low dose (50 μg) compared to the higher dose (>800 μg) of free-MOG. This was associated with reduced proliferation of splenocytes and pro-inflammatory cytokines secretion, decreased spinal cord inflammation, demyelination and axonal damage. Notably, biodegradable porous SNP showed an enhanced disease suppression assisted by elevated levels of regulatory T cells and programmed-death ligands (PD-L1/2) in splenic and lymph node cells. Our results demonstrate that targeting nano-conjugated autoantigens to tissue-resident macrophages in lymphoid organs can effectively suppress autoimmunity.
Collapse
Affiliation(s)
- Prashant Sadanandan
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India; Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Natalie L Payne
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Guizhi Sun
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Anusha Ashokan
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Siddaramana G Gowd
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Arsha Lal
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Kumar M K Satheesh
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Sreeranjini Pulakkat
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Shantikumar V Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Krishnakumar N Menon
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| | - Claude C A Bernard
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia.
| | - Manzoor Koyakutty
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
23
|
Liu J, Gao H, Xu LP, Mo XD, Liu R, Liang S, Wu N, Wang M, Wang Z, Chang YJ, Wang Y, Zhang XH, Huang XJ. Immunosuppressant indulges EBV reactivation and related lymphoproliferative disease by inhibiting Vδ2 + T cells activities after hematopoietic transplantation for blood malignancies. J Immunother Cancer 2021; 8:jitc-2019-000208. [PMID: 32221014 PMCID: PMC7206968 DOI: 10.1136/jitc-2019-000208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2020] [Indexed: 12/16/2022] Open
Abstract
Background Following the extensive use of immunosuppressive drugs in the clinic, immunosuppression-associated side effects have received increasing attention. Epstein-Barr virus (EBV) reactivation and related lymphoproliferative diseases (LPD) are the lethal complications observed after allogeneic hematopoietic cell transplantation (alloHCT). While studies generally suggest an association between immunosuppressants and EBV reactivation, the effects of specific immunosuppressive drugs and which T-cell subsets mediate these correlations are unclear. Vδ2+ T cells are correlated with EBV reactivation after alloHCT. Researchers have not determined whether Vδ2+ T-cell activities are affected by immunosuppressants and thereby facilitate EBV reactivation and related LPD. Methods A clinical cohort study of 170 patients with hematopoietic malignancies who received haploidentical hematopoietic cell transplantation (haploHCT) was performed to investigate whether the early cessation of mycophenolate mofetil (MMF) decreases EBV reactivation and related LPD and to determine whether this change is associated with the recovery of Vδ2 + T cells after transplantation. The effects of MMF on the expansion and anti-EBV capacity of Vδ2+ T cells were detected in vitro and in an immunodeficient mouse model. Results A reduction in the course of MMF significantly improved the recovery of Vδ2+ T cells from 30 to 90 days after haploHCT (p=0.002, p=0.042 and p=0.035, respectively), accompanied by a significant decrease in EBV reactivation (from 26% to 13%, p=0.033) and EBV-LPD (from 10.6% to 2.4%, p=0.029). The day-30 Vδ2+ T level remained an independent factor for EBV reactivation in patients with different MMF durations (p=0.007). In the in-vitro experiments, MMF inhibited Vδ2+ T-cell expansion and its cytotoxicity on EBV-transformed malignant cells. Furthermore, the therapeutic and prophylactic effects of adoptively transferred human Vδ2+ T cells were attenuated by the MMF treatment in immunodeficient mice with EBV-LPD. Conclusions These results elucidated a negative effect of immunosuppressants on the anti-EBV capacity of Vδ2+ T cells. Strategies that appropriately relieve the immunosuppression may improve anti-EBV immunity by increasing the activity of Vδ2+ T cells after alloHCT.
Collapse
Affiliation(s)
- Jiangying Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Haitao Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Dong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ruoyang Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Shuang Liang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ning Wu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ming Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhidong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China .,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Smallwood TB, Navarro S, Cristofori-Armstrong B, Watkins TS, Tungatt K, Ryan RYM, Haigh OL, Lutzky VP, Mulvenna JP, Rosengren KJ, Loukas A, Miles JJ, Clark RJ. Synthetic hookworm-derived peptides are potent modulators of primary human immune cell function that protect against experimental colitis in vivo. J Biol Chem 2021; 297:100834. [PMID: 34051231 PMCID: PMC8239465 DOI: 10.1016/j.jbc.2021.100834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
The prevalence of autoimmune diseases is on the rise globally. Currently, autoimmunity presents in over 100 different forms and affects around 9% of the world's population. Current treatments available for autoimmune diseases are inadequate, expensive, and tend to focus on symptom management rather than cure. Clinical trials have shown that live helminthic therapy can decrease chronic inflammation associated with inflammatory bowel disease and other gastrointestinal autoimmune inflammatory conditions. As an alternative and better controlled approach to live infection, we have identified and characterized two peptides, Acan1 and Nak1, from the excretory/secretory component of parasitic hookworms for their therapeutic activity on experimental colitis. We synthesized Acan1 and Nak1 peptides from the Ancylostoma caninum and Necator americanus hookworms and assessed their structures and protective properties in human cell-based assays and in a mouse model of acute colitis. Acan1 and Nak1 displayed anticolitic properties via significantly reducing weight loss and colon atrophy, edema, ulceration, and necrosis in 2,4,6-trinitrobenzene sulfonic acid-exposed mice. These hookworm peptides prevented mucosal loss of goblet cells and preserved intestinal architecture. Acan1 upregulated genes responsible for the repair and restitution of ulcerated epithelium, whereas Nak1 downregulated genes responsible for epithelial cell migration and apoptotic cell signaling within the colon. These peptides were nontoxic and displayed key immunomodulatory functions in human peripheral blood mononuclear cells by suppressing CD4+ T cell proliferation and inhibiting IL-2 and TNF production. We conclude that Acan1 and Nak1 warrant further development as therapeutics for the treatment of autoimmunity, particularly gastrointestinal inflammatory conditions.
