201
|
Abstract
Strategies to enhance, suppress, or qualitatively shape the immune response are of importance for diverse biomedical applications, such as the development of new vaccines, treatments for autoimmune diseases and allergies, strategies for regenerative medicine, and immunotherapies for cancer. However, the intricate cellular and molecular signals regulating the immune system are major hurdles to predictably manipulating the immune response and developing safe and effective therapies. To meet this challenge, biomaterials are being developed that control how, where, and when immune cells are stimulated in vivo, and that can finely control their differentiation in vitro. We review recent advances in the field of biomaterials for immunomodulation, focusing particularly on designing biomaterials to provide controlled immunostimulation, targeting drugs and vaccines to lymphoid organs, and serving as scaffolds to organize immune cells and emulate lymphoid tissues. These ongoing efforts highlight the many ways in which biomaterials can be brought to bear to engineer the immune system.
Collapse
Affiliation(s)
- Nathan A Hotaling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
| | - Li Tang
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
| | - Darrell J Irvine
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| | - Julia E Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia 30332;
| |
Collapse
|
202
|
Kontos S, Grimm AJ, Hubbell JA. Engineering antigen-specific immunological tolerance. Curr Opin Immunol 2015; 35:80-8. [DOI: 10.1016/j.coi.2015.05.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/04/2015] [Accepted: 05/13/2015] [Indexed: 01/07/2023]
|
203
|
Irvine DJ, Hanson MC, Rakhra K, Tokatlian T. Synthetic Nanoparticles for Vaccines and Immunotherapy. Chem Rev 2015; 115:11109-46. [PMID: 26154342 DOI: 10.1021/acs.chemrev.5b00109] [Citation(s) in RCA: 560] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Darrell J Irvine
- The Ragon Institute of MGH, Massachusetts Institute of Technology and Harvard University , 400 Technology Square, Cambridge, Massachusetts 02139, United States.,Howard Hughes Medical Institute , Chevy Chase, Maryland 20815, United States
| | | | | | | |
Collapse
|
204
|
Salazar-González JA, González-Ortega O, Rosales-Mendoza S. Gold nanoparticles and vaccine development. Expert Rev Vaccines 2015; 14:1197-211. [DOI: 10.1586/14760584.2015.1064772] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Jorge Alberto Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | - Omar González-Ortega
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | | |
Collapse
|
205
|
Zhang P, Chiu YC, Tostanoski LH, Jewell CM. Polyelectrolyte Multilayers Assembled Entirely from Immune Signals on Gold Nanoparticle Templates Promote Antigen-Specific T Cell Response. ACS NANO 2015; 9:6465-77. [PMID: 26035231 DOI: 10.1021/acsnano.5b02153] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Materials that allow modular, defined assembly of immune signals could support a new generation of rationally designed vaccines that promote tunable immune responses. Toward this goal, we have developed the first polyelectrolyte multilayer (PEM) coatings built entirely from immune signals. These immune-PEMs (iPEMs) are self-assembled on gold nanoparticle templates through stepwise electrostatic interactions between peptide antigen and polyanionic toll-like receptor (TLR) agonists that serve as molecular adjuvants. iPEMs do not require solvents or mixing, offer direct control over the composition and loading of vaccine components, and can be coated on substrates at any scale. These films also do not require other structural components, eliminating the potentially confounding effects caused by the inherent immune-stimulatory characteristics of many synthetic polymers. iPEM loading on gold nanoparticle substrates is tunable, and cryoTEM reveals iPEM shells coated on gold cores. These nanoparticles are efficiently internalized by primary dendritic cells (DCs), resulting in activation, selective triggering of TLR signaling, and presentation of the antigens used to assemble iPEMs. In coculture, iPEMs drive antigen-specific T cell proliferation and effector cytokines but not cytokines associated with more generalized inflammation. Compared to mice treated with soluble antigen and adjuvant, iPEM immunization promotes high levels of antigen-specific CD8(+) T cells in peripheral blood after 1 week. These enhancements result from increased DC activation and antigen presentation in draining lymph nodes. iPEM-immunized mice also exhibit a potent recall response after boosting, supporting the potential of iPEMs for designing well-defined vaccine coatings that provide high cargo density and eliminate synthetic film components.
Collapse
Affiliation(s)
- Peipei Zhang
- †Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Yu-Chieh Chiu
- †Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Lisa H Tostanoski
- †Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Christopher M Jewell
- †Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
- ‡Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
- §Marlene and Stewart Greenebaum Cancer Center, Baltimore, Maryland 21201, United States
| |
Collapse
|
206
|
Getts DR, Shea LD, Miller SD, King NJC. Harnessing nanoparticles for immune modulation. Trends Immunol 2015; 36:419-27. [PMID: 26088391 PMCID: PMC4603374 DOI: 10.1016/j.it.2015.05.007] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/15/2015] [Accepted: 05/17/2015] [Indexed: 01/18/2023]
Abstract
NPs can be generated from numerous biocompatible compounds. Specific physiochemical characteristics can be manipulated to modulate the immune response. Severe inflammation can be treated using NP-based approaches. Antigen delivery via NPs can restore peripheral immune tolerance.
Recent approaches using nanoparticles engineered for immune regulation have yielded promising results in preclinical models of disease. The number of nanoparticle therapies is growing, fueled by innovations in nanotechnology and advances in understanding of the underlying pathogenesis of immune-mediated diseases. In particular, recent mechanistic insight into the ways in which nanoparticles interact with the mononuclear phagocyte system and impact its function during homeostasis and inflammation have highlighted the potential of nanoparticle-based therapies for controlling severe inflammation while concurrently restoring peripheral immune tolerance in autoimmune disease. Here we review recent advances in nanoparticle-based approaches aimed at immune-modulation, and discuss these in the context of concepts in polymeric nanoparticle development, including particle modification, delivery and the factors associated with successful clinical deployment.
Collapse
Affiliation(s)
- Daniel R Getts
- The Discipline of Pathology, School of Medical Sciences, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Cour Pharmaceutical Development Company, Elmhurst, IL, USA.
| | - Lonnie D Shea
- Department of Chemical and Biomedical Engineering, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nicholas J C King
- The Discipline of Pathology, School of Medical Sciences, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
207
|
Carambia A, Freund B, Schwinge D, Bruns OT, Salmen SC, Ittrich H, Reimer R, Heine M, Huber S, Waurisch C, Eychmüller A, Wraith DC, Korn T, Nielsen P, Weller H, Schramm C, Lüth S, Lohse AW, Heeren J, Herkel J. Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice. J Hepatol 2015; 62:1349-56. [PMID: 25617499 DOI: 10.1016/j.jhep.2015.01.006] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/19/2014] [Accepted: 01/05/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS It is well-known that the liver can induce immune tolerance, yet this knowledge could, thus far, not be translated into effective treatments for autoimmune diseases. We have previously shown that liver sinusoidal endothelial cells (LSECs) could substantially contribute to hepatic tolerance through their ability to induce CD4+ Foxp3+ regulatory T cells (Tregs). Here, we explored whether the Treg-inducing potential of LSECs could be harnessed for the treatment of autoimmune disease. METHODS We engineered a polymeric nanoparticle (NP) carrier for the selective delivery of autoantigen peptides to LSECs in vivo. In the well-characterized autoimmune disease model of experimental autoimmune encephalomyelitis (EAE), we investigated whether administration of LSEC-targeting autoantigen peptide-loaded NPs could protect mice from autoimmune disease. RESULTS We demonstrate that NP-based autoantigen delivery to LSECs could completely and permanently prevent the onset of clinical EAE. More importantly, in a therapeutic approach, mice with already established EAE improved rapidly and substantially following administration of a single dose of autoantigen peptide-loaded NPs, whereas the control group deteriorated. Treatment efficacy seemed to depend on Tregs. The Treg frequencies in the spleens of mice treated with autoantigen peptide-loaded NPs were significantly higher than those in vehicle-treated mice. Moreover, NP-mediated disease control was abrogated after Treg depletion by repeated administration of Treg-depleting antibody. CONCLUSION Our findings provide proof of principle that the selective delivery of autoantigen peptides to LSECs by NPs can induce antigen-specific Tregs and enable effective treatment of autoimmune disease. These findings highlight the importance of Treg induction by LSECs for immune tolerance.
