1
|
Stan G, Lorimer GH, Thirumalai D. Friends in need: How chaperonins recognize and remodel proteins that require folding assistance. Front Mol Biosci 2022; 9:1071168. [PMID: 36479385 PMCID: PMC9720267 DOI: 10.3389/fmolb.2022.1071168] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/07/2022] [Indexed: 08/19/2023] Open
Abstract
Chaperonins are biological nanomachines that help newly translated proteins to fold by rescuing them from kinetically trapped misfolded states. Protein folding assistance by the chaperonin machinery is obligatory in vivo for a subset of proteins in the bacterial proteome. Chaperonins are large oligomeric complexes, with unusual seven fold symmetry (group I) or eight/nine fold symmetry (group II), that form double-ring constructs, enclosing a central cavity that serves as the folding chamber. Dramatic large-scale conformational changes, that take place during ATP-driven cycles, allow chaperonins to bind misfolded proteins, encapsulate them into the expanded cavity and release them back into the cellular environment, regardless of whether they are folded or not. The theory associated with the iterative annealing mechanism, which incorporated the conformational free energy landscape description of protein folding, quantitatively explains most, if not all, the available data. Misfolded conformations are associated with low energy minima in a rugged energy landscape. Random disruptions of these low energy conformations result in higher free energy, less folded, conformations that can stochastically partition into the native state. Two distinct mechanisms of annealing action have been described. Group I chaperonins (GroEL homologues in eubacteria and endosymbiotic organelles), recognize a large number of misfolded proteins non-specifically and operate through highly coordinated cooperative motions. By contrast, the less well understood group II chaperonins (CCT in Eukarya and thermosome/TF55 in Archaea), assist a selected set of substrate proteins. Sequential conformational changes within a CCT ring are observed, perhaps promoting domain-by-domain substrate folding. Chaperonins are implicated in bacterial infection, autoimmune disease, as well as protein aggregation and degradation diseases. Understanding the chaperonin mechanism and the specific proteins they rescue during the cell cycle is important not only for the fundamental aspect of protein folding in the cellular environment, but also for effective therapeutic strategies.
Collapse
Affiliation(s)
- George Stan
- Department of Chemistry, University of Cincinnati, Cincinnati, OH, United States
| | - George H. Lorimer
- Center for Biomolecular Structure and Organization, Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, United States
| | - D. Thirumalai
- Department of Chemistry, University of Texas, Austin, TX, United States
- Department of Physics, University of Texas, Austin, TX, United States
| |
Collapse
|
2
|
Lessons Learned from Two Decades of Modeling the Heat-Shock Response. Biomolecules 2022; 12:biom12111645. [DOI: 10.3390/biom12111645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
The Heat Shock Response (HSR) is a highly conserved genetic system charged with protecting the proteome in a wide range of organisms and species. Experiments since the early 1980s have elucidated key elements in these pathways and revealed a canonical mode of regulation, which relies on a titration feedback. This system has been subject to substantial modeling work, addressing questions about resilience, design and control. The compact core regulatory circuit, as well as its apparent conservation, make this system an ideal ‘hydrogen atom’ model for the regulation of stress response. Here we take a broad view of the models of the HSR, focusing on the different questions asked and the approaches taken. After 20 years of modeling work, we ask what lessons had been learned that would have been hard to discover without mathematical models. We find that while existing models lay strong foundations, many important questions that can benefit from quantitative modeling are still awaiting investigation.
Collapse
|
3
|
Guerra PV, Andrade CM, Nunes IV, Gama BC, Tibúrcio R, Santos WLC, Azevedo VA, Tavares NM, Rebouças JDS, Maiolii TU, Faria AMC, Brodskyn CI. Oral Tolerance Induced by Heat Shock Protein 65-Producing Lactococcus lactis Mitigates Inflammation in Leishmania braziliensis Infection. Front Immunol 2021; 12:647987. [PMID: 34248935 PMCID: PMC8264454 DOI: 10.3389/fimmu.2021.647987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 12/05/2022] Open
Abstract
Cutaneous leishmaniasis caused by L. braziliensis induces a pronounced Th1 inflammatory response characterized by IFN-γ production. Even in the absence of parasites, lesions result from a severe inflammatory response in which inflammatory cytokines play an important role. Different approaches have been used to evaluate the therapeutic potential of orally administrated heat shock proteins (Hsp). These proteins are evolutionarily preserved from bacteria to humans, highly expressed under inflammatory conditions and described as immunodominant antigens. Tolerance induced by the oral administration of Hsp65 is capable of suppressing inflammation and inducing differentiation in regulatory cells, and has been successfully demonstrated in several experimental models of autoimmune and inflammatory diseases. We initially administered recombinant Lactococcus lactis (L. lactis) prior to infection as a proof of concept, in order to verify its immunomodulatory potential in the inflammatory response arising from L. braziliensis. Using this experimental approach, we demonstrated that the oral administration of a recombinant L. lactis strain, which produces and secretes Hsp65 from Mycobacterium leprae directly into the gut, mitigated the effects of inflammation caused by L. braziliensis infection in association or not with PAM 3CSK4 (N-α-Palmitoyl-S-[2,3-bis(palmitoyloxy)-(2RS)-propyl]-L-cysteine, a TLR2 agonist). This was evidenced by the production of anti-inflammatory cytokines and the expansion of regulatory T cells in the draining lymph nodes of BALB/c mice. Our in vitro experimental results suggest that IL-10, TLR-2 and LAP are important immunomodulators in L. braziliensis infection. In addition, recombinant L. lactis administered 4 weeks after infection was observed to decrease lesion size, as well as the number of parasites, and produced a higher IL-10 production and decrease IFN-γ secretion. Together, these results indicate that Hsp65-producing L. lactis can be considered as an alternative candidate for treatment in both autoimmune diseases, as well as in chronic infections that cause inflammatory disease.