Collapse
Affiliation(s)
- Taylor B Smallwood
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Severine Navarro
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia; Woolworths Centre for Child Nutrition Research, Institute of Health and Biomedical Innovation, Queensland University of Technology, QLD, Australia
| | - Ben Cristofori-Armstrong
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Thomas S Watkins
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Centre for Molecular Therapeutics, The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia
| | - Katie Tungatt
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Centre for Molecular Therapeutics, The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia; Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, QLD, Australia
| | - Rachael Y M Ryan
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia; Centre for Molecular Therapeutics, The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia
| | - Oscar L Haigh
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Viviana P Lutzky
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jason P Mulvenna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - K Johan Rosengren
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, QLD, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia
| | - John J Miles
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia; Centre for Molecular Therapeutics, The Australian Institute of Tropical Health and Medicine, James Cook University, QLD, Australia; Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, QLD, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff, Wales, United Kingdom.
| | - Richard J Clark
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, QLD, Australia.
| |
Collapse
|
25
|
Au KM, Medik Y, Ke Q, Tisch R, Wang AZ. Immune Checkpoint-Bioengineered Beta Cell Vaccine Reverses Early-Onset Type 1 Diabetes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101253. [PMID: 33963786 PMCID: PMC8222180 DOI: 10.1002/adma.202101253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/18/2021] [Indexed: 05/14/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease that results from autoreactive T cells destroying insulin-producing pancreatic beta (β) cells. The development of T1DM is associated with the deficiency of co-inhibitory immune checkpoint ligands (e.g., PD-L1, CD86, and Gal-9) in β cells. Here, a new translational approach based on metabolic glycoengineering and bioorthogonal click chemistry, which bioengineers β cells with co-inhibitory immune checkpoint molecules that induce antigen-specific immunotolerance and reverse early-onset hyperglycemia is reported. To achieve this goal, a subcutaneous injectable acellular pancreatic extracellular matrix platform for localizing the bioengineered β cells while creating a pancreas-like immunogenic microenvironment, in which the autoreactive T cells can interface with the β cells, is devised.
Collapse
Affiliation(s)
- Kin Man Au
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yusra Medik
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Qi Ke
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Roland Tisch
- Department of Microbiology and Immunology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
26
|
Shi Y, Lu Y, Zhu C, Luo Z, Li X, Liu Y, Jiang M, Liu X, Luo L, Du Y, You J. Targeted regulation of lymphocytic ER stress response with an overall immunosuppression to alleviate allograft rejection. Biomaterials 2021; 272:120757. [PMID: 33798960 DOI: 10.1016/j.biomaterials.2021.120757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/20/2022]
Abstract
Transplantation is the most effective, and sometimes the only resort for end-stage organ failure. However, allogeneic graft suffers greatly from lymphocyte-mediated immunorejection, which bears close relationship with a hyperactivation of endoplasmic reticulum (ER) stress response in host lymphocytes, especially in CD8+ T cells (T-8). Therefore, regulating lymphocytic ER unfolded protein response (UPR) might be a potential therapeutic breakthrough in alleviating graft rejection. Here, ER-targetable liposome is prepared via the surface modification of ER-targeting peptide (Pardaxin), which efficiently loads and directly delivers small molecule inhibitor of UPR sensor IRE1α into the ER of lymphocytes, inducing a systemic immunosuppression that facilitates tumorigenesis and metastasis in the tumor inoculation challenge in vivo. And in vitro, a stage-differential dependency of IRE1α in the phase transition of T-8 is identified. Specifically, inhibiting IRE1α at the early responding stages of T-8, especially at the activation phase, results in a shrunk proliferation, impaired effector function, and limited memory commitment, which might contribute centrally to the induced overall immunosuppression. Based on this, a classical acute rejection model, murine full-thickness trunk skin allograft that primary arises from the hyperactivity of T-lymphocyte, is used. Results suggest that lymphocytic IRE1α inactivation attenuates transplant rejection and prolongs graft survival, with a limited effector function and memory commitment of host T-8. Moreover, an even higher immunosuppressive effect is obtained when IRE1α inhibition is used in combination with immunosuppressant tacrolimus (FK506), which might owe to a synergistic regulation of inflammatory transcription factors. These findings provide a deeper insight into the biological polarization and stress response of lymphocytes, which might guide the future development of allogeneic transplantation.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Xiang Li
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Yu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Yongzhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
27
|
Jaime-Pérez JC, Ramos-Dávila EM, Meléndez-Flores JD, Gómez-De León A, Gómez-Almaguer D. Insights on chronic immune thrombocytopenia pathogenesis: A bench to bedside update. Blood Rev 2021; 49:100827. [PMID: 33771403 DOI: 10.1016/j.blre.2021.100827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Immune thrombocytopenia (ITP) is a heterogeneous disease with an unpredictable course. Chronicity can develop in up to two-thirds of adults and 20-25% of children, representing a significant burden on patients' quality of life. Despite acceptable responses to treatment, precise etiology and pathophysiology phenomena driving evolution to chronicity remain undefined. We analyzed reported risk factors for chronic ITP and associated them with proposed underlying mechanisms in its pathogenesis, including bone marrow (BM) microenvironment disturbances, clinical features, and immunological markers. Their understanding has diagnostic implications, such as screening for the presence of specific antibodies or BM examination employing molecular tools, which could help predict prognosis and recognize main pathogenic pathways in each patient. Identifying these underlying mechanisms could guide the use of personalized therapies such as all-trans retinoic acid, mTor inhibitors, FcRn inhibitors, oseltamivir, and others. Further research should lead to tailored treatments and chronic course prevention, improving patients' quality of life.