Collapse
Affiliation(s)
- Antonella Carambia
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Freund
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorothee Schwinge
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver T Bruns
- Department of Electron Microscopy and Micro Technology, Heinrich-Pette Institute, Hamburg, Germany
| | - Sunhild C Salmen
- Institute of Physical Chemistry, University of Hamburg, Hamburg, Germany
| | - Harald Ittrich
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rudolph Reimer
- Department of Electron Microscopy and Micro Technology, Heinrich-Pette Institute, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - David C Wraith
- Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Thomas Korn
- Department of Neurology, TU München, München, Germany
| | - Peter Nielsen
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Horst Weller
- Institute of Physical Chemistry, University of Hamburg, Hamburg, Germany
| | - Christoph Schramm
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Lüth
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Herkel
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
208
|
Gammon JM, Tostanoski LH, Adapa AR, Chiu YC, Jewell CM. Controlled delivery of a metabolic modulator promotes regulatory T cells and restrains autoimmunity. J Control Release 2015; 210:169-78. [PMID: 26002150 DOI: 10.1016/j.jconrel.2015.05.277] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022]
Abstract
Autoimmune disorders occur when the immune system abnormally recognizes and attacks self-molecules. Dendritic cells (DCs) play a powerful role in initiating adaptive immune response, and are therefore a recent target for autoimmune therapies. N-Phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC), a small molecule glutamate receptor enhancer, alters how DCs metabolize glutamate, skewing cytokine secretion to bias T cell function. These effects provide protection in mouse models of multiple sclerosis (MS) by polarizing T cells away from inflammatory TH17 cells and toward regulatory T cells (TREG) when mice receive daily systemic injections of PHCCC. However, frequent, continued treatment is required to generate and maintain therapeutic benefits. Thus, the use of PHCCC is limited by poor solubility, the need for frequent dosing, and cell toxicity. We hypothesized that controlled release of PHCCC from degradable nanoparticles (NPs) might address these challenges by altering DC function to maintain efficacy with reduced treatment frequency and toxicity. This idea could serve as a new strategy for harnessing biomaterials to polarize immune function through controlled delivery of metabolic modulators. PHCCC was readily encapsulated in nanoparticles, with controlled release of 89% of drug into media over three days. Culture of primary DCs or DC and T cell co-cultures with PHCCC NPs reduced DC activation and secretion of pro-inflammatory cytokines, while shifting T cells away from TH17 and toward TREG phenotypes. Importantly, PHCCC delivered to cells in NPs was 36-fold less toxic compared with soluble PHCCC. Treatment of mice with PHCCC NPs every three days delayed disease onset and decreased disease severity compared with mice treated with soluble drug at the same dose and frequency. These results highlight the potential to promote tolerance through controlled delivery of metabolic modulators that alter DC signaling to polarize T cells, and suggest future gains that could be realized by engineering materials that provide longer term release.
Collapse
Affiliation(s)
- Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Arjun R Adapa
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yu-Chieh Chiu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, United States; Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, United States.
| |
Collapse
|
209
|
Therapeutic applications of nanomedicine in autoimmune diseases: From immunosuppression to tolerance induction. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1003-18. [DOI: 10.1016/j.nano.2014.12.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 01/13/2023]
|
210
|
Lewis JS, Dolgova NV, Zhang Y, Xia CQ, Wasserfall CH, Atkinson MA, Clare-Salzler MJ, Keselowsky BG. A combination dual-sized microparticle system modulates dendritic cells and prevents type 1 diabetes in prediabetic NOD mice. Clin Immunol 2015; 160:90-102. [PMID: 25842187 DOI: 10.1016/j.clim.2015.03.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/16/2015] [Accepted: 03/24/2015] [Indexed: 12/17/2022]
Abstract
We developed a novel poly(lactic-co-glycolic acid)-based, microparticle (MP) system providing concurrent delivery of multiple encapsulated immuno-suppressive factors and antigen, for in vivo conditioning of dendritic cells (DCs) toward a tolerance promoting pathway. Subcutaneous administration prevents onset of type 1 diabetes (T1D) in NOD mice. Two MP sizes were made: phagocytosable MPs were fabricated encapsulating vitamin D3 or insulin B(9-23) peptide, while unphagocytosable MPs were fabricated encapsulating TGF-β1 or GM-CSF. The combination of Vit D3/TGF-β1 MPs confers an immature and LPS activation-resistant phenotype to DCs, and MP-delivered antigen is efficiently and functionally presented. Notably, two subcutaneous injections into 4week old NOD mice using the combination of MPs encapsulating Vit D3, Ins B, TGF-β1 and GM-CSF protected 40% of mice from T1D development, significant in comparison to the control. This work represents one of the first applications of a biomaterial-based, MP vaccine system to successfully prevent autoimmune diabetes.
Collapse
Affiliation(s)
- Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Natalia V Dolgova
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ying Zhang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Chang Qing Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
211
|
Serra P, Santamaria P. Nanoparticle-based autoimmune disease therapy. Clin Immunol 2015; 160:3-13. [PMID: 25704658 DOI: 10.1016/j.clim.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 01/10/2023]
Abstract
The goal of immunotherapy against autoimmunity is to block pathogenic inflammation without impairing immunity against infections and tumours. Regulatory T-cells (Tregs) play a central role in maintaining immune homeostasis, and autoimmune inflammation is frequently associated with decreased numbers and/or function of these T-cells. Therapies harnessing Tregs to treat autoimmune inflammation remain under-developed with caveats ranging from the lack of antigenic and disease specificity to the potential phenotypic and functional instability of in vitro-expanded Treg cells in vivo. Here, we review nanotechnology-based approaches designed to promote immune tolerance through various mechanisms, ranging from systemic or local suppression of antigen-presenting cells and deletion of antigen-specific T-cells, to the systemic expansion of antigen- and disease-specific Treg cells in vivo.
Collapse
Affiliation(s)
- Pau Serra
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain.
| | - Pere Santamaria
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain; Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cummings School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
212
|
Hartwell BL, Antunez L, Sullivan BP, Thati S, Sestak JO, Berkland C. Multivalent Nanomaterials: Learning from Vaccines and Progressing to Antigen-Specific Immunotherapies. J Pharm Sci 2015; 104:346-61. [DOI: 10.1002/jps.24273] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/26/2014] [Accepted: 10/28/2014] [Indexed: 12/28/2022]
|
213
|
Gendelman HE, Anantharam V, Bronich T, Ghaisas S, Jin H, Kanthasamy AG, Liu X, McMillan J, Mosley RL, Narasimhan B, Mallapragada SK. Nanoneuromedicines for degenerative, inflammatory, and infectious nervous system diseases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:751-67. [PMID: 25645958 DOI: 10.1016/j.nano.2014.12.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 12/01/2022]
Abstract
Interest in nanoneuromedicine has grown rapidly due to the immediate need for improved biomarkers and therapies for psychiatric, developmental, traumatic, inflammatory, infectious and degenerative nervous system disorders. These, in whole or in part, are a significant societal burden due to growth in numbers of affected people and in disease severity. Lost productivity of the patient and his or her caregiver, and the emotional and financial burden cannot be overstated. The need for improved health care, treatment and diagnostics is immediate. A means to such an end is nanotechnology. Indeed, recent developments of health-care enabling nanotechnologies and nanomedicines range from biomarker discovery including neuroimaging to therapeutic applications for degenerative, inflammatory and infectious disorders of the nervous system. This review focuses on the current and future potential of the field to positively affect clinical outcomes. From the clinical editor: Many nervous system disorders remain unresolved clinical problems. In many cases, drug agents simply cannot cross the blood-brain barrier (BBB) into the nervous system. The advent of nanomedicines can enhance the delivery of biologically active molecules for targeted therapy and imaging. This review focused on the use of nanotechnology for degenerative, inflammatory, and infectious diseases in the nervous system.