Collapse
Affiliation(s)
- Priscila Valera Guerra
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Curso de Medicina, Centro Universitário Christus, Fortaleza, Brazil
| | - Camila Mattos Andrade
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Ivanéia Valeriano Nunes
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Brena Cardoso Gama
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Rafael Tibúrcio
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Washington Luis Conrado Santos
- Laboratório de Patologia Estrutural e Molecular (LAPEM), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Departamento de Patologia e Medicina Legal Faculdade de Medicina da Universidade Federal da Bahia, Salvador, Brazil
| | - Vasco Ariston Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biomédicas, Universidade Federal de Minais Gerais, Belo Horizonte, Brazil
| | - Natalia Machado Tavares
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Juliana de Souza Rebouças
- Instituto de Ciências Biológicas, Programa de Pós Graduação em Ciências da Saúde, Universidade de Pernambuco, Recife, Brazil
| | - Tatiani Uceli Maiolii
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cláudia Ida Brodskyn
- Laboratório da Interação Parasita-Hospedeiro e Epidemiologia (LAIPHE) Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
4
|
Moin ASM, Nandakumar M, Diane A, Dehbi M, Butler AE. The Role of Heat Shock Proteins in Type 1 Diabetes. Front Immunol 2021; 11:612584. [PMID: 33584694 PMCID: PMC7873876 DOI: 10.3389/fimmu.2020.612584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/30/2020] [Indexed: 01/08/2023] Open
Abstract
Type 1 diabetes (T1D) is a T-cell mediated autoimmune disease characterized by recognition of pancreatic β-cell proteins as self-antigens, called autoantigens (AAgs), followed by loss of pancreatic β-cells. (Pre-)proinsulin ([P]PI), glutamic acid decarboxylase (GAD), tyrosine phosphatase IA-2, and the zinc transporter ZnT8 are key molecules in T1D pathogenesis and are recognized by autoantibodies detected in routine clinical laboratory assays. However, generation of new autoantigens (neoantigens) from β-cells has also been reported, against which the autoreactive T cells show activity. Heat shock proteins (HSPs) were originally described as “cellular stress responders” for their role as chaperones that regulate the conformation and function of a large number of cellular proteins to protect the body from stress. HSPs participate in key cellular functions under both physiological and stressful conditions, including suppression of protein aggregation, assisting folding and stability of nascent and damaged proteins, translocation of proteins into cellular compartments and targeting irreversibly damaged proteins for degradation. Low HSP expression impacts many pathological conditions associated with diabetes and could play a role in diabetic complications. HSPs have beneficial effects in preventing insulin resistance and hyperglycemia in type 2 diabetes (T2D). HSPs are, however, additionally involved in antigen presentation, presenting immunogenic peptides to class I and class II major histocompatibility molecules; thus, an opportunity exists for HSPs to be employed as modulators of immunologic responses in T1D and other autoimmune disorders. In this review, we discuss the multifaceted roles of HSPs in the pathogenesis of T1D and in autoantigen-specific immune protection against T1D development.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Manjula Nandakumar
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Abdoulaye Diane
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Mohammed Dehbi
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Alexandra E Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
5
|
Loss of gut barrier integrity triggers activation of islet-reactive T cells and autoimmune diabetes. Proc Natl Acad Sci U S A 2019; 116:15140-15149. [PMID: 31182588 PMCID: PMC6660755 DOI: 10.1073/pnas.1814558116] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Functional loss of gut barrier integrity with subsequent increased antigen trafficking and occurrence of low-grade intestinal inflammation precede the onset of type 1 diabetes (T1D) in patients and preclinical models, thus suggesting that these events are mechanistically linked to the autoimmune pathogenesis of the disease. However, a causal relationship between increased intestinal permeability and autoimmune diabetes was never demonstrated. Our data show that breakage of gut barrier continuity leads to activation of islet-reactive T cells in the intestine, thus triggering autoimmune diabetes. An important implication of our findings is that restoration of a healthy gut barrier through microbiota and diet modulation in diabetes-prone individuals could reduce intestinal activation of islet-reactive T cells and prevent T1D occurrence. Low-grade intestinal inflammation and alterations of gut barrier integrity are found in patients affected by extraintestinal autoimmune diseases such as type 1 diabetes (T1D), but a direct causal link between enteropathy and triggering of autoimmunity is yet to be established. Here, we found that onset of autoimmunity in preclinical models of T1D is associated with alterations of the mucus layer structure and loss of gut barrier integrity. Importantly, we showed that breakage of the gut barrier integrity in BDC2.5XNOD mice carrying a transgenic T cell receptor (TCR) specific for a beta cell autoantigen leads to activation of islet-reactive T cells within the gut mucosa and onset of T1D. The intestinal activation of islet-reactive T cells requires the presence of gut microbiota and is abolished when mice are depleted of endogenous commensal microbiota by antibiotic treatment. Our results indicate that loss of gut barrier continuity can lead to activation of islet-specific T cells within the intestinal mucosa and to autoimmune diabetes and provide a strong rationale to design innovative therapeutic interventions in “at-risk” individuals aimed at restoring gut barrier integrity to prevent T1D occurrence.
Collapse
|
6
|
Xin GLL, Khee YP, Ying TY, Chellian J, Gupta G, Kunnath AP, Nammi S, Collet T, Hansbro PM, Dua K, Chellappan DK. Current Status on Immunological Therapies for Type 1 Diabetes Mellitus. Curr Diab Rep 2019; 19:22. [PMID: 30905013 DOI: 10.1007/s11892-019-1144-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) occurs when there is destruction of beta cells within the islets of Langerhans in the pancreas due to autoimmunity. It is considered a complex disease, and different complications can surface and worsen the condition if T1D is not managed well. Since it is an incurable disease, numerous treatments and therapies have been postulated in order to control T1D by balancing hyperglycemia control while minimizing hypoglycemic episodes. The purpose of this review is to primarily look into the current state of the available immunological therapies and their advantages for the treatment of T1D. RECENT FINDINGS Over the years, immunological therapy has become the center of attraction to treat T1D. Immunomodulatory approaches on non-antigens involving agents such as cyclosporine A, mycophenolate mofetil, anti-CD20, cytotoxic T cells, anti-TNF, anti-CD3, and anti-thymocyte globulin as well as immunomodulative approaches on antigens such as insulin, glutamic acid decarboxylase, and heat shock protein 60 have been studied. Aside from these two approaches, studies and trials have also been conducted on regulatory T cells, dendritic cells, interleukin 2, interleukin 4, M2 macrophages, and rapamycin/interleukin 2 combination therapy to test their effects on patients with T1D. Many of these agents have successfully suppressed T1D in non-obese diabetic (NOD) mice and in human trials. However, some have shown negative results. To date, the insights into the management of the immune system have been increasing rapidly to search for potential therapies and treatments for T1D. Nevertheless, some of the challenges are still inevitable. A lot of work and effort need to be put into the investigation on T1D through immunological therapy, particularly to reduce complications to improve and enhance clinical outcomes.