Collapse
Affiliation(s)
- José Carlos Jaime-Pérez
- Department of Hematology, Internal Medicine Division, Dr. Jose E. González University Hospital and School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico.
| | - Eugenia M Ramos-Dávila
- Department of Hematology, Internal Medicine Division, Dr. Jose E. González University Hospital and School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Jesús D Meléndez-Flores
- Department of Hematology, Internal Medicine Division, Dr. Jose E. González University Hospital and School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Andrés Gómez-De León
- Department of Hematology, Internal Medicine Division, Dr. Jose E. González University Hospital and School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - David Gómez-Almaguer
- Department of Hematology, Internal Medicine Division, Dr. Jose E. González University Hospital and School of Medicine, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
28
|
Volfson-Sedletsky V, Jones A, Hernandez-Escalante J, Dooms H. Emerging Therapeutic Strategies to Restore Regulatory T Cell Control of Islet Autoimmunity in Type 1 Diabetes. Front Immunol 2021; 12:635767. [PMID: 33815387 PMCID: PMC8015774 DOI: 10.3389/fimmu.2021.635767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Despite many decades of investigation uncovering the autoimmune mechanisms underlying Type 1 Diabetes (T1D), translating these findings into effective therapeutics has proven extremely challenging. T1D is caused by autoreactive T cells that become inappropriately activated and kill the β cells in the pancreas, resulting in insulin insufficiency and hyperglycemia. A large body of evidence supports the idea that the unchecked activation and expansion of autoreactive T cells in T1D is due to defects in immunosuppressive regulatory T cells (Tregs) that are critical for maintaining peripheral tolerance to islet autoantigens. Hence, repairing these Treg deficiencies is a much sought-after strategy to treat the disease. To accomplish this goal in the most precise, effective and safest way possible, restored Treg functions will need to be targeted towards suppressing the autoantigen-specific immune responses only and/or be localized in the pancreas. Here we review the most recent developments in designing Treg therapies that go beyond broad activation or expansion of non-specific polyclonal Treg populations. We focus on two cutting-edge strategies namely ex vivo generation of optimized Tregs for re-introduction in T1D patients vs direct in situ stimulation and restoration of endogenous Treg function.
Collapse
Affiliation(s)
- Victoria Volfson-Sedletsky
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Albert Jones
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Jaileene Hernandez-Escalante
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| | - Hans Dooms
- Arthritis and Autoimmune Diseases Research Center, Rheumatology Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,Department of Microbiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
29
|
Phillips BE, Garciafigueroa Y, Engman C, Liu W, Wang Y, Lakomy RJ, Meng WS, Trucco M, Giannoukakis N. Arrest in the Progression of Type 1 Diabetes at the Mid-Stage of Insulitic Autoimmunity Using an Autoantigen-Decorated All- trans Retinoic Acid and Transforming Growth Factor Beta-1 Single Microparticle Formulation. Front Immunol 2021; 12:586220. [PMID: 33763059 PMCID: PMC7982719 DOI: 10.3389/fimmu.2021.586220] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes (T1D) is a disorder of impaired glucoregulation due to lymphocyte-driven pancreatic autoimmunity. Mobilizing dendritic cells (DC) in vivo to acquire tolerogenic activity is an attractive therapeutic approach as it results in multiple and overlapping immunosuppressive mechanisms. Delivery of agents that can achieve this, in the form of micro/nanoparticles, has successfully prevented a number of autoimmune conditions in vivo. Most of these formulations, however, do not establish multiple layers of immunoregulation. all-trans retinoic acid (RA) together with transforming growth factor beta 1 (TGFβ1), in contrast, has been shown to promote such mechanisms. When delivered in separate nanoparticle vehicles, they successfully prevent the progression of early-onset T1D autoimmunity in vivo. Herein, we show that the approach can be simplified into a single microparticle formulation of RA + TGFβ1 with surface decoration with the T1D-relevant insulin autoantigen. We show that the onset of hyperglycemia is prevented when administered into non-obese diabetic mice that are at the mid-stage of active islet-selective autoimmunity. Unexpectedly, the preventive effects do not seem to be mediated by increased numbers of regulatory T-lymphocytes inside the pancreatic lymph nodes, at least following acute administration of microparticles. Instead, we observed a mild increase in the frequency of regulatory B-lymphocytes inside the mesenteric lymph nodes. These data suggest additional and potentially-novel mechanisms that RA and TGFβ1 could be modulating to prevent progression of mid-stage autoimmunity to overt T1D. Our data further strengthen the rationale to develop RA+TGFβ1-based micro/nanoparticle “vaccines” as possible treatments of pre-symptomatic and new-onset T1D autoimmunity.