Collapse
Affiliation(s)
- Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA.
| | | | - Tatiana Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Iowa State University, Ames, IA USA
| | - Huajun Jin
- Department of Biomedical Sciences, Iowa State University, Ames, IA USA
| | | | - Xinming Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA USA
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA USA.
| |
Collapse
|
214
|
Ballerini C, Baldi G, Aldinucci A, Maggi P. Nanomaterial applications in multiple sclerosis inflamed brain. J Neuroimmune Pharmacol 2015; 10:1-13. [PMID: 25616566 DOI: 10.1007/s11481-015-9588-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/14/2015] [Indexed: 12/22/2022]
Abstract
In the last years scientific progress in nanomaterials, where size and shape make the difference, has increased their utilization in medicine with the development of a promising new translational science: nanomedicine. Due to their surface and core biophysical properties, nanomaterials hold the promise for medical applications in central nervous system (CNS) diseases: inflammatory, degenerative and tumors. The present review is focused on nanomaterials at the neuro-immune interface, evaluating two aspects: the possible CNS inflammatory response induced by nanomaterials and the developments of nanomaterials to improve treatment and diagnosis of neuroinflammatory diseases, with a focus on multiple sclerosis (MS). Indeed, nanomedicine allows projecting new ways of drug delivery and novel techniques for CNS imaging. Despite the wide field of application in neurological diseases of nanomaterials, our topic here is to review the more recent development of nanomaterials that cross blood brain barrier (BBB) and reach specific target during CNS inflammatory diseases, a crucial strategy for CNS early diagnosis and drug delivery, indeed the main challenges of nanomedicine.
Collapse
Affiliation(s)
- Clara Ballerini
- Department of Neurofarba, University of Florence, Viale Pieraccini, 6, 50137, Florence, Italy,
| | | | | | | |
Collapse
|
215
|
Esser C, Rannug A. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev 2015; 67:259-79. [PMID: 25657351 DOI: 10.1124/pr.114.009001] [Citation(s) in RCA: 377] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is an evolutionarily old transcription factor belonging to the Per-ARNT-Sim-basic helix-loop-helix protein family. AhR translocates into the nucleus upon binding of various small molecules into the pocket of its single-ligand binding domain. AhR binding to both xenobiotic and endogenous ligands results in highly cell-specific transcriptome changes and in changes in cellular functions. We discuss here the role of AhR for immune cells of the barrier organs: skin, gut, and lung. Both adaptive and innate immune cells require AhR signaling at critical checkpoints. We also discuss the current two prevailing views-namely, 1) AhR as a promiscuous sensor for small chemicals and 2) a role for AhR as a balancing factor for cell differentiation and function, which is controlled by levels of endogenous high-affinity ligands. AhR signaling is considered a promising drug and preventive target, particularly for cancer, inflammatory, and autoimmune diseases. Therefore, understanding its biology is of great importance.
Collapse
Affiliation(s)
- Charlotte Esser
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany (C.E.); and Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.R.)
| | - Agneta Rannug
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany (C.E.); and Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (A.R.)
| |
Collapse
|
216
|
Sriramoju B, Kanwar RK, Kanwar JR. Neurobehavioral burden of multiple sclerosis with nanotheranostics. Neuropsychiatr Dis Treat 2015; 11:2675-89. [PMID: 26508863 PMCID: PMC4610886 DOI: 10.2147/ndt.s82768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating neurological disorder affecting people worldwide; women are affected more than men. MS results in serious neurological deficits along with behavioral compromise, the mechanisms of which still remain unclear. Behavioral disturbances such as depression, anxiety, cognitive impairment, psychosis, euphoria, sleep disturbances, and fatigue affect the quality of life in MS patients. Among these, depression and psychosis are more common than any other neurological disorders. In addition, depression is associated with other comorbidities. Although anxiety is often misdiagnosed in MS patients, it can induce suicidal ideation if it coexists with depression. An interrelation between sleep abnormalities and fatigue is also reported among MS patients. In addition, therapeutics for MS is always a challenge because of the presence of the blood-brain barrier, adding to the lack of detailed understanding of the disease pathology. In this review, we tried to summarize various behavioral pathologies and their association with MS, followed by its conventional treatment and nanotheranostics.
Collapse
Affiliation(s)
- Bhasker Sriramoju
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine, Molecular and Medical Research, Faculty of Health, Deakin University, VIC, Australia
| | - Rupinder K Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine, Molecular and Medical Research, Faculty of Health, Deakin University, VIC, Australia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), School of Medicine, Molecular and Medical Research, Faculty of Health, Deakin University, VIC, Australia
| |
Collapse
|
217
|
Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci U S A 2014; 112:E156-65. [PMID: 25548186 DOI: 10.1073/pnas.1408686111] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current treatments to control pathological or unwanted immune responses often use broadly immunosuppressive drugs. New approaches to induce antigen-specific immunological tolerance that control both cellular and humoral immune responses are desirable. Here we describe the use of synthetic, biodegradable nanoparticles carrying either protein or peptide antigens and a tolerogenic immunomodulator, rapamycin, to induce durable and antigen-specific immune tolerance, even in the presence of potent Toll-like receptor agonists. Treatment with tolerogenic nanoparticles results in the inhibition of CD4+ and CD8+ T-cell activation, an increase in regulatory cells, durable B-cell tolerance resistant to multiple immunogenic challenges, and the inhibition of antigen-specific hypersensitivity reactions, relapsing experimental autoimmune encephalomyelitis, and antibody responses against coagulation factor VIII in hemophilia A mice, even in animals previously sensitized to antigen. Only encapsulated rapamycin, not the free form, could induce immunological tolerance. Tolerogenic nanoparticle therapy represents a potential novel approach for the treatment of allergies, autoimmune diseases, and prevention of antidrug antibodies against biologic therapies.
Collapse
|
218
|
Andorko JI, Hess KL, Jewell CM. Harnessing biomaterials to engineer the lymph node microenvironment for immunity or tolerance. AAPS JOURNAL 2014; 17:323-38. [PMID: 25533221 PMCID: PMC4365095 DOI: 10.1208/s12248-014-9708-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 11/27/2014] [Indexed: 01/06/2023]
Abstract
Nanoparticles, microparticles, and other biomaterials are advantageous in vaccination because these materials provide opportunities to modulate specific characteristics of immune responses. This idea of “tuning” immune responses has recently been used to combat infectious diseases and cancer, and to induce tolerance during organ transplants or autoimmune disease. Lymph nodes and other secondary lymphoid organs such as the spleen play crucial roles in determining if and how these responses develop following vaccination or immunotherapy. Thus, by manipulating the local microenvironments within these immunological command centers, the nature of systemic immune response can be controlled. This review provides recent examples that harness the interactions between biomaterials and lymph nodes or other secondary lymphoid organs to generate immunity or promote tolerance. These strategies draw on mechanical properties, surface chemistry, stability, and targeting to alter the interactions of cells, signals, and vaccine components in lymph nodes. While there are still many unanswered questions surrounding how best to design biomaterial-based vaccines to promote specific structures or functions in lymph nodes, features such as controlled release and targeting will help pave the way for the next generation of vaccines and immunotherapies that generate immune responses tuned for specific applications.
Collapse
Affiliation(s)
- James I Andorko
- Fischell Department of Bioengineering, University of Maryland, 2212 Jeong H. Kim Engineering Building, College Park, Maryland, 20742, USA
| | | | | |
Collapse
|
219
|
Mansilla MJ, Sellès-Moreno C, Fàbregas-Puig S, Amoedo J, Navarro-Barriuso J, Teniente-Serra A, Grau-López L, Ramo-Tello C, Martínez-Cáceres EM. Beneficial effect of tolerogenic dendritic cells pulsed with MOG autoantigen in experimental autoimmune encephalomyelitis. CNS Neurosci Ther 2014; 21:222-30. [PMID: 25403984 DOI: 10.1111/cns.12342] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment with tolerogenic dendritic cells (TolDC) is a promising, cell-based strategy to regulate autoimmune diseases such as multiple sclerosis (MS) in an antigen-specific way. This technique involves the use of TolDC from MS patients cultured in the presence of vitamin D(3) (VitD3) and pulsed with myelin peptides to induce a stable hyporesponsiveness in myelin-specific autologous T cells. AIM The purpose of this study was to analyze the in vivo effect of VitD3-TolDC treatment on experimental autoimmune encephalomyelitis, an animal model of MS. METHODS Bone marrow-derived TolDC cultured in the presence of VitD3 and pulsed with peptide 40-55 of the myelin oligodendrocyte glycoprotein (MOG(40-55)) were administrated preventively, preclinically, and therapeutically to EAE-induced mice. RESULTS We found that VitD3-TolDC-MOG treatment showed a beneficial effect, not only decreasing the incidence of the disease but also reducing the severity of the clinical signs mediated by induction of regulatory T cells (Treg), as well as IL-10 production and reduction of Ag-specific lymphoproliferation. Our results support VitD3-TolDC-peptide(s) treatment as a potential strategy to restore tolerance in autoimmune diseases such as MS.