Collapse
Affiliation(s)
- Griselda Lim Loo Xin
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Yap Pui Khee
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Tan Yoke Ying
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India
| | - Anil Philip Kunnath
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Sydney, NSW, 2751, Australia
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, 2751, Australia
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, 4059, Australia
| | - Philip Michael Hansbro
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Pockley AG, Henderson B. Extracellular cell stress (heat shock) proteins-immune responses and disease: an overview. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0522. [PMID: 29203707 DOI: 10.1098/rstb.2016.0522] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2017] [Indexed: 12/11/2022] Open
Abstract
Extracellular cell stress proteins are highly conserved phylogenetically and have been shown to act as powerful signalling agonists and receptors for selected ligands in several different settings. They also act as immunostimulatory 'danger signals' for the innate and adaptive immune systems. Other studies have shown that cell stress proteins and the induction of immune reactivity to self-cell stress proteins can attenuate disease processes. Some proteins (e.g. Hsp60, Hsp70, gp96) exhibit both inflammatory and anti-inflammatory properties, depending on the context in which they encounter responding immune cells. The burgeoning literature reporting the presence of stress proteins in a range of biological fluids in healthy individuals/non-diseased settings, the association of extracellular stress protein levels with a plethora of clinical and pathological conditions and the selective expression of a membrane form of Hsp70 on cancer cells now supports the concept that extracellular cell stress proteins are involved in maintaining/regulating organismal homeostasis and in disease processes and phenotype. Cell stress proteins, therefore, form a biologically complex extracellular cell stress protein network having diverse biological, homeostatic and immunomodulatory properties, the understanding of which offers exciting opportunities for delivering novel approaches to predict, identify, diagnose, manage and treat disease.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- A Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Brian Henderson
- Division of Microbial Diseases, UCL Eastman Dental Institute, London WC1X 8LD, UK
| |
Collapse
|
8
|
Meng Q, Li BX, Xiao X. Toward Developing Chemical Modulators of Hsp60 as Potential Therapeutics. Front Mol Biosci 2018; 5:35. [PMID: 29732373 PMCID: PMC5920047 DOI: 10.3389/fmolb.2018.00035] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/26/2018] [Indexed: 12/22/2022] Open
Abstract
The 60 kDa heat shock protein (Hsp60) is classically known as a mitochondrial chaperonin protein working together with co-chaperonin 10 kDa heat shock protein (Hsp10). This chaperonin complex is essential for folding proteins newly imported into mitochondria. However, Hsp60, and/or Hsp10 have also been shown to reside in other subcellular compartments including extracellular space, cytosol, and nucleus. The proteins in these extra-mitochondrial compartments may possess a wide range of functions dependent or independent of its chaperoning activity. But the mechanistic details remain unknown. Mutations in Hsp60 gene have been shown to be associated with neurodegenerative disorders. Abnormality in expression level and/or subcellular localization have also been detected from different diseased tissues including inflammatory diseases and various cancers. Therefore, there is a strong interest in developing small molecule modulators of Hsp60. Most of the reported inhibitors were discovered through various chemoproteomics strategies. In this review, we will describe the recent progress in this area with reported inhibitors from both natural products and synthetic compounds. The former includes mizoribine, epolactaene, myrtucommulone, stephacidin B, and avrainvillamide while the latter includes o-carboranylphenoxyacetanilides and gold (III) porphyrins. The potencies of the known inhibitors range from low micromolar to millimolar concentrations. The potential applications of these inhibitors include anti-cancer, anti-inflammatory diseases, and anti-autoimmune diseases.
Collapse
Affiliation(s)
- Qianli Meng
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, United States
| | - Bingbing X Li
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, United States
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
9
|
Chlamydia trachomatis: the Persistent Pathogen. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00203-17. [PMID: 28835360 DOI: 10.1128/cvi.00203-17] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium whose only natural host is humans. Although presenting as asymptomatic in most women, genital tract chlamydial infections are a leading cause of pelvic inflammatory disease, tubal factor infertility, and ectopic pregnancy. C. trachomatis has evolved successful mechanisms to avoid destruction by autophagy and the host immune system and persist within host epithelial cells. The intracellular form of this organism, the reticulate body, can enter into a persistent nonreplicative but viable state under unfavorable conditions. The infectious form of the organism, the elementary body, is again generated when the immune attack subsides. In its persistent form, C. trachomatis ceases to produce its major structural and membrane components, but synthesis of its 60-kDa heat shock protein (hsp60) is greatly upregulated and released from the cell. The immune response to hsp60, perhaps exacerbated by repeated cycles of productive infection and persistence, may promote damage to fallopian tube epithelial cells, scar formation, and tubal occlusion. The chlamydial and human hsp60 proteins are very similar, and hsp60 is one of the first proteins produced by newly formed embryos. Thus, the development of immunity to epitopes in the chlamydial hsp60 that are also present in the corresponding human hsp60 may increase susceptibility to pregnancy failure in infected women. Delineation of host factors that increase the likelihood that C. trachomatis will avoid immune destruction and survive within host epithelial cells and utilization of this knowledge to design individualized preventative and treatment protocols are needed to more effectively combat infections by this persistent pathogen.
Collapse
|
10
|
Shen L, Lu S, Huang D, Li G, Liu K, Cao R, Zong L, Jin L, Wu J. A rationally designed peptide IA-2-P2 against type 1 diabetes in streptozotocin-induced diabetic mice. Diab Vasc Dis Res 2017; 14:184-190. [PMID: 28467202 DOI: 10.1177/1479164116664189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Recent studies have investigated the potential of type 1 diabetes mellitus-related autoantigens, such as heat shock protein 60, to induce immunological tolerance or to suppress the immune response. A functional 24-residue peptide derived from heat shock protein 60 (P277) has shown anti-type 1 diabetes mellitus potential in experimental animals and in clinical studies, but it also carries a potential atherogenic effect. In this study, we have modified P277 to retain an anti-type 1 diabetes mellitus effect and minimize the atherogenic potential by replacing the P277 B epitope with another diabetes-associated autoantigen, insulinoma antigen-2 (IA-2), to create the fusion peptide IA-2-P2. In streptozotocin-induced diabetic C57BL/6J mice, the IA-2-P2 peptide displayed similar anti-diabetic effects to the control P277 peptide. Also, the IA-2-P2 peptide did not show atherogenic activity in a rabbit model. Our findings indicate the potential of IA-2-P2 as a promising vaccine against type 1 diabetes mellitus.