Collapse
Affiliation(s)
- Brett E Phillips
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| | - Yesica Garciafigueroa
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| | - Carl Engman
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| | - Wen Liu
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States.,Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Yiwei Wang
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Robert J Lakomy
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Massimo Trucco
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| | - Nick Giannoukakis
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Immunogenicity Challenges Associated with Subcutaneous Delivery of Therapeutic Proteins. BioDrugs 2021; 35:125-146. [PMID: 33523413 PMCID: PMC7848667 DOI: 10.1007/s40259-020-00465-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/12/2022]
Abstract
The subcutaneous route of administration has provided convenient and non-inferior delivery of therapeutic proteins compared to intravenous infusion, but there is potential for enhanced immunogenicity toward subcutaneously administered proteins in a subset of patients. Unwanted anti-drug antibody response toward proteins or monoclonal antibodies upon repeated administration is shown to impact the pharmacokinetics and efficacy of multiple biologics. Unique immunogenicity challenges of the subcutaneous route have been realized through various preclinical and clinical examples, although subcutaneous delivery has often demonstrated comparable immunogenicity to intravenous administration. Beyond route of administration as a treatment-related factor of immunogenicity, certain product-related risk factors are particularly relevant to subcutaneously administered proteins. This review attempts to provide an overview of the mechanism of immune response toward proteins administered subcutaneously (subcutaneous proteins) and comments on product-related risk factors related to protein structure and stability, dosage form, and aggregation. A two-wave mechanism of antigen presentation in the immune response toward subcutaneous proteins is described, and interaction with dynamic antigen-presenting cells possessing high antigen processing efficiency and migratory activity may drive immunogenicity. Mitigation strategies for immunogenicity are discussed, including those in general use clinically and those currently in development. Mechanistic insights along with consideration of risk factors involved inspire theoretical strategies to provide antigen-specific, long-lasting effects for maintaining the safety and efficacy of therapeutic proteins.
Collapse
|
31
|
Li J, Chen J, Schroeder JA, Hu J, Williams CB, Shi Q. Platelet gene therapy induces robust immune tolerance even in a primed model via peripheral clonal deletion of antigen-specific T cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 23:719-730. [PMID: 33575117 PMCID: PMC7851450 DOI: 10.1016/j.omtn.2020.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/30/2020] [Indexed: 11/08/2022]
Abstract
While platelet-specific gene therapy is effective in inducing immune tolerance to a targeted protein, how the reactivity of pre-existing immunity affects the efficacy, and whether CD8 T cells were involved in tolerization, is unclear. In this study, ovalbumin (OVA) was used as a surrogate protein. Platelet-OVA expression was introduced by 2bOVA lentivirus transduction of Sca-1+ cells from either wild-type (WT)/CD45.2 or OT-II/CD45.2 donors followed by transplantation into OVA-primed WT/CD45.1 recipients preconditioned with 6.6 Gy of irradiation. Sustained platelet-OVA expression was achieved in >85% of OVA-primed recipients but abolished in animals with high-reactive pre-existing immunity. As confirmed by OVA rechallenge and skin graft transplantation, immune tolerance was achieved in 2bOVA-transduced recipients. We found that there is a negative correlation between platelet-OVA expression and the percentage of OVA-specific CD4 T cells and a positive correlation with the OVA-specific regulatory T (Treg) cells. Using the OT-I/WT model, we showed that antigen-specific CD8 T cells were partially deleted in recipients after platelet-targeted gene transfer. Taken together, our studies demonstrate that robust antigen-specific immune tolerance can be achieved through platelet-specific gene therapy via peripheral clonal deletion of antigen-specific CD4 and CD8 T effector cells and induction of antigen-specific Treg cells. There is an antagonistic dynamic process between immune responses and immune tolerance after platelet-targeted gene therapy.
Collapse
Affiliation(s)
- Jing Li
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA.,Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Juan Chen
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA
| | - Jocelyn A Schroeder
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jianda Hu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Calvin B Williams
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.,Children's Research Institute, Children's Wisconsin, Milwaukee, WI, USA
| | - Qizhen Shi
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI, USA.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.,Children's Research Institute, Children's Wisconsin, Milwaukee, WI, USA.,Midwest Athletes Against Childhood Cancer Fund Research Center, Milwaukee, WI, USA
| |
Collapse
|
32
|
Xie M, Viviani M, Fussenegger M. Engineering precision therapies: lessons and motivations from the clinic. Synth Biol (Oxf) 2020; 6:ysaa024. [PMID: 33817342 PMCID: PMC7998714 DOI: 10.1093/synbio/ysaa024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
In the past decade, gene- and cell-based therapies have been at the forefront of the biomedical revolution. Synthetic biology, the engineering discipline of building sophisticated 'genetic software' to enable precise regulation of gene activities in living cells, has been a decisive success factor of these new therapies. Here, we discuss the core technologies and treatment strategies that have already gained approval for therapeutic applications in humans. We also review promising preclinical work that could either enhance the efficacy of existing treatment strategies or pave the way for new precision medicines to treat currently intractable human conditions.
Collapse
Affiliation(s)
- Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Mirta Viviani
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Titus HE, Chen Y, Podojil JR, Robinson AP, Balabanov R, Popko B, Miller SD. Pre-clinical and Clinical Implications of "Inside-Out" vs. "Outside-In" Paradigms in Multiple Sclerosis Etiopathogenesis. Front Cell Neurosci 2020; 14:599717. [PMID: 33192332 PMCID: PMC7654287 DOI: 10.3389/fncel.2020.599717] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is an immune-mediated neurological disorder, characterized by central nervous system (CNS) inflammation, oligodendrocyte loss, demyelination, and axonal degeneration. Although autoimmunity, inflammatory demyelination and neurodegeneration underlie MS, the initiating event has yet to be clarified. Effective disease modifying therapies need to both regulate the immune system and promote restoration of neuronal function, including remyelination. The challenge in developing an effective long-lived therapy for MS requires that three disease-associated targets be addressed: (1) self-tolerance must be re-established to specifically inhibit the underlying myelin-directed autoimmune pathogenic mechanisms; (2) neurons must be protected from inflammatory injury and degeneration; (3) myelin repair must be engendered by stimulating oligodendrocyte progenitors to remyelinate CNS neuronal axons. The combined use of chronic and relapsing remitting experimental autoimmune encephalomyelitis (C-EAE, R-EAE) (“outside-in”) as well as progressive diphtheria toxin A chain (DTA) and cuprizone autoimmune encephalitis (CAE) (“inside-out”) mouse models allow for the investigation and specific targeting of all three of these MS-associated disease parameters. The “outside-in” EAE models initiated by myelin-specific autoreactive CD4+ T cells allow for the evaluation of both myelin-specific tolerance in the absence or presence of neuroprotective and/or remyelinating agents. The “inside-out” mouse models of secondary inflammatory demyelination are triggered by toxin-induced oligodendrocyte loss or subtle myelin damage, which allows evaluation of novel therapeutics that could promote remyelination and neuroprotection in the CNS. Overall, utilizing these complementary pre-clinical MS models will open new avenues for developing therapeutic interventions, tackling MS from the “outside-in” and/or “inside-out”.