Collapse
Affiliation(s)
- María José Mansilla
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain; Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Quintana FJ, Patel B, Yeste A, Nyirenda M, Kenison J, Rahbari R, Fetco D, Hussain M, O'Mahony J, Magalhaes S, McGowan M, Johnson T, Rajasekharan S, Narayanan S, Arnold DL, Weiner HL, Banwell B, Bar-Or A. Epitope spreading as an early pathogenic event in pediatric multiple sclerosis. Neurology 2014; 83:2219-26. [PMID: 25381299 DOI: 10.1212/wnl.0000000000001066] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES For most adults with initial clinical presentation of multiple sclerosis (MS), biological disease was likely initiated many years prior. Pediatric-onset MS provides an opportunity to study early disease processes. METHODS Using antigen microarrays, including CNS-related proteins, lipids, and other autoantigens, we studied early immunologic events involved in clinical onset of pediatric MS. Serum samples were collected at the time of incident acquired CNS demyelinating syndromes (ADS) in children who, in subsequent prospective follow-up, were ascertained to have either pediatric MS (ADS-MS) or a monophasic illness (ADS-mono). Samples were obtained both at the time of ADS presentation and 3 months into follow-up. We used an initial training set of samples to implicate antibody signatures associated with each group, and then a test set. An additional set of follow-up samples (stability set) was used as a form of internal validation. RESULTS Children with ADS-MS tended to have distinguishable serum antibody patterns both at the time of ADS presentation and 3 months into follow-up. At the time of ADS, serum samples from patients with ADS-MS or ADS-mono reacted against similar numbers of CNS antigens, although CNS antigens implicated in adult MS were more often targeted in children with ADS-MS. The follow-up ADS-MS samples reacted against a broader panel of CNS antigens, while corresponding ADS-mono samples exhibited a contraction of the initial antibody response. CONCLUSIONS Our findings in this prospective cohort of pediatric-onset CNS demyelinating diseases point to an active process of epitope spreading during early stages of MS, not seen in monophasic CNS inflammatory conditions.
Collapse
Affiliation(s)
- Francisco J Quintana
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Bonny Patel
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Ada Yeste
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Mukanthu Nyirenda
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Jessica Kenison
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Roya Rahbari
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Dumitru Fetco
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Mohammad Hussain
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Julia O'Mahony
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sandra Magalhaes
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Melissa McGowan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Trina Johnson
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sathy Rajasekharan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Sridar Narayanan
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Douglas L Arnold
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Howard L Weiner
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Brenda Banwell
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA
| | - Amit Bar-Or
- From the Center for Neurologic Diseases (F.J.Q., B.P., A.Y., J.K., R.R., M.H., H.L.W.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Neuroimmunology Unit (M.N., S.M., A.B.-O.) and the McConnell Brain Imaging Centre (D.F., S.N., D.L.A.), Montreal Neurological Institute and Hospital, McGill University, Montreal; Hospital for Sick Children (J.O., M.M., B.B.) and the Institute of Health Policy, Management, and Evaluation (J.O.), University of Toronto; Experimental Therapeutics Program (T.J., S.R., A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; and The Children's Hospital of Philadelphia (B.B.), PA.
| | | |
Collapse
|
221
|
Wei P, Hu GH, Kang HY, Yao HB, Kou W, Zhang C, Hong SL. Role of the aryl hydrocarbon receptor in the pathogenesis of chronic rhinosinusitis with nasal polyps. Inflammation 2014; 37:387-95. [PMID: 24092408 DOI: 10.1007/s10753-013-9751-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A predominant Th17 population is a marker of chronic rhinosinusitis with nasal polyps (CRSwNP) in Chinese patients. As a ligand-activated transcription factor, the aryl hydrocarbon receptor (AhR) plays a vital role in promoting or inhibiting specific Th cell development. However, its role in CRSwNP remains to be defined. The aim of the present study was to investigate whether AhR, which regulates Th17 cell differentiation, played a role in the pathogenesis of CRSwNP by evaluating AhR expression in nasal polyps and peripheral blood mononuclear cells (PBMCs) obtained from CRSwNP patients. Forty-eight patients (atopic, 24; non-atopic, 24) and 13 controls were studied. To explore the role of AhR in CRSwNP, we analyzed the expression of AhR, retinoid-related orphan receptor C (RORC), interleukin (IL)-17, and IL-10 and the differentiation of Th17 using mRNA or protein detection methods. Notably, the expression of AhR was reduced in CRSwNP, and the expression of AhR was lower in the atopic group than in the non-atopic group. However, there was a very low level of Th17 and its associated factors (RORC, IL-17) in the control group compared to the two CRSwNP groups. In particular, the polarization of Th17 cells in atopic CRSwNP patients was increased compared with non-atopic individuals. In addition, ITE intervention in PBMCs promoted AhR expression and attenuated Th17 responses, demonstrating that AhR was more likely to suppress Th17 cells differentiation in Chinese CRSwNP patients. This information is valuable for obtaining a clear understanding of the pathogenesis of CRSwNP. Moreover, patients with atopic CRSwNP may exhibit reduced expression of AhR, leading to aggravation of the disproportionate distribution of Th17 cells in polyp tissues and PBMCs, thereby suggesting that atopic CRSwNP has a distinct pathogenesis from that of non-atopic CRSwNP.
Collapse
Affiliation(s)
- Ping Wei
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Yixueyuan Road 1#, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
222
|
Ilinskaya AN, Dobrovolskaia MA. Immunosuppressive and anti-inflammatory properties of engineered nanomaterials. Br J Pharmacol 2014; 171:3988-4000. [PMID: 24724793 PMCID: PMC4243973 DOI: 10.1111/bph.12722] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/24/2014] [Accepted: 04/03/2014] [Indexed: 12/24/2022] Open
Abstract
Nanoparticle interactions with various components of the immune system are determined by their physicochemical properties such as size, charge, hydrophobicity and shape. Nanoparticles can be engineered to either specifically target the immune system or to avoid immune recognition. Nevertheless, identifying their unintended impacts on the immune system and understanding the mechanisms of such accidental effects are essential for establishing a nanoparticle's safety profile. While immunostimulatory properties have been reviewed before, little attention in the literature has been given to immunosuppressive and anti-inflammatory properties. The purpose of this review is to fill this gap. We will discuss intended immunosuppression achieved by either nanoparticle engineering, or the use of nanoparticles to carry immunosuppressive or anti-inflammatory drugs. We will also review unintended immunosuppressive properties of nanoparticles per se and consider how such properties could be either beneficial or adverse.
Collapse
Affiliation(s)
- A N Ilinskaya
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research IncFrederick, MD, USA
| | - M A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research IncFrederick, MD, USA
| |
Collapse
|
223
|
Role and therapeutic value of dendritic cells in central nervous system autoimmunity. Cell Death Differ 2014; 22:215-24. [PMID: 25168240 DOI: 10.1038/cdd.2014.125] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/23/2014] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that control the generation of adaptive immunity. Consequently, DCs have a central role in the induction of protective immunity to pathogens and also in the pathogenic immune response responsible for the development and progression of autoimmune disorders. Thus the study of the molecular pathways that control DC development and function is likely to result in new strategies for the therapeutic manipulation of the immune response. In this review, we discuss the role and therapeutic value of DCs in autoimmune diseases, with a special focus on multiple sclerosis.