Collapse
MESH Headings
- Animals
- Atherosclerosis/chemically induced
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Cell Proliferation/drug effects
- Cells, Cultured
- Chaperonin 60/administration & dosage
- Chaperonin 60/pharmacology
- Chaperonin 60/toxicity
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Drug Design
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/toxicity
- Immunization
- Lymphocyte Activation/drug effects
- Male
- Mice, Inbred C57BL
- Peptide Fragments/administration & dosage
- Peptide Fragments/pharmacology
- Peptide Fragments/toxicity
- Rabbits
- Receptor-Like Protein Tyrosine Phosphatases, Class 8/administration & dosage
- Receptor-Like Protein Tyrosine Phosphatases, Class 8/pharmacology
- Receptor-Like Protein Tyrosine Phosphatases, Class 8/toxicity
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/pharmacology
- Recombinant Fusion Proteins/toxicity
- Streptozocin
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
- Vaccines/administration & dosage
- Vaccines/pharmacology
- Vaccines/toxicity
Collapse
Affiliation(s)
- Lili Shen
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shiping Lu
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dongcheng Huang
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Guoliang Li
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Kunfeng Liu
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Rongyue Cao
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Li Zong
- 2 Institute of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liang Jin
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jie Wu
- 1 Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Abstract
In spite of modern techniques, the burden for patients with type 1 diabetes mellitus will not disappear, and type 1 diabetes will remain a life-threatening disease causing severe complications and increased mortality. We have to learn of ways to stop the destructive process, preserve residual insulin secretion or even improve the disease via β-cell regeneration. This will give a milder disease, a more stable metabolism, simpler treatment and perhaps even cure. Therapies based on single drugs have not shown sufficient efficacy; however, there are several treatments with encouraging efficacy and no apparent, or rather mild, adverse events. As the disease process is heterogeneous, treatments have to be chosen to fit relevant subgroups of patients, and step by step efficacy can possibly be improved by the use of combination therapies. Thus immunosuppressive therapies like anti-CD3 and anti-CD20 monoclonal antibodies might be combined with fusion proteins such as etanercept [tumor necrosis factor (TNF)-α inhibitor] and/or abatacept (CTLA4-Ig) early after onset to stop the destructive process, supported by β-cell protective agents. The effect may be prolonged by using autoantigen therapy [glutamate decarboxylase (GAD) proinsulin], and by adding agents facilitating β-cell regeneration [e.g. glucagon-like peptide-1 (GLP-1)] there should be a good chance to make the disease milder, perhaps leading to cure in some patients.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, 58185, Linköping, Sweden.
| |
Collapse
|
12
|
Abstract
Diabetes is a chronic disease, and its prevalence continues to rise and can increase the risk for the progression of microvascular (such as nephropathy, retinopathy and neuropathy) and also macrovascular complications. Diabetes is a condition in which the oxidative stress and inflammation rise. Heat shock proteins (HSPs) are a highly conserved family of proteins that are expressed by all cells exposed to environmental stress, and they have diverse functions. In patients with diabetes, the expression and levels of HSPs decrease, but these chaperones can aid in improving some complications of diabetes, such as oxidative stress and inflammation. (The suppression of some HSPs is associated with a generalized increase in tissue inflammation.) In this review, we summarize the current understanding of HSPs in diabetes as well as their complications, and we also highlight their potential role as therapeutic targets in diabetes.
Collapse
|
13
|
Fusion protein His-Hsp65-6IA2P2 prevents type 1 diabetes through nasal immunization in NOD Mice. Int Immunopharmacol 2016; 35:235-242. [PMID: 27082999 DOI: 10.1016/j.intimp.2016.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 03/15/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022]
Abstract
Human heat shock protein 60 (Hsp60), is an endogenous β-cells autoantigen, it could postpone the onset of insulitis and sooner type 1 diabetes mellitus. P277 is one of Hsp65 determinants at position 437-469 of amino acids cascaded. Meanwhile, it's already well-known that there were several better anti-diabetic B epitopes, such as insulinoma antigen-2 (IA-2). Currently, fusion protein IA2P2 has constructed in order to enhance its pharmacological efficacy. In addition, added homologous bacterial-derived Hsp65 and His tag were beneficial to protein immunogenicity and purification separately. So, finally we examined a fusion protein His-Hsp65-6IA2P2 could regulate Th2 immune response and reduce natural diabetic incidence in NOD mice. We constructed two express vector pET28a-His-Hsp65-6P277 and pET28a-His-Hsp65-6IA2P2. After purification, we observed that triple intranasal administration of these two fusion protein in 4-week-old NOD mice maintained normal blood glucose and weight, with a lower diabetic or insulitis incidence. Consistent with induced splenic T cells proliferation and tolerance, His-Hsp65-6IA2P2-treated mice performed reduced IFN-γ and increased IL-10 level. In conclusion, we suggested that fusion protein His-Hsp65-6IA2P2 could be reconstructed and purified successively. Furthermore, nasal administration of this fusion protein could rebalance T cells population and prevent T1DM.