Collapse
Affiliation(s)
- Haley E Titus
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yanan Chen
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph R Podojil
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Cour Pharmaceutical Development Company, Inc., Northbrook, IL, United States
| | - Andrew P Robinson
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Roumen Balabanov
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Brian Popko
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Cour Pharmaceutical Development Company, Inc., Northbrook, IL, United States.,Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
34
|
Affiliation(s)
- Jeffrey A Bluestone
- From the Sean N. Parker Autoimmune Research Laboratory (J.A.B.) and the Diabetes Center (J.A.B., M.A.), University of California, San Francisco, San Francisco
| | - Mark Anderson
- From the Sean N. Parker Autoimmune Research Laboratory (J.A.B.) and the Diabetes Center (J.A.B., M.A.), University of California, San Francisco, San Francisco
| |
Collapse
|
35
|
Dumitrescu M, Trusca VG, Savu L, Stancu IG, Ratiu AC, Simionescu M, Gafencu AV. Adenovirus-Mediated FasL Minigene Transfer Endows Transduced Cells with Killer Potential. Int J Mol Sci 2020; 21:ijms21176011. [PMID: 32825521 PMCID: PMC7504687 DOI: 10.3390/ijms21176011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Fas ligand (First apoptosis signal ligand, FasL, also known as CD95L) is the common executioner of apoptosis within the tumor necrosis factor (TNF) superfamily. We aimed to induce functional FasL expression in transduced cells using an adenovirus vector, which has the advantage of strong and transient induction of the gene included in the adenoviral genome. Here, we report that the adenovirus carrying a truncated FasL gene, named FasL minigene, encoding the full-length FasL protein (Ad-gFasL) is more efficient than the adenovirus carrying FasL cDNA (Ad-cFasL) in the induction of FasL expression in transduced cells. FasL minigene (2887 bp) lacking the second intron and a part of the 3'-UTR was created to reduce the gene length due to the size limitation of the adenoviral genome. The results show that, in transduced hepatocytes, strong expression of mRNA FasL appeared after 10 h for Ad-gFasL, while for Ad-cFasL, a faint expression appeared after 16 h. For Ad-gFasL, the protein expression was noticed starting with 0.5 transfection units (TU)/cell, while for Ad-cFasL, it could not be revealed. FasL-expressing endothelial cells induced apoptosis of A20 cells in co-culture experiments. FasL-expressing cells may be exploitable in various autoimmune diseases such as graft-versus-host disease, chronic colitis, and type I diabetes.
Collapse
Affiliation(s)
- Madalina Dumitrescu
- Gene Regulation and Molecular Therapies Laboratory, Institute of Cellular Biology and Pathology “N. Simionescu”, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (M.D.); (V.G.T.); (M.S.)
| | - Violeta Georgeta Trusca
- Gene Regulation and Molecular Therapies Laboratory, Institute of Cellular Biology and Pathology “N. Simionescu”, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (M.D.); (V.G.T.); (M.S.)
| | - Lorand Savu
- Molecular Biology Department, Genetic Lab, 9 Căpitan Nicolae Drossu Street, 012071 Bucharest, Romania; (L.S.); (I.G.S.); (A.C.R.)
| | - Ioana Georgeta Stancu
- Molecular Biology Department, Genetic Lab, 9 Căpitan Nicolae Drossu Street, 012071 Bucharest, Romania; (L.S.); (I.G.S.); (A.C.R.)
| | - Attila Cristian Ratiu
- Molecular Biology Department, Genetic Lab, 9 Căpitan Nicolae Drossu Street, 012071 Bucharest, Romania; (L.S.); (I.G.S.); (A.C.R.)
| | - Maya Simionescu
- Gene Regulation and Molecular Therapies Laboratory, Institute of Cellular Biology and Pathology “N. Simionescu”, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (M.D.); (V.G.T.); (M.S.)
| | - Anca Violeta Gafencu
- Gene Regulation and Molecular Therapies Laboratory, Institute of Cellular Biology and Pathology “N. Simionescu”, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania; (M.D.); (V.G.T.); (M.S.)
- Correspondence: ; Tel.: +40-21-319-2327 (ext. 222)
| |
Collapse
|
36
|
Colazo JM, Evans BC, Farinas AF, Al-Kassis S, Duvall CL, Thayer WP. Applied Bioengineering in Tissue Reconstruction, Replacement, and Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 25:259-290. [PMID: 30896342 DOI: 10.1089/ten.teb.2018.0325] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPACT STATEMENT The use of autologous tissue in the reconstruction of tissue defects has been the gold standard. However, current standards still face many limitations and complications. Improving patient outcomes and quality of life by addressing these barriers remain imperative. This article provides historical perspective, covers the major limitations of current standards of care, and reviews recent advances and future prospects in applied bioengineering in the context of tissue reconstruction, replacement, and regeneration.