Collapse
|
224
|
Smith BR, Ghosn EEB, Rallapalli H, Prescher JA, Larson T, Herzenberg LA, Gambhir SS. Selective uptake of single-walled carbon nanotubes by circulating monocytes for enhanced tumour delivery. NATURE NANOTECHNOLOGY 2014; 9:481-7. [PMID: 24727688 PMCID: PMC4236538 DOI: 10.1038/nnano.2014.62] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 02/26/2014] [Indexed: 05/05/2023]
Abstract
In cancer imaging, nanoparticle biodistribution is typically visualized in living subjects using 'bulk' imaging modalities such as magnetic resonance imaging, computerized tomography and whole-body fluorescence. Accordingly, nanoparticle influx is observed only macroscopically, and the mechanisms by which they target cancer remain elusive. Nanoparticles are assumed to accumulate via several targeting mechanisms, particularly extravasation (leakage into tumour). Here, we show that, in addition to conventional nanoparticle-uptake mechanisms, single-walled carbon nanotubes are almost exclusively taken up by a single immune cell subset, Ly-6C(hi) monocytes (almost 100% uptake in Ly-6C(hi) monocytes, below 3% in all other circulating cells), and delivered to the tumour in mice. We also demonstrate that a targeting ligand (RGD) conjugated to nanotubes significantly enhances the number of single-walled carbon nanotube-loaded monocytes reaching the tumour (P < 0.001, day 7 post-injection). The remarkable selectivity of this tumour-targeting mechanism demonstrates an advanced immune-based delivery strategy for enhancing specific tumour delivery with substantial penetration.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Eliver Eid Bou Ghosn
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Harikrishna Rallapalli
- Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA
| | - Jennifer A Prescher
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2]
| | - Timothy Larson
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Sanjiv Sam Gambhir
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA [3] Department of Bioengineering and Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
225
|
Purwada A, Roy K, Singh A. Engineering vaccines and niches for immune modulation. Acta Biomater 2014; 10:1728-40. [PMID: 24373907 DOI: 10.1016/j.actbio.2013.12.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/16/2013] [Accepted: 12/18/2013] [Indexed: 12/14/2022]
Abstract
Controlled modulation of immune response, especially the balance between immunostimulatory and immunosuppressive responses, is critical for a variety of clinical applications, including immunotherapies against cancer and infectious diseases, treatment of autoimmune disorders, transplant surgeries, regenerative medicine, prosthetic implants, etc. Our ability to precisely modify both innate and adaptive immune responses could provide new therapeutic directions in a variety of diseases. In the context of vaccines and immunotherapies, the interplay between antigen-presenting cells (e.g. dendritic cells and macrophages), B cells, T helper and killer subtypes, and regulatory T- and B-cell responses is critical for generating effective immunity against cancer, infectious diseases and autoimmune diseases. In recent years, immunoengineering has emerged as a new field that uses quantitative engineering tools to understand molecular-, cellular- and system-level interactions of the immune system and to develop design-driven approaches to control and modulate immune responses. Biomaterials are an integral part of this engineering toolbox and can exploit the intrinsic biological and mechanical cues of the immune system to directly modulate and train immune cells and direct their response to a particular phenotype. A large body of literature exists on strategies to evade or suppress the immune response in implants, transplantation and regenerative medicine. This review specifically focuses on the use of biomaterials for immunostimulation and controlled modulation, especially in the context of vaccines and immunotherapies against cancer, infectious diseases and autoimmune disorders. Bioengineering smart systems that can simultaneously deliver multiple bioactive agents in a controlled manner or can work as a niche for in situ priming and modulation of the immune system could significantly enhance the efficacy of next-generation immunotherapeutics. In this review, we describe our perspective on the important design aspects for the development of biomaterials that can actively modulate immune responses by stimulating receptor complexes and cells, and delivering multiple immunomodulatory biomolecules.
Collapse
|
226
|
Hunter Z, McCarthy DP, Yap WT, Harp CT, Getts DR, Shea LD, Miller SD. A biodegradable nanoparticle platform for the induction of antigen-specific immune tolerance for treatment of autoimmune disease. ACS NANO 2014; 8:2148-60. [PMID: 24559284 PMCID: PMC3990004 DOI: 10.1021/nn405033r] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Targeted immune tolerance is a coveted therapy for the treatment of a variety of autoimmune diseases, as current treatment options often involve nonspecific immunosuppression. Intravenous (iv) infusion of apoptotic syngeneic splenocytes linked with peptide or protein autoantigens using ethylene carbodiimide (ECDI) has been demonstrated to be an effective method for inducing peripheral, antigen-specific tolerance for treatment of autoimmune disease. Here, we show the ability of biodegradable poly(lactic-co-glycolic acid) (PLG) nanoparticles to function as a safe, cost-effective, and highly efficient alternative to cellular carriers for the induction of antigen-specific T cell tolerance. We describe the formulation of tolerogenic PLG particles and demonstrate that administration of myelin antigen-coupled particles both prevented and treated relapsing-remitting experimental autoimmune encephalomyelitis (R-EAE), a CD4 T cell-mediated mouse model of multiple sclerosis (MS). PLG particles made on-site with surfactant modifications surpass the efficacy of commercially available particles in their ability to couple peptide and to prevent disease induction. Most importantly, myelin antigen-coupled PLG nanoparticles are able to significantly ameliorate ongoing disease and subsequent relapses when administered at onset or at peak of acute disease, and minimize epitope spreading when administered during disease remission. Therapeutic treatment results in significantly reduced CNS infiltration of encephalitogenic Th1 (IFN-γ) and Th17 (IL-17a) cells as well as inflammatory monocytes/macrophages. Together, these data describe a platform for antigen display that is safe, low-cost, and highly effective at inducing antigen-specific T cell tolerance. The development of such a platform carries broad implications for the treatment of a variety of immune-mediated diseases.
Collapse
Affiliation(s)
- Zoe Hunter
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 East Chicago Avenue, Chicago, Illinois 60611, United States
| | - Derrick P. McCarthy
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 East Chicago Avenue, Chicago, Illinois 60611, United States
| | - Woon Teck Yap
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Christopher T. Harp
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 East Chicago Avenue, Chicago, Illinois 60611, United States
| | - Daniel R. Getts
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 East Chicago Avenue, Chicago, Illinois 60611, United States
| | - Lonnie D. Shea
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Address correspondence to
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 6-713 Tarry Building, 303 East Chicago Avenue, Chicago, Illinois 60611, United States
- Address correspondence to
| |
Collapse
|
227
|
McCarthy DP, Hunter ZN, Chackerian B, Shea LD, Miller SD. Targeted immunomodulation using antigen-conjugated nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 6:298-315. [PMID: 24616452 DOI: 10.1002/wnan.1263] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/23/2014] [Accepted: 02/01/2014] [Indexed: 12/20/2022]
Abstract
The growing prevalence of nanotechnology in the fields of biology, medicine, and the pharmaceutical industry is confounded by the relatively small amount of data on the impact of these materials on the immune system. In addition to concerns surrounding the potential toxicity of nanoparticle (NP)-based delivery systems, there is also a demand for a better understanding of the mechanisms governing interactions of NPs with the immune system. Nanoparticles can be tailored to suppress, enhance, or subvert recognition by the immune system. This 'targeted immunomodulation' can be achieved by delivery of unmodified particles, or by modifying particles to deliver drugs, proteins/peptides, or genes to a specific site. In order to elicit the desired, beneficial immune response, considerations should be made at every step of the design process: the NP platform itself, ligands, and other modifiers, the delivery route, and the immune cells that will encounter the conjugated NPs can all impact host immune responses.
Collapse
Affiliation(s)
- Derrick P McCarthy
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
228
|
Fazio F, Zappulla C, Notartomaso S, Busceti C, Bessede A, Scarselli P, Vacca C, Gargaro M, Volpi C, Allegrucci M, Lionetto L, Simmaco M, Belladonna ML, Nicoletti F, Fallarino F. Cinnabarinic acid, an endogenous agonist of type-4 metabotropic glutamate receptor, suppresses experimental autoimmune encephalomyelitis in mice. Neuropharmacology 2014; 81:237-43. [PMID: 24565643 DOI: 10.1016/j.neuropharm.2014.02.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 02/06/2014] [Accepted: 02/13/2014] [Indexed: 01/18/2023]
Abstract
Cinnabarinic acid (CA) is an endogenous metabolite of the kynurenine pathway which acts as an orthosteric agonist of type-4 metabotropic glutamate receptor (mGlu4). We now report that systemic administration of CA (0.1-10 mg/kg, i.p.) was highly protective against experimental autoimmune encephalomyelitis (EAE) induced by the myelin oligodendrocyte glycoprotein (MOG35-55) peptide, which models multiple sclerosis in mice. Full protection against EAE required daily injections of CA since the time of immunization, similarly to what reported for the mGlu4 enhancer N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1acarboxamide (PHCCC). CA treatment boosted an immune response dominated by regulatory T (Treg) cells at the expenses of Th17 cells. In addition, exogenous CA enhanced endogenous CA formation in lymphocytes, suggesting the occurrence of a positive feedback loop sustaining immune tolerance. To examine whether activation of mGlu4 could account for the protective activity of CA against EAE, we used mGlu4 knockout mice. As expected, these mice displayed a more severe form of EAE in response to immunization. CA was still protective against EAE in mGlu4-deficient mice, although its action was significantly reduced both at high and low CA doses. This suggests that the action of CA against neuroinflammation involves multiple mechanisms including the activation of mGlu4. These data further suggest that CA is one possible bridge between activation of the kynurenine pathway and immune tolerance aimed at restraining neuroinflammation.