Collapse
|
14
|
Wang X, Wang J, Liang Y, Ni H, Shi L, Xu C, Zhou Y, Su Y, Mou X, Chen D, Mao C. Schistosoma japonicum HSP60-derived peptide SJMHE1 suppresses delayed-type hypersensitivity in a murine model. Parasit Vectors 2016; 9:147. [PMID: 26971312 PMCID: PMC4789290 DOI: 10.1186/s13071-016-1434-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/05/2016] [Indexed: 12/20/2022] Open
Abstract
Background Parasite-derived molecules with immunomodulatory properties, which have been optimised during host-parasite co-evolution, exhibit potential applications as novel immunotherapeutics. We have previously demonstrated that Schistosoma japonicum HSP60-derived peptide SJMHE1 induces CD4+CD25+ regulatory T-cells (Tregs) and that adoptively transferred SJMHE1-induced CD4+CD25+ Tregs inhibit delayed-type hypersensitivity (DTH) in mice. However, multiple concerns regarding this method render this treatment unsuitable. To gain further insights into the potential effects of SJMHE1, we used ovalbumin (OVA)-induced DTH and evaluated the effect of SJMHE1 on DTH mice. Methods BALB/c mice were sensitised with OVA alone or combined with SJMHE1 and then challenged with OVA to induce DTH. We first analysed the potential effects of SJMHE1 by measuring DTH responses, T-cell responses, cytokine secretion, and Treg proportions. We then evaluated the expression levels of IL-10 and TGF-β1 in CD4+CD25+ T-cells during DTH and Treg generation to identify the mechanism by which SJMHE1 suppresses DTH. Results SJMHE1 modulated the effector response against OVA-induced DTH and stimulated the production of the anti-inflammatory cytokines IL-10 and TGF-β1 in immunised mice through a mechanism involving CD4+CD25+ Tregs. SJMHE1-induced CD4+CD25+ Tregs expressed high levels of CTLA-4, IL-10, and TGF-β1, which substantially contributed to the suppressive activity during DTH. The administration of SJMHE1 to DTH in mice led to the expansion of CD4+CD25+ Tregs from CD4+CD25− T-cells in the periphery, which inhibited DTH responses. Conclusions Our study proves that the parasite-driven peptide suppresses DTH in mice, which may confer a new option for inflammation treatment.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China. .,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Jun Wang
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Yong Liang
- Clinical Laboratory, Huai'an Hospital Affiliated of Xuzhou Medical College, Huaian, Jiangsu, 223300, China
| | - Hongchang Ni
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Liang Shi
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chengcheng Xu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuting Su
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiao Mou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Deyu Chen
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chaoming Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
15
|
Promotion of atherosclerosis in high cholesterol diet-fed rabbits by immunization with the P277 peptide. Immunol Lett 2015; 170:80-7. [PMID: 26730848 DOI: 10.1016/j.imlet.2015.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/12/2015] [Accepted: 12/20/2015] [Indexed: 01/25/2023]
Abstract
Previous evidence has proved the ability of immunization with heat shock protein (HSP) 60/65 to induce atherosclerosis. P277, a 24-residue peptide of human HSP60, is a promising peptide vaccine against autoimmune diabetes. But as a fragment of HSP60, its potential ability of promoting atherosclerosis has never been investigated yet. In the present study, the rabbits fed with normal standard diet or high cholesterol diet were immunized with P277 or PBS emulsified in incomplete Freund's adjuvant 4 times at 4-week intervals. Atherosclerotic lesions of the rabbits receiving P277 treatment and fed with high cholesterol diet increased significantly compared with those of the rabbits receiving PBS treatment and the same diet. However, no obvious lesions were found in the two groups of rabbits fed with the normal standard diet. Significant expression of P277 was detected in the high cholesterol diet-induced atherosclerotic lesions and heat-stressed endothelial cells. Surface exposure of P277 was also observed in the stressed cells. In the subsequent assay of endothelial cells in vitro, the purified anti-P277 antibodies mediated a noticeable cytotoxicity to the stressed cells with the participation of complement. In conclusion, subcutaneous immunization with P277 emulsified in IFA can aggravate the atherosclerosis in high cholesterol diet-fed rabbits. Surface expression of P277 was observed on stressed endothelial cells, and were suggested to mediate the autoimmune attack and promote the disease.
Collapse
|
16
|
Ilyas Z, Shah HS, Al-Oweini R, Kortz U, Iqbal J. Antidiabetic potential of polyoxotungstates: in vitro and in vivo studies. Metallomics 2015; 6:1521-6. [PMID: 24887259 DOI: 10.1039/c4mt00106k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetes mellitus is a chronic metabolic disorder continuously affecting people all over the world. A common way to treat diabetes mellitus is to limit the conversion of carbohydrates into glucose which is mediated by glucosidase enzymes. Diabetes mellitus is also famous for its life-threatening microvascular (retinopathy, neuropathy and nephropathy) and macrovascular (atherosclerosis) complications. Aldose reductases present in eye lens (ALR1) and kidney (ALR2) are responsible for microvascular complications. The production of advanced glycation end products (AGEs) is involved in the development of atherosclerosis. The present work was aimed at the synthesis and in vitro/in vivo evaluation of different polyoxotungstates against glucosidases (α- and β), aldose reductases (ALR1 and ALR2) and AGEs to discover a new treatment which may limit the complications associated with diabetes mellitus. The polyanion [P6W18O79](20-) was found to be the most potent inhibitor of α-glucosidase (IC50 = 1.33 ± 0.41 μM), ALR1 (IC50 = 0.4 ± 0.009 μM) and ALR2 (IC50 = 0.38 ± 0.02 μM). Animal studies showed that the polyanion [H2W12O40](6-) was very effective in reducing the blood glucose level to 84.25 ± 5.07 mg dL(-1) when compared with standard antidiabetic drug glibenclamide (150.62 ± 9.35 mg dL(-1)) measured after maximum 8 h of dose administration. The data obtained from in vitro and in vivo experiments confirm that [P6W18O79](20-) and [H2W12O40](6-) could be used as a new treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Zaitoon Ilyas
- Centre for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad, Postal Code 22060, Pakistan.
| | | | | | | | | |
Collapse
|
17
|
Sarikonda G, Sachithanantham S, Miller JF, Pagni PP, Coppieters KT, von Herrath M. The Hsp60 peptide p277 enhances anti-CD3 mediated diabetes remission in non-obese diabetic mice. J Autoimmun 2015; 59:61-6. [PMID: 25772283 DOI: 10.1016/j.jaut.2015.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells leading to inadequate glycemic control. Trials with immunomodulatory monotherapies have shown that the disease course can in principle be altered. The observed preservation of endogenous insulin secretion however is typically transient and chronic treatment is often associated with significant side effects. Here we combined anti-CD3 with the Hsp60 peptide p277, two drugs that have been evaluated in Phase 3 trials, to test for enhanced efficacy. Female NOD mice with recent onset diabetes were given 5 μg anti-CD3 i.v., on three consecutive days in combination with 100 μg of p277 peptide in IFA s.c., once weekly for four weeks. Anti-CD3 alone restored normoglycemia in 44% of the mice while combination therapy with anti-CD3 and p277 induced stable remission in 83% of mice. The observed increase in protection occurred only in part through TLR2 signaling and was characterized by increased Treg numbers and decreased insulitis. These results have important implications for the design of combination therapies for the treatment of T1D.