Collapse
Affiliation(s)
- Juan M Colazo
- 1Vanderbilt University School of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,2Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brian C Evans
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Angel F Farinas
- 4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Salam Al-Kassis
- 4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Craig L Duvall
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Wesley P Thayer
- 3Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,4Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
37
|
Irvine DJ, Aung A, Silva M. Controlling timing and location in vaccines. Adv Drug Deliv Rev 2020; 158:91-115. [PMID: 32598970 PMCID: PMC7318960 DOI: 10.1016/j.addr.2020.06.019] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023]
Abstract
Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.
Collapse
Affiliation(s)
- Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Murillo Silva
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
38
|
Li B, Yuan Z, Jain P, Hung HC, He Y, Lin X, McMullen P, Jiang S. De novo design of functional zwitterionic biomimetic material for immunomodulation. SCIENCE ADVANCES 2020; 6:eaba0754. [PMID: 32523997 PMCID: PMC7259941 DOI: 10.1126/sciadv.aba0754] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/30/2020] [Indexed: 05/14/2023]
Abstract
Superhydrophilic zwitterionic polymers are a class of nonfouling materials capable of effectively resisting any nonspecific interactions with biological systems. We designed here a functional zwitterionic polymer that achieves a trade-off between nonspecific interactions providing the nonfouling property and a specific interaction for bioactive functionality. Built from phosphoserine, an immune-signaling molecule in nature, this zwitterionic polymer exhibits both nonfouling and immunomodulatory properties. Its conjugation to uricase is shown to proactively eradicate all unwanted immune response, outperforming the zwitterionic polymers. On the other hand, this polymer could significantly prolong the half-life of protein drugs in vivo, overcoming the innate drawback of phosphoserine in inducing accelerated clearance. Our demonstration of a nonfouling zwitterionic material with built-in immunomodulatory functionality provides new insights into the fundamental design of biomaterials, as well as far-reaching implications for broad applications such as drug delivery, implants, and cell therapy.
Collapse
Affiliation(s)
- Bowen Li
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Zhefan Yuan
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Priyesh Jain
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Hsiang-Chieh Hung
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yuwei He
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xiaojie Lin
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Patrick McMullen
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Shaoyi Jiang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
39
|
Forte E, Zhang Z, Thorp EB, Hummel M. Cytomegalovirus Latency and Reactivation: An Intricate Interplay With the Host Immune Response. Front Cell Infect Microbiol 2020; 10:130. [PMID: 32296651 PMCID: PMC7136410 DOI: 10.3389/fcimb.2020.00130] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
CMV is an ancient herpesvirus that has co-evolved with its host over millions of years. The 236 kbp genome encodes at least 165 genes, four non-coding RNAs and 14 miRNAs. Of the protein-coding genes, 43-44 are core replication genes common to all herpesviruses, while ~30 are unique to betaherpesviruses. Many CMV genes are involved in evading detection by the host immune response, and others have roles in cell tropism. CMV replicates systemically, and thus, has adapted to various biological niches within the host. Different biological niches may place competing demands on the virus, such that genes that are favorable in some contexts are unfavorable in others. The outcome of infection is dependent on the cell type. In fibroblasts, the virus replicates lytically to produce infectious virus. In other cell types, such as myeloid progenitor cells, there is an initial burst of lytic gene expression, which is subsequently silenced through epigenetic repression, leading to establishment of latency. Latently infected monocytes disseminate the virus to various organs. Latency is established through cell type specific mechanisms of transcriptional silencing. In contrast, reactivation is triggered through pathways activated by inflammation, infection, and injury that are common to many cell types, as well as differentiation of myeloid cells to dendritic cells. Thus, CMV has evolved a complex relationship with the host immune response, in which it exploits cell type specific mechanisms of gene regulation to establish latency and to disseminate infection systemically, and also uses the inflammatory response to infection as an early warning system which allows the virus to escape from situations in which its survival is threatened, either by cellular damage or infection of the host with another pathogen. Spontaneous reactivation induced by cellular aging/damage may explain why extensive expression of lytic genes has been observed in recent studies using highly sensitive transcriptome analyses of cells from latently infected individuals. Recent studies with animal models highlight the potential for harnessing the host immune response to blunt cellular injury induced by organ transplantation, and thus, prevent reactivation of CMV and its sequelae.
Collapse
Affiliation(s)
- Eleonora Forte
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Zheng Zhang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Edward B. Thorp
- Department of Pathology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Mary Hummel
- Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
40
|
Emerging Role of Myeloid-derived Suppressor Cells in the Biology of Transplantation Tolerance. Transplantation 2020; 104:467-475. [DOI: 10.1097/tp.0000000000002996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Kuang ZS, Yang YL, Wei W, Wang JL, Long XY, Li KY, Tong CY, Sun Z, Song ZJ. Clinical characteristics and prognosis of community-acquired pneumonia in autoimmune disease-induced immunocompromised host: A retrospective observational study. World J Emerg Med 2020; 11:145-151. [PMID: 32351646 DOI: 10.5847/wjem.j.1920-8642.2020.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Community-acquired pneumonia (CAP) in autoimmune diseases (AID)-induced immunocompromised host (ICH) had a high incidence and poor prognosis. However, only a few studies had determined the clinical characteristics of these patients. Our study was to explore the characteristics and predictors of mortality in CAP patients accompanied with AID-induced ICH. METHODS From 2013 to 2018, a total of 94 CAP patients accompanied with AID-induced ICH, admitted to Emergency Department of Zhongshan Hospital, Fudan University, were enrolled in this study. Clinical data and the risk regression estimates of repeated predictors were evaluated by generalized estimating equations (GEEs) analysis. An open-cohort approach was used to classify patient's outcomes into the survival or non-survival group. RESULTS The hospital mortality of patients with CAP occurring in AID-induced ICH was 60.64%. No significant differences were found with respect to clinical symptoms and lung images between survival and non-survival groups, while renal insufficiency and dysfunction of coagulation had higher proportions in non-survival patients (P<0.05). Both noninvasive ventilation (NIV) and invasive mechanical ventilation (IMV) were performed more frequently in non-survival group (P< 0.05). By the multivariate GEEs analysis, the repeated measured longitudinal indices of neutrophil-to-lymphocyte ratio (NLR) (odds ratio [OR]=1.055, 95% confidence interval [95%CI] 1.025-1.086), lactate dehydrogenase (LDH) (OR=1.004, 95%CI 1.002-1.006) and serum creatinine (sCr) (OR=1.018, 95%CI 1.008-1.028), were associated with a higher risk of mortality. CONCLUSION The CAP patients in AID-induced ICH had a high mortality. A significant relationship was demonstrated between the factors of NLR, LDH, sCr and mortality risk in these patients.