Collapse
Affiliation(s)
- Francesco Fazio
- I.R.C.C.S. Neuromed, Pozzilli, Parco Tecnologico, Località Camerelle, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Cristina Zappulla
- I.R.C.C.S. Neuromed, Pozzilli, Parco Tecnologico, Località Camerelle, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Serena Notartomaso
- I.R.C.C.S. Neuromed, Pozzilli, Parco Tecnologico, Località Camerelle, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Carla Busceti
- I.R.C.C.S. Neuromed, Pozzilli, Parco Tecnologico, Località Camerelle, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | | | - Pamela Scarselli
- I.R.C.C.S. Neuromed, Pozzilli, Parco Tecnologico, Località Camerelle, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Massimo Allegrucci
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Luana Lionetto
- NESMOS Department, Advanced Molecular Diagnostic Unit, Sant'Andrea Hospital, University of Rome 'Sapienza', Via di Grotta Rossa 1035-39, 00189 Rome, Italy
| | - Maurizio Simmaco
- NESMOS Department, Advanced Molecular Diagnostic Unit, Sant'Andrea Hospital, University of Rome 'Sapienza', Via di Grotta Rossa 1035-39, 00189 Rome, Italy
| | - Maria Laura Belladonna
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Ferdinando Nicoletti
- Department of Human Physiology and Pharmacology, University of Rome 'Sapienza', Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Francesca Fallarino
- Department of Experimental Medicine, University of Perugia, Polo Didattico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy.
| |
Collapse
|
229
|
Punj S, Kopparapu P, Jang HS, Phillips JL, Pennington J, Rohlman D, O’Donnell E, Iversen PL, Kolluri SK, Kerkvliet NI. Benzimidazoisoquinolines: a new class of rapidly metabolized aryl hydrocarbon receptor (AhR) ligands that induce AhR-dependent Tregs and prevent murine graft-versus-host disease. PLoS One 2014; 9:e88726. [PMID: 24586378 PMCID: PMC3929365 DOI: 10.1371/journal.pone.0088726] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/09/2014] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that plays multiple roles in regulation of immune and inflammatory responses. The ability of certain AhR ligands to induce regulatory T cells (Tregs) has generated interest in developing AhR ligands for therapeutic treatment of immune-mediated diseases. To this end, we designed a screen for novel Treg-inducing compounds based on our understanding of the mechanisms of Treg induction by the well-characterized immunosuppressive AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). We screened a ChemBridge small molecule library and identified 10-chloro-7H-benzimidazo[2,1-a]benzo[de]Iso-quinolin-7-one (10-Cl-BBQ) as a potent AhR ligand that was rapidly metabolized and not cytotoxic to proliferating T cells. Like TCDD,10-Cl-BBQ altered donor CD4+ T cell differentiation during the early stages of a graft versus host (GVH) response resulting in expression of high levels of CD25, CTLA-4 and ICOS, as well as several genes associated with Treg function. The Treg phenotype required AhR expression in the donor CD4+ T cells. Foxp3 was not expressed in the AhR-induced Tregs implicating AhR as an independent transcription factor for Treg induction. Structure-activity studies showed that unsubstituted BBQ as well as 4, 11-dichloro-BBQ were capable of inducing AhR-Tregs. Other substitutions reduced activation of AhR. Daily treatment with 10-Cl-BBQ during the GVH response prevented development of GVH disease in an AhR-dependent manner with no overt toxicity. Together, our data provide strong support for development of select BBQs that activate the AhR to induce Tregs for treatment of immune-mediated diseases.
Collapse
Affiliation(s)
- Sumit Punj
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Prasad Kopparapu
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Hyo Sang Jang
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Jessica L. Phillips
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Jamie Pennington
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Diana Rohlman
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Edmond O’Donnell
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Patrick L. Iversen
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
| | - Siva Kumar Kolluri
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, United States of America
| | - Nancy I. Kerkvliet
- Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, Oregon, United States of America
- Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
230
|
Peine KJ, Guerau-de-Arellano M, Lee P, Kanthamneni N, Severin M, Probst GD, Peng H, Yang Y, Vangundy Z, Papenfuss TL, Lovett-Racke AE, Bachelder EM, Ainslie KM. Treatment of experimental autoimmune encephalomyelitis by codelivery of disease associated Peptide and dexamethasone in acetalated dextran microparticles. Mol Pharm 2014; 11:828-35. [PMID: 24433027 PMCID: PMC3993881 DOI: 10.1021/mp4005172] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system that can cause loss of motor function and is thought to result, in part, from chronic inflammation due to an antigen-specific T cell immune response. Current treatments suppress the immune system without antigen specificity, increasing the risks of cancer, chronic infection, and other long-term side effects. In this study, we show treatment of experimental autoimmune encephalomyelitis (EAE), a model of MS, by coencapsulating the immunodominant peptide of myelin oligodendrocyte glycoprotein (MOG) with dexamethasone (DXM) into acetalated dextran (Ac-DEX) microparticles (DXM/MOG/MPs) and administering the microparticles subcutaneously. The clinical score of the mice was reduced from 3.4 to 1.6 after 3 injections 3 days apart with the coencapsulated microparticulate formulation (MOG 17.6 μg and DXM 8 μg). This change in clinical score was significantly greater than observed with phosphate-buffered saline (PBS), empty MPs, free DXM and MOG, DXM/MPs, and MOG/MPs. Additionally, treatment with DXM/MOG/MPs significantly inhibited disease-associated cytokine (e.g., IL-17, GM-CSF) expression in splenocytes isolated in treated mice. Here we show a promising approach for the therapeutic treatment of MS using a polymer-based microparticle delivery platform.
Collapse
Affiliation(s)
- Kevin J Peine
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University , Columbus, Ohio, 43210, United States
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Clemente-Casares X, Santamaria P. Nanomedicine in autoimmunity. Immunol Lett 2014; 158:167-74. [PMID: 24406504 DOI: 10.1016/j.imlet.2013.12.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 12/09/2013] [Accepted: 12/20/2013] [Indexed: 11/15/2022]
Abstract
The application of nanotechnology to the diagnosis and therapy of human diseases is already a reality and is causing a real revolution in how we design new therapies and vaccines. In this review we focus on the applications of nanotechnology in the field of autoimmunity. First, we review scenarios in which iron oxide nanoparticles have been used in the diagnosis of autoimmune diseases, mostly through magnetic resonance imaging (MRI), both in animal models and patients. Second, we discuss the potential of nanoparticles as an immunotherapeutic platform for autoimmune diseases, for now exclusively in pre-clinical models. Finally, we discuss the potential of this field to generate the 'perfect drug' with the capacity to report on its therapeutic efficacy in real time, that is, the birth of theranostics in autoimmunity.