Collapse
Affiliation(s)
- Ghanashyam Sarikonda
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | - Jacqueline F Miller
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Philippe P Pagni
- Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA
| | - Ken T Coppieters
- Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA
| | - Matthias von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Type 1 Diabetes R&D Center, Novo Nordisk, Inc., Seattle, WA, USA.
| |
Collapse
|
18
|
Doran TM, Simanski S, Kodadek T. Discovery of native autoantigens via antigen surrogate technology: application to type 1 diabetes. ACS Chem Biol 2015; 10:401-12. [PMID: 25474415 PMCID: PMC4339956 DOI: 10.1021/cb5007618] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/04/2014] [Indexed: 01/23/2023]
Abstract
A fundamental goal in understanding the mechanisms of autoimmune disease is the characterization of autoantigens that are targeted by autoreactive antibodies and T cells. Unfortunately, the identification of autoantigens is a difficult problem. We have begun to explore a novel route to the discovery of autoantibody/autoantigen pairs that involves comparative screening of combinatorial libraries of unnatural, synthetic molecules for compounds that bind antibodies present at much higher levels in the serum of individuals with a given autoimmune disease than in the serum of control individuals. We have shown that this approach can yield "antigen surrogates" capable of capturing disease-specific autoantibodies from serum. In this report, we demonstrate that the synthetic antigen surrogates can be used to affinity purify the autoantibodies from serum and that these antibodies can then be used to identify their cognate autoantigen in an appropriate tissue lysate. Specifically, we report the discovery of a peptoid able to bind autoantibodies present in about one-third of nonobese diabetic (NOD) mice. The peptoid-binding autoantibodies were highly enriched through peptoid affinity chromatography and employed to probe mouse pancreatic and brain lysates. This resulted in identification of murine GAD65 as the native autoantigen. GAD65 is a known humoral autoantigen in human type 1 diabetes mellitus (T1DM), but its existence in mice had been controversial. This study demonstrates the potential of this chemical approach for the unbiased identification of autoantigen/autoantibody complexes.
Collapse
Affiliation(s)
- Todd M. Doran
- Departments
of Chemistry
& Cancer Biology, The Scripps Research
Institute, 130 Scripps
Way, Jupiter, Florida 33458, United States
| | - Scott Simanski
- Departments
of Chemistry
& Cancer Biology, The Scripps Research
Institute, 130 Scripps
Way, Jupiter, Florida 33458, United States
| | - Thomas Kodadek
- Departments
of Chemistry
& Cancer Biology, The Scripps Research
Institute, 130 Scripps
Way, Jupiter, Florida 33458, United States
| |
Collapse
|
19
|
Exosomal Hsp70 mediates immunosuppressive activity of the myeloid-derived suppressor cells via phosphorylation of Stat3. Med Oncol 2015; 32:453. [PMID: 25603952 DOI: 10.1007/s12032-014-0453-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 12/11/2014] [Indexed: 01/04/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs), one of the main cell populations, are responsible for regulating the immune response, which accumulates in tumor-bearing mice and humans contributing to cancer development. Exosomes produced by tumor cells have been involved in tumor-associated immune suppression. However, the role of exosomes is unclear in the activation of MDSCs. Here, we have purified tumor-derived exosomes from the supernatants of Renca cell cultures. Transmission electron microscopy was used to confirm their morphology, and Western blot analysis showed that Hsp70 was rich in these isolated exosomes compared with the whole-cell lysates of Renca cells. Then, we demonstrated that there was a more powerful activity of exosomal Hsp70-mediated induction of proinflammation cytokines, tumor growth factors of MDSCs and tumor progression than exosomes pre-incubated with anti-Hsp70 antibody. Furthermore, we show that an interactive exosomal HSP70 and MDSCs determine the suppressive activity of the MDSCs via phosphorylation of Stat3 (p-Stat3). Finally, we show that exosomal Hsp70 triggers p-Stat3 in MDSCs in a TLR2-MyD88-dependent manner. Meanwhile, we also find that there is a more significant increase in the percentage of CD11b+Gr-1+ cells in the mice, which are treated with exosomal Hsp70 than that exosomes pre-incubated with anti-Hsp70 antibody. Hence, we believe that the signaling pathway activation by exosomal Hsp70 within MDSCs may be a significant target in future treatment of renal cell carcinoma.
Collapse
|
20
|
Grönholm J, Lenardo MJ. Novel diagnostic and therapeutic approaches for autoimmune diabetes--a prime time to treat insulitis as a disease. Clin Immunol 2014; 156:109-18. [PMID: 25486604 DOI: 10.1016/j.clim.2014.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/22/2014] [Indexed: 02/09/2023]
Abstract
Type 1 diabetes is a progressive autoimmune disease with no curative treatment, making prevention critical. At the time of diagnosis, a majority of the insulin secreting β-cells have already been destroyed. Insulitis, lymphocytic infiltration to the pancreatic islets, is believed to begin months to years before the clinical symptoms of insulin deficiency appear. Insulitis should be treated as its own disease, for it is a known precursor to autoimmune diabetes. Because it is difficult to detect insulitic cellular infiltrates noninvasively, considerable interest has been focused on the levels of islet autoantibodies in blood as measurable diagnostic markers for islet autoimmunity. The traditional islet autoantibody detection assays have many limitations. New electrochemiluminescence-based autoantibody detection assays have the potential to overcome these challenges and they offer promising, cost-effective screening tools in identifying high-risk individuals for trials of preventive interventions. Here, we outline diagnostic and therapeutic strategies to overcome pancreatic β-cell destroying insulitis.
Collapse
Affiliation(s)
- Juha Grönholm
- Molecular Development of the Immune System Section, Laboratory of Immunology, NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immunology, NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Cohen IR. Activation of benign autoimmunity as both tumor and autoimmune disease immunotherapy: A comprehensive review. J Autoimmun 2014; 54:112-7. [DOI: 10.1016/j.jaut.2014.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 05/19/2014] [Indexed: 12/25/2022]
|
22
|
Husseiny MI, Kaye A, Zebadua E, Kandeel F, Ferreri K. Tissue-specific methylation of human insulin gene and PCR assay for monitoring beta cell death. PLoS One 2014; 9:e94591. [PMID: 24722187 PMCID: PMC3983232 DOI: 10.1371/journal.pone.0094591] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/17/2014] [Indexed: 01/12/2023] Open
Abstract
The onset of metabolic dysregulation in type 1 diabetes (T1D) occurs after autoimmune destruction of the majority of pancreatic insulin-producing beta cells. We previously demonstrated that the DNA encoding the insulin gene is uniquely unmethylated in these cells and then developed a methylation-specific PCR (MSP) assay to identify circulating beta cell DNA in streptozotocin-treated mice prior to the rise in blood glucose. The current study extends to autoimmune non-obese diabetic (NOD) mice and humans, showing in NOD mice that beta cell death occurs six weeks before the rise in blood sugar and coincides with the onset of islet infiltration by immune cells, demonstrating the utility of MSP for monitoring T1D. We previously reported unique patterns of methylation of the human insulin gene, and now extend this to other human tissues. The methylation patterns of the human insulin promoter, intron 1, exon 2, and intron 2 were determined in several normal human tissues. Similar to our previous report, the human insulin promoter was unmethylated in beta cells, but methylated in all other tissues tested. In contrast, intron 1, exon 2 and intron 2 did not exhibit any tissue-specific DNA methylation pattern. Subsequently, a human MSP assay was developed based on the methylation pattern of the insulin promoter and human islet DNA was successfully detected in circulation of T1D patients after islet transplantation therapy. Signal levels of normal controls and pre-transplant samples were shown to be similar, but increased dramatically after islet transplantation. In plasma the signal declines with time but in whole blood remains elevated for at least two weeks, indicating that association of beta cell DNA with blood cells prolongs the signal. This assay provides an effective method to monitor beta cell destruction in early T1D and in islet transplantation therapy.