Collapse
Affiliation(s)
- Zhong-Shu Kuang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Lin Yang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Wei
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian-Li Wang
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiang-Yu Long
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ke-Yong Li
- Department of Pharmacology, University of Virginia School of Medicine Charlottesville, Virginia, USA
| | - Chao-Yang Tong
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhan Sun
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhen-Ju Song
- Department of Emergency Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
42
|
Engineering human stellate cells for beta cell replacement therapy promotes in vivo recruitment of regulatory T cells. Mater Today Bio 2019; 2:100006. [PMID: 32159143 PMCID: PMC7061575 DOI: 10.1016/j.mtbio.2019.100006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/25/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by destruction of pancreatic β cells. One of the promising therapeutic approaches in T1D is the transplantation of islets; however, it has serious limitations. To address these limitations, immunotherapeutic strategies have focused on restoring immunologic tolerance, preventing transplanted cell destruction by patients’ own immune system. Macrophage-derived chemokines such as chemokine-ligand-22 (CCL22) can be utilized for regulatory T cell (Treg) recruitment and graft tolerance. Stellate cells (SCs) have various immunomodulatory functions: recruitment of Tregs and induction of T-cell apoptosis. Here, we designed a unique immune-privileged microenvironment around implantable islets through overexpression of CCL22 proteins by SCs. We prepared pseudoislets with insulin-secreting mouse insulinoma-6 (MIN6) cells and human SCs as a model to mimic naive islet morphology. Our results demonstrated that transduced SCs can secrete CCL22 and recruit Tregs toward the implantation site in vivo. This study is promising to provide a fundamental understanding of SC-islet interaction and ligand synthesis and transport from SCs at the graft site for ensuring local immune tolerance. Our results also establish a new paradigm for creating tolerable grafts for other chronic diseases such as diabetes, anemia, and central nervous system (CNS) diseases, and advance the science of graft tolerance.
Collapse
|
43
|
Thangavelu G, Blazar BR. Achievement of Tolerance Induction to Prevent Acute Graft-vs.-Host Disease. Front Immunol 2019; 10:309. [PMID: 30906290 PMCID: PMC6419712 DOI: 10.3389/fimmu.2019.00309] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/06/2019] [Indexed: 01/04/2023] Open
Abstract
Acute graft-vs.-host disease (GVHD) limits the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT), a main therapy to treat various hematological disorders. Despite rapid progress in understanding GVHD pathogenesis, broad immunosuppressive agents are most often used to prevent and remain the first line of therapy to treat GVHD. Strategies enhancing immune tolerance in allo-HSCT would permit reductions in immunosuppressant use and their associated undesirable side effects. In this review, we discuss the mechanisms responsible for GVHD and advancement in strategies to achieve immune balance and tolerance thereby avoiding GVHD and its complications.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
44
|
Clinical hepatocyte transplantation. GASTROENTEROLOGIA Y HEPATOLOGIA 2019; 42:202-208. [DOI: 10.1016/j.gastrohep.2018.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/21/2018] [Accepted: 10/10/2018] [Indexed: 12/18/2022]
|
45
|
Gammon JM, Jewell CM. Engineering Immune Tolerance with Biomaterials. Adv Healthc Mater 2019; 8:e1801419. [PMID: 30605264 PMCID: PMC6384133 DOI: 10.1002/adhm.201801419] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/05/2018] [Indexed: 12/28/2022]
Abstract
Autoimmune diseases, rejection of transplanted organs and grafts, chronic inflammatory diseases, and immune-mediated rejection of biologic drugs impact a large number of people across the globe. New understanding of immune function is revealing exciting opportunities to help tackle these challenges by harnessing-or correcting-the specificity of immune function. However, realizing this potential requires precision control over the interaction between regulatory immune cues, antigens attacked during inflammation, and the tissues where these processes occur. Engineered materials-such as polymeric and lipid particles, scaffolds, and inorganic materials-offer powerful features that can help to selectively regulate immune function during disease without compromising healthy immune functions. This review highlights some of the exciting developments to leverage biomaterials as carriers, depots, scaffolds-and even as agents with intrinsic immunomodulatory features-to promote immunological tolerance.