Collapse
Affiliation(s)
- Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada; Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona, Spain.
| |
Collapse
|
232
|
Toward understanding the role of aryl hydrocarbon receptor in the immune system: current progress and future trends. BIOMED RESEARCH INTERNATIONAL 2014; 2014:520763. [PMID: 24527450 PMCID: PMC3914515 DOI: 10.1155/2014/520763] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/14/2013] [Indexed: 01/03/2023]
Abstract
The immune system is regulated by distinct signaling pathways that control the development and function of the immune cells. Accumulating evidence suggest that ligation of aryl hydrocarbon receptor (Ahr), an environmentally responsive transcription factor, results in multiple cross talks that are capable of modulating these pathways and their downstream responsive genes. Most of the immune cells respond to such modulation, and many inflammatory response-related genes contain multiple xenobiotic-responsive elements (XREs) boxes upstream. Active research efforts have investigated the physiological role of Ahr in inflammation and autoimmunity using different animal models. Recently formed paradigm has shown that activation of Ahr by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 3,3′-diindolylmethane (DIM) prompts the differentiation of CD4+Foxp3+ regulatory T cells (Tregs) and inhibits T helper (Th)-17 suggesting that Ahr is an innovative therapeutic strategy for autoimmune inflammation. These promising findings generate a basis for future clinical practices in humans. This review addresses the current knowledge on the role of Ahr in different immune cell compartments, with a particular focus on inflammation and autoimmunity.
Collapse
|
233
|
Quintana FJ. Regulation of central nervous system autoimmunity by the aryl hydrocarbon receptor. Semin Immunopathol 2013; 35:627-35. [PMID: 23999753 PMCID: PMC3819215 DOI: 10.1007/s00281-013-0397-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 08/22/2013] [Indexed: 12/22/2022]
Abstract
The ligand-activated transcription factor aryl hydrocarbon receptor controls the activity of several components of the immune system, many of which play an important role in neuroinflammation. This review discusses the role of AhR in T cells and dendritic cells, its relevance for the control of autoimmunity in the central nervous system, and its potential as a therapeutic target for immune-mediated disorders.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA,
| |
Collapse
|
234
|
Irvine DJ, Swartz MA, Szeto GL. Engineering synthetic vaccines using cues from natural immunity. NATURE MATERIALS 2013; 12:978-90. [PMID: 24150416 PMCID: PMC3928825 DOI: 10.1038/nmat3775] [Citation(s) in RCA: 462] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 09/09/2013] [Indexed: 05/17/2023]
Abstract
Vaccines aim to protect against or treat diseases through manipulation of the immune response, promoting either immunity or tolerance. In the former case, vaccines generate antibodies and T cells poised to protect against future pathogen encounter or attack diseased cells such as tumours; in the latter case, which is far less developed, vaccines block pathogenic autoreactive T cells and autoantibodies that target self tissue. Enormous challenges remain, however, as a consequence of our incomplete understanding of human immunity. A rapidly growing field of research is the design of vaccines based on synthetic materials to target organs, tissues, cells or intracellular compartments; to co-deliver immunomodulatory signals that control the quality of the immune response; or to act directly as immune regulators. There exists great potential for well-defined materials to further our understanding of immunity. Here we describe recent advances in the design of synthetic materials to direct immune responses, highlighting successes and challenges in prophylactic, therapeutic and tolerance-inducing vaccines.
Collapse
Affiliation(s)
- Darrell J. Irvine
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Department of Biological Engineering, MIT, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, United States
- The Ragon Institute of MGH, MIT, and Harvard, East 149 13th Street, Charlestown, MA 02129, United States
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815, United States
| | - Melody A. Swartz
- Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Gregory L. Szeto
- Department of Materials Science and Engineering, Massachusetts Institute of Technology (MIT), 77 Massachusetts Avenue, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, United States
- The Ragon Institute of MGH, MIT, and Harvard, East 149 13th Street, Charlestown, MA 02129, United States
| |
Collapse
|
235
|
Kaufman JJ, Ottman R, Tao G, Shabahang S, Banaei EH, Liang X, Johnson SG, Fink Y, Chakrabarti R, Abouraddy AF. In-fiber production of polymeric particles for biosensing and encapsulation. Proc Natl Acad Sci U S A 2013; 110:15549-54. [PMID: 24019468 PMCID: PMC3785740 DOI: 10.1073/pnas.1310214110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Polymeric micro- and nanoparticles are becoming a mainstay in biomedicine, medical diagnostics, and therapeutics, where they are used in implementing sensing mechanisms, as imaging contrast agents, and in drug delivery. Current approaches to the fabrication of such particles are typically finely tuned to specific monomer or polymer species, size ranges, and structures. We present a general scalable methodology for fabricating uniformly sized spherical polymeric particles from a wide range of polymers produced with complex internal architectures and continuously tunable diameters extending from the millimeter scale down to 50 nm. Controllable access to such a wide range of sizes enables broad applications in cancer treatment, immunology, and vaccines. Our approach harnesses thermally induced, predictable fluid instabilities in composite core/cladding polymer fibers drawn from a macroscopic scaled-up model called a "preform." Through a stack-and-draw process, we produce fibers containing a multiplicity of identical cylindrical cores made of the polymers of choice embedded in a polymer cladding. The instability leads to the breakup of the initially intact cores, independent of the polymer chemistry, into necklaces of spherical particles held in isolation within the cladding matrix along the entire fiber length. We demonstrate here surface functionalization of the extracted particles for biodetection through specific protein-protein interactions, volumetric encapsulation of a biomaterial in spherical polymeric shells, and the combination of both surface and volumetric functionalities in the same particle. These particles used in distinct modalities may be produced from the desired biocompatible polymer by changing only the geometry of the macroscopic preform from which the fiber is drawn.
Collapse
Affiliation(s)
- Joshua J. Kaufman
- Center for Research and Education in Optics and Lasers (CREOL), The College of Optics and Photonics
| | - Richard Ottman
- Burnett School of Biomedical Sciences, College of Medicine, and
| | - Guangming Tao
- Center for Research and Education in Optics and Lasers (CREOL), The College of Optics and Photonics
| | - Soroush Shabahang
- Center for Research and Education in Optics and Lasers (CREOL), The College of Optics and Photonics
| | - Esmaeil-Hooman Banaei
- Department of Electrical Engineering and Computer Science, University of Central Florida, Orlando, FL 32816; and
| | | | | | - Yoel Fink
- Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Ayman F. Abouraddy
- Center for Research and Education in Optics and Lasers (CREOL), The College of Optics and Photonics
| |
Collapse
|
236
|
Mascanfroni ID, Yeste A, Vieira SM, Burns EJ, Patel B, Sloma I, Wu Y, Mayo L, Ben-Hamo R, Efroni S, Kuchroo VK, Robson SC, Quintana FJ. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat Immunol 2013; 14:1054-63. [PMID: 23995234 DOI: 10.1038/ni.2695] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 07/19/2013] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) control the balance between effector T cells and regulatory T cells in vivo. Hence, the study of DCs might identify mechanisms of disease pathogenesis and guide new therapeutic approaches for disorders mediated by the immune system. We found that interleukin 27 (IL-27) signaling in mouse DCs limited the generation of effector cells of the TH1 and TH17 subsets of helper T cells and the development of experimental autoimmune encephalomyelitis (EAE). The effects of IL-27 were mediated at least in part through induction of the immunoregulatory molecule CD39 in DCs. IL-27-induced CD39 decreased the extracellular concentration of ATP and downregulated nucleotide-dependent activation of the NLRP3 inflammasome. Finally, therapeutic vaccination with IL-27-conditioned DCs suppressed established relapsing-remitting EAE. Thus, IL-27 signaling in DCs limited pathogenic T cell responses and the development of autoimmunity.
Collapse
Affiliation(s)
- Ivan D Mascanfroni
- 1] Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA. [2]
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that belongs to the family of basic helix-loop-helix transcription factors. Although the AhR was initially recognized as the receptor mediating the pathologic effects of dioxins and other pollutants, the activation of AhR by endogenous and environmental factors has important physiologic effects, including the regulation of the immune response. Thus, the AhR provides a molecular pathway through which environmental factors modulate the immune response in health and disease. In this review, we discuss the role of AhR in the regulation of the immune response, the source and chemical nature of AhR ligands, factors controlling production and degradation of AhR ligands, and the potential to target the AhR for therapeutic immunomodulation.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
238
|
Hao N, Whitelaw ML. The emerging roles of AhR in physiology and immunity. Biochem Pharmacol 2013; 86:561-70. [PMID: 23856287 DOI: 10.1016/j.bcp.2013.07.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/04/2013] [Accepted: 07/04/2013] [Indexed: 02/06/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is traditionally defined as a transcriptional regulator involved in adaptive xenobiotic response, however, emerging evidence supports physiological functions of AhR in normal cell development and immune response. The role of AhR in immunomodulation is multi-dimensional. On the one hand, activation of AhR by TCDD and other ligands leads to profound immunosuppression, potentially via skewed Th1/Th2 cell balance toward Th1 dominance, and boosted Treg cell differentiation. On the other hand, activation of AhR can also induce Th17 cell polarization and increase the severity of autoimmune disease. In addition to T lymphocytes, the AhR also appears to play a vital role in B cell maturation, and regulates the activity of macrophages, dendritic cells and neutrophils following lipopolysaccharide challenge or influenza virus infection. In these scenarios, activation of AhR is associated with decreased host response and reduced survival. Furthermore, gene knock out studies suggest that AhR is indispensable for the postnatal maintenance of intestinal intraepithelial lymphocytes and skin-resident dendritic epidermal gamma delta T cells, providing a potential link between AhR and gut immunity and wound healing. It is well accepted that the magnitude and the type of immune response is dependent on the local cytokine milieu and the AhR appears to be one of the key factors involved in the fine turning of this cytokine balance.