Collapse
Affiliation(s)
- Mohamed I. Husseiny
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Alexander Kaye
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Emily Zebadua
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Fouad Kandeel
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Kevin Ferreri
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Coppieters KT, Harrison LC, von Herrath MG. Trials in type 1 diabetes: Antigen-specific therapies. Clin Immunol 2013; 149:345-55. [PMID: 23490422 PMCID: PMC5777514 DOI: 10.1016/j.clim.2013.02.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 02/05/2013] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) results from an aberrant immunological response against the insulin-producing beta cells in the islets of the pancreas. The ideal therapy would restore immune balance in a safe and lasting fashion, stopping the process of beta cell decay. The efficacy of immune suppressive agents such as cyclosporin underscores the notion that T1D can in principle be prevented, albeit at an unacceptable long-term safety risk. Immune modulatory drugs such as monoclonal anti-CD3 antibody, on the other hand, have recently had rather disappointing results in phase 3 trials, possibly due to inadequate dosing or choice of inappropriate endpoints. Therefore, it is argued that striking the right balance between safety and efficacy, together with careful trial design, will be paramount in preventing T1D. Here we outline the concept of antigen-specific tolerization as a strategy to safely induce long-term protection against T1D, focusing on available clinical trial data, key knowledge gaps and potential future directions.
Collapse
Affiliation(s)
| | - Leonard C. Harrison
- The Walter and Eliza Hall Institute of Medical Research and Department of Clinical Immunology and Burnet Clinical Research Unit, The Royal Melbourne Hospital, Melbourne, Australia
| | - Matthias G. von Herrath
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA
- Type 1 Diabetes Center, The La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
24
|
Rydén AKE, Wesley JD, Coppieters KT, Von Herrath MG. Non-antigenic and antigenic interventions in type 1 diabetes. Hum Vaccin Immunother 2013; 10:838-46. [PMID: 24165565 PMCID: PMC4896560 DOI: 10.4161/hv.26890] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Type 1 diabetes (T1D) results from autoimmune destruction of the pancreatic β-cells. Current T1D therapies are exclusively focused on regulating glycemia rather than the underlying immune response. A handful of trials have sought to alter the clinical course of T1D using various broad immune-suppressors, e.g., cyclosporine A and azathioprine.1–3 The effect on β-cell preservation was significant, however, these therapies were associated with unacceptable side-effects. In contrast, more recent immunomodulators, such as anti-CD3 and antigenic therapies such as DiaPep277, provide a more targeted immunomodulation and have been generally well-tolerated and safe; however, as a monotherapy there appear to be limitations in terms of therapeutic benefit. Therefore, we argue that this new generation of immune-modifying agents will likely work best as part of a combination therapy. This review will summarize current immune-modulating therapies under investigation and discuss how to move the field of immunotherapy in T1D forward.
Collapse
Affiliation(s)
- Anna K E Rydén
- Type 1 Diabetes R&D Center; Novo Nordisk Inc.; Seattle, WA USA; Pacific Northwest Diabetes Research Institute; Seattle, WA USA
| | | | | | | |
Collapse
|
25
|
Xu D, Prasad S, Miller SD. Inducing immune tolerance: a focus on Type 1 diabetes mellitus. ACTA ACUST UNITED AC 2013; 3:415-426. [PMID: 24505231 DOI: 10.2217/dmt.13.36] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tolerogenic strategies that specifically target diabetogenic immune cells in the absence of complications of immunosuppression are the desired treatment for the prevention or even reversal of Type 1 diabetes (T1D). Antigen (Ag)-based therapies must not only suppress disease-initiating diabetogenic T cells that are already activated, but, more importantly, prevent activation of naive auto-Ag-specific T cells that may become autoreactive through epitope spreading as a result of Ag liberation from damaged islet cells. Therefore, identification of auto-Ags relevant to T1D initiation and progression is critical to the design of effective Ag-specific therapies. Animal models of T1D have been successfully employed to identify potential diabetogenic Ags, and have further facilitated translation of Ag-specific tolerance strategies into human clinical trials. In this review, we highlight important advances using animal models in Ag-specific T1D immunotherapies, and the application of the preclinical findings to human subjects. We provide an up-to-date overview of the strengths and weaknesses of various tolerance-inducing strategies, including infusion of soluble Ags/peptides by various routes of delivery, genetic vaccinations, cell- and inert particle-based tolerogenic approaches, and various other strategies that target distinct tolerance-inducing pathways.
Collapse
Affiliation(s)
- Dan Xu
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | - Suchitra Prasad
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
26
|
Clemente-Casares X, Tsai S, Huang C, Santamaria P. Antigen-specific therapeutic approaches in Type 1 diabetes. Cold Spring Harb Perspect Med 2013; 2:a007773. [PMID: 22355799 DOI: 10.1101/cshperspect.a007773] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Development of strategies capable of specifically curbing pathogenic autoimmune responses in a disease- and organ-specific manner without impairing foreign or tumor antigen-specific immune responses represents a long sought-after goal in autoimmune disease research. Unfortunately, our current understanding of the intricate details of the different autoimmune diseases that affect mankind, including type 1 diabetes, is rudimentary. As a result, progress in the development of the so-called "antigen-specific" therapies for autoimmunity has been slow and fraught with limitations that interfere with bench-to-bedside translation. Absent or incomplete understanding of mechanisms of action and lack of adequate immunological biomarkers, for example, preclude the rational design of effective drug development programs. Here, we provide an overview of antigen-specific approaches that have been tested in preclinical models of T1D and, in some cases, human subjects. The evidence suggests that effective translation of these approaches through clinical trials and into patients will continue to meet with failure unless detailed mechanisms of action at the level of the organism are defined.