Collapse
Affiliation(s)
- Joshua M. Gammon
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive RM 5110, College Park, MD 20742, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive RM 5110, College Park, MD 20742, USA ; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD 20742, USA; United States Department of Veterans Affairs, Baltimore VA Medical center, 10. N Green Street, Baltimore, Maryland 21201, USA; Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
46
|
Seyed MA. A comprehensive review on Phyllanthus derived natural products as potential chemotherapeutic and immunomodulators for a wide range of human diseases. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2019. [DOI: 10.1016/j.bcab.2019.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
47
|
Ohnuma K, Hatano R, Dang NH, Morimoto C. Rheumatic diseases associated with immune checkpoint inhibitors in cancer immunotherapy. Mod Rheumatol 2018; 29:721-732. [DOI: 10.1080/14397595.2018.1532559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kei Ohnuma
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Ryo Hatano
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nam H. Dang
- Division of Hematology/Oncology, University of Florida, Gainesville, FL, USA
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
48
|
Li J, Qiu D, Liu Y, Xiong J, Wang Y, Yang X, Fu X, Zheng L, Luo G, Xing M, Wu Y. Cytomembrane Infused Polymer Accelerating Delivery of Myelin Antigen Peptide to Treat Experimental Autoimmune Encephalomyelitis. ACS NANO 2018; 12:11579-11590. [PMID: 30265798 DOI: 10.1021/acsnano.8b06575] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
While there has been extensive development of soluble epitope-specific peptides to induce immune tolerance for the treatment of autoimmune diseases, the clinical efficacy of soluble-peptides-based immunotherapy was still uncertain. Recent strategies to develop antigen carriers coupled with peptides have shown promising results in preclinical animal models. Here we developed functional amphiphilic hyperbranched (HB) polymers with different grafting degrees of hydrophobic chains as antigen myelin antigen oligodendrocyte glycoprotein (MOG) peptide carriers and evaluated their ability to induce immune tolerance. We show that these polymers could efficiently deliver antigen peptide, and the uptake amount by bone marrow dendritic cells (BMDCs) was correlated with the hydrophobicity of polymers. We observe that these polymers have a higher ability to activate BMDCs and a higher efficacy to induce antigen-specific T cell apoptosis than soluble peptides, irrespective of hydrophobicity. We show that intravenous injection of polymer-conjugated MOG peptide, but not soluble peptide, markedly treats the clinical symptoms of experimental autoimmune encephalomyelitis in mice. Together, these results demonstrate the potential for using amphiphilic HB polymers as antigen carriers to deliver peptides for pathogenic autoreactive T cell deletion/tolerance strategies to treat autoimmune disorders.
Collapse
Affiliation(s)
- Jian Li
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| | - Ding Qiu
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| | - Yuqing Liu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , 400038 , China
- Department of Mechanical Engineering , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Jian Xiong
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| | - Ying Wang
- Department of Mechanical Engineering , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Xia Yang
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| | - Xiaolan Fu
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Gaoxing Luo
- Department of Mechanical Engineering , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Malcolm Xing
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , 400038 , China
- Department of Mechanical Engineering , University of Manitoba , Winnipeg , MB R3T 2N2 , Canada
| | - Yuzhang Wu
- Institute of Immunology, PLA , Third Military Medical University (Army Medical University) , Chongqing 400038 , China
| |
Collapse
|
49
|
Zai K, Yuzuriha K, Kishimura A, Mori T, Katayama Y. Preparation of Complexes between Ovalbumin Nanoparticles and Retinoic Acid for Efficient Induction of Tolerogenic Dendritic Cells. ANAL SCI 2018; 34:1243-1248. [PMID: 29962375 DOI: 10.2116/analsci.18p252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The induction of antigen-specific immunotolerance has been gathering attention concerning the therapy of allergy and autoimmune diseases. Tolerogenic dendritic cells (tDCs) play crucial roles in immunotolerance therapy because they induce anergic responses for auto-reactive helper T cells, and also enhance differentiation to regulatory T cells to maintain tolerance against auto-antigens. All-trans retinoic acid (ATRA) is one of the representative molecules used to induce tDCs. We have proposed a simple formulation of ovalbumin nanoparticles complexed with ATRA (OVA/RA NPs). OVA/RA NPs were taken up by DCs and successfully induced phenotypes of tDCs.
Collapse
Affiliation(s)
- Khadijah Zai
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University
| | | | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of Systems Life Sciences, Kyushu University.,Center for Future Chemistry, Kyushu University.,International Research Center for Molecular Systems, Kyushu University
| | - Takeshi Mori
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of Systems Life Sciences, Kyushu University.,Center for Future Chemistry, Kyushu University
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University.,Graduate School of Systems Life Sciences, Kyushu University.,Center for Future Chemistry, Kyushu University.,International Research Center for Molecular Systems, Kyushu University.,Centre for Advanced Medicine Innovation, Kyushu University.,Department of Biomedical Engineering, Chung Yuan Christian University
| |
Collapse
|
50
|
Pearson RM, Podojil JR, Shea LD, King NJC, Miller SD, Getts DR. Overcoming challenges in treating autoimmuntity: Development of tolerogenic immune-modifying nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 18:282-291. [PMID: 30352312 DOI: 10.1016/j.nano.2018.10.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/02/2018] [Accepted: 10/12/2018] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases, such as celiac disease, multiple sclerosis, and type 1 diabetes, are leading causes of morbidity and mortality in the United States. In these disease states, immune regulatory mechanisms fail that result in T and B cell-mediated destruction of self-tissues. The known role of T cells in mediating autoimmune diseases has led to the emergence of numerous therapies aimed at inactivating T cells, however successful 'tolerance-inducing' strategies have not yet emerged for approved standard-of-care clinical use. In this review, we describe relevant examples of antigen-specific tolerance approaches that have been applied in clinical trials for human diseases. Furthermore, we describe the evolution of biomaterial approaches from cell-based therapies to induce immune tolerance with a focus on the Tolerogenic Immune-Modifying nanoParticle (TIMP) platform. The TIMP platform can be designed to treat various autoimmune conditions and is currently in clinical trials testing its ability to reverse celiac disease.
Collapse
Affiliation(s)
- Ryan M Pearson
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph R Podojil
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lonnie D Shea
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas J C King
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Bosch Institute and Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Medical Sciences Sydney Medical School, University of Sydney, Australia
| | - Stephen D Miller
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniel R Getts
- Research & Development, Cour Pharmaceuticals Development Company, Northbrook, IL, USA; Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|