Collapse
Affiliation(s)
- Nan Hao
- School of Molecular and Biomedical Science (Biochemistry), The University of Adelaide, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
239
|
Gallo PM, Gallucci S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front Immunol 2013; 4:138. [PMID: 23772226 PMCID: PMC3677085 DOI: 10.3389/fimmu.2013.00138] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/23/2013] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) initiate and control immune responses, participate in the maintenance of immunological tolerance and are pivotal players in the pathogenesis of autoimmunity. In patients with autoimmune disease and in experimental animal models of autoimmunity, DCs show abnormalities in both numbers and activation state, expressing immunogenic levels of costimulatory molecules and pro-inflammatory cytokines. Exogenous and endogenous danger signals activate DCs to stimulate the immune response. Classic endogenous danger signals are released, activated, or secreted by host cells and tissues experiencing stress, damage, and non-physiologic cell death; and are therefore referred to as damage-associated molecular patterns (DAMPs). Some DAMPs are released from cells, where they are normally sequestered, during necrosis (e.g., heat shock proteins, uric acid, ATP, HMGB1, mitochondria-derived molecules). Others are actively secreted, like Type I Interferons. Here we discuss important DAMPs in the context of autoimmunity. For some, there is a clear pathogenic link (e.g., nucleic acids and lupus). For others, there is less evidence. Additionally, we explore emerging danger signals. These include inorganic materials and man-made technologies (e.g., nanomaterials) developed as novel therapeutic approaches. Some nanomaterials can activate DCs and may trigger unintended inflammatory responses. Finally, we will review “homeostatic danger signals,” danger signals that do not derive directly from pathogens or dying cells but are associated with perturbations of tissue/cell homeostasis and may signal pathological stress. These signals, like acidosis, hypoxia, and changes in osmolarity, also play a role in inflammation and autoimmunity.
Collapse
Affiliation(s)
- Paul M Gallo
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple Autoimmunity Center, Temple University School of Medicine , Philadelphia, PA , USA
| | | |
Collapse
|
240
|
The aryl hydrocarbon receptor: a novel target for immunomodulation in organ transplantation. Transplantation 2013; 95:983-90. [PMID: 23263608 DOI: 10.1097/tp.0b013e31827a3d1d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The aryl hydrocarbon receptor (AHR), which has been central to studies in toxicology for years as the receptor for the toxicant dioxin, is rapidly gaining interest in immunology based on its ability to influence T-cell differentiation. Multiple studies have documented that binding of this receptor with certain ligands favors T-cell differentiation toward regulatory T cells, and paradoxically, binding of this same receptor with different ligands enhances Th17 effector cell differentiation. This finding has been confirmed in both in vitro and in vivo models, where different ligands are able to either ameliorate or conversely aggravate autoimmunity in experimental autoimmune encephalomyelitis. The AHR has both an endogenous role that is important in development and normal physiology and an exogenous role as a receptor for manmade toxicants, with their binding leading to transcription of cytochrome P450 enzymes that metabolize these same ligands. Based on recent reports that will be summarized in this overview, we will consider the role that the AHR might play as a sensor to the outside environment, leading to alteration of the acquired immune system that might have relevance in transplantation or other medical conditions. In addition to describing the data in normal physiology and T-cell differentiation, we will present examples of the importance of this receptor in preclinical models of disease and highlight specific ligands that target the AHR and will have efficacy in treating transplant rejection and in tolerance protocols.
Collapse
|
241
|
Quintana FJ. Nanoparticles for the induction of antigen-specific Tregs. Immunotherapy 2013; 5:437-40. [DOI: 10.2217/imt.13.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham & Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston 02115, MA, USA
| |
Collapse
|
242
|
Quintana FJ. The aryl hydrocarbon receptor: a molecular pathway for the environmental control of the immune response. Immunology 2013. [PMID: 23190340 DOI: 10.1111/imm.12046] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Environmental factors have significant effects on the development of autoimmune diseases. The ligand-activated transcription factor aryl hydrocarbon receptor (AHR) is controlled by endogenous and environmental small molecules. Hence, AHR provides a molecular pathway by which endogenous and environmental signals can influence the immune response and the development of autoimmune diseases. AHR also provides a target for therapeutic intervention in immune-mediated disorders. In this review, we discuss the role of AHR in the regulation of T-cell differentiation and autoimmunity.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
243
|
Abstract
BACKGROUND The immune response involves the activation of heterogeneous populations of T cells and B cells that show different degrees of affinity and specificity for target antigens. Although several techniques have been developed to study the molecular pathways that control immunity, there is a need for high-throughput assays to monitor the specificity of the immune response. CONTENT Antigen microarrays provide a new tool to study the immune response. We reviewed the literature on antigen microarrays and their advantages and limitations, and we evaluated their use for the study of autoimmune diseases. Antigen arrays have been successfully used for several purposes in the investigation of autoimmune disorders: for disease diagnosis, to monitor disease progression and response to therapy, to discover mechanisms of pathogenesis, and to tailor antigen-specific therapies to the autoimmune response of individual patients. In this review we discuss the use of antigen microarrays for the study of 4 common autoimmune diseases and their animal models: type 1 diabetes, systemic lupus erythematosus, rheumatoid arthritis, and multiple sclerosis. CONCLUSIONS Antigen microarrays constitute a new tool for the investigation of the immune response in autoimmune disorders and also in other conditions such as tumors and allergies. Once current limitations are overcome, antigen microarrays have the potential to revolutionize the investigation and management of autoimmune diseases.
Collapse
Affiliation(s)
- Ada Yeste
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
244
|
Benson JM, Beamer CA, Seaver BP, Shepherd DM. INDOLE-3-CARBINOL EXERTS SEX-SPECIFIC EFFECTS IN MURINE COLITIS. EUR J INFLAMM 2012; 10:335-346. [PMID: 33024444 DOI: 10.1177/1721727x1201000309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to the severe adverse effects that can accompany conventional therapies for Crohn's disease, the search for natural complementary therapies has increased dramatically in recent years. Indole-3-carbinol (I3C), a constituent of cruciferous vegetables, possesses anti-inflammatory properties; however, its effects on intestinal inflammation have yet to be evaluated. To test the hypothesis that I3C dampens intestinal inflammation, C57Bl/6 mice were treated with I3C and exposed to 2,4,6-trinitrobenzenesulfonic acid (TNBS) to induce colitis. Several parameters of disease severity and inflammation were subsequently evaluated. I3C dampened the disease severity, as indicated by decreased body weight loss and decreased severity of clinical signs. Interestingly, this effect was observed in female but not male mice, which displayed a trend towards exacerbated colitis. Differential effects were observed in the profiles of cytokine production, as the production of pro-inflammatory cytokines was increased in males. The sex-specific effect of I3C in TNBS-induced colitis is a novel finding and warrants further investigation since this is a common dietary compound and is also available commercially.
Collapse
Affiliation(s)
- J M Benson
- Department of Biomedical and Pharmaceutical Sciences and Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| | - C A Beamer
- Department of Biomedical and Pharmaceutical Sciences and Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| | - B P Seaver
- Department of Biomedical and Pharmaceutical Sciences and Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| | - D M Shepherd
- Department of Biomedical and Pharmaceutical Sciences and Center for Environmental Health Sciences, University of Montana, Missoula, MT, USA
| |
Collapse
|