Collapse
Affiliation(s)
- Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre, University of Calgary, NW Calgary, Alberta T2N 4N1, Canada
| | | | | | | |
Collapse
|
27
|
Recombinant heat shock protein 60 (Hsp60/GroEL) of Salmonella enterica serovar Typhi elicits cross-protection against multiple bacterial pathogens in mice. Vaccine 2013; 31:2035-41. [DOI: 10.1016/j.vaccine.2013.02.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 01/22/2013] [Accepted: 02/25/2013] [Indexed: 11/17/2022]
|
28
|
Zucchi FCR, Tsanaclis AMC, Moura-Dias Q, Silva CL, Pelegrini-da-Silva A, Neder L, Takayanagui OM. Modulation of angiogenic factor VEGF by DNA-hsp65 vaccination in a murine CNS tuberculosis model. Tuberculosis (Edinb) 2013; 93:373-80. [PMID: 23491717 DOI: 10.1016/j.tube.2013.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 02/03/2013] [Indexed: 12/22/2022]
Abstract
Tuberculosis (TB) is a serious public health problem. Development of experimental models and vaccines are essential to elucidate physiopathological mechanisms and to control the disease. Vascular endothelial growth factor (VEGF) is a potent activator of vascular permeability and angiogenesis. VEGF seems to participate in breakdown of the blood brain-barrier (BBB) in tuberculous meningitis (TBM), contributing to worsening of disease. Therefore, the objective here was to extent the characterization of our previously described murine model of central nervous system TB (CNS-TB) by describing the VEGF participation in the CNS disease, and suggesting a vaccination plan in mice. Plasmid encoding DNA protein antigen DNA-hsp65 has been described as a protector against TB infection and was used here to test its effectiveness in the prevention of VEGF production and TB disease. Vaccinated mice and its controls were injected with Mycobacterium bovis bacillus Calmette-Guerin (BCG) in cerebellum. Four weeks after BCG injection, mice were perfused and brains were paraffin-embedded for VEGF expression analysis. We observed VEGF immunohistochemical expression in TBM and granulomas in non-vaccinated mice. The DNA-hsp65 treatment blocked the expression of VEGF in mice TBM. Therefore, our murine model indicated the VEGF participation in the physiopathology of CNS-TB and the potential prevention of the DNA-hsp65 in the disease progression.
Collapse
Affiliation(s)
- Fabíola C R Zucchi
- Department of Neuroscience, University of São Paulo at Ribeirão Preto School of Medicine, Ribeirão Preto, SP, Brazil.
| | | | | | | | | | | | | |
Collapse
|
29
|
Coppieters KT, Sehested Hansen B, von Herrath MG. Clinical potential of antigen-specific therapies in type 1 diabetes. Rev Diabet Stud 2012; 9:328-37. [PMID: 23804270 PMCID: PMC3740700 DOI: 10.1900/rds.2012.9.328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/21/2013] [Accepted: 02/08/2013] [Indexed: 12/31/2022] Open
Abstract
In type 1 diabetes (T1D), pancreatic beta-cells are attacked and destroyed by the immune system, which leads to a loss of endogenous insulin secretion. The desirable outcome of therapeutic intervention in autoimmune diseases is the restoration of immune tolerance to prevent organ damage. Past trials with immune suppressive drugs highlight the fact that T1D is in principle a curable condition. However, the barrier in T1D therapy in terms of drug safety is set particularly high because of the predominantly young population and the good prognosis associated with modern exogenous insulin therapy. Thus, there is a general consensus that chronic immune suppression is associated with unacceptable long-term safety risks. On the other hand, immune-modulatory biologicals have recently failed to confer significant protection in phase 3 clinical trials. However, the concept of antigen-specific tolerization may offer a unique strategy to safely induce long-term protection against T1D. In this review, we analyze the potential reasons for the failure of the different tolerization therapies, and describe how the concept of antigen-specific toleraization may overcome the obstacles associated with clinical therapy in T1D.
Collapse
Affiliation(s)
| | | | - Matthias G. von Herrath
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
30
|
Hassan GA, Sliem HA, Ellethy AT, Salama MES. Role of immune system modulation in prevention of type 1 diabetes mellitus. Indian J Endocrinol Metab 2012; 16:904-909. [PMID: 23226634 PMCID: PMC3510959 DOI: 10.4103/2230-8210.102989] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
An increased incidence of Type 1 diabetes mellitus (T1DM) is expected worldwide. Eventually, T1DM is fatal unless treated with insulin. The expansion of interventions to prevent diabetes and the use of alternative treatments to insulin is a dream to be fulfilled. The pathophysiology in T1DM is basically a destruction of beta cells in the pancreas, regardless of which risk factors or causative entities have been present. Individual risk factors can have separate patho-physiological processes to, in turn, cause this beta cell destruction. Currently, autoimmunity is considered the major factor in the pathophysiology of T1DM. In a genetically susceptible individual, viral infection may stimulate the production of antibodies against a viral protein that trigger an autoimmune response against antigenically similar beta cell molecules. Many components of the immune system have been implicated in autoimmunity leading to β-cell destruction, including cytotoxic and helper T-cells, B-cells, macrophages, and dendritic cells. The inflammatory process in early diabetes is thought to be initiated and propagated by the effect of Th1-secreted cytokines (e.g. g interferon) and suppressed by Th2-secreted antiinflammatory cytokines (interleukins). Structure and function of β-cell may be modulated by using Th1/Th2-secreted cytokines. Several experimental and clinical trials of applying GAD65, Hsp60, peptide-MHC, pepetide-277 immunization, anti-CD3 infusion, and interleukins to modulate immune response in T1DM were done. Applying such trials in patients with prediabetes, will most likely be the future key in preventing Type 1 autoimmune diabetes.
Collapse
Affiliation(s)
- Gamal Abdulrhman Hassan
- Department of Anatomy and Genetics, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Hamdy Ahmad Sliem
- Department of Internal Medicine, College of Dentistry, Qassim University, Saudi Arabia
| | | | - Mahmoud El-Sawy Salama
- Department of Basic Oral and Medical Science, College of Dentistry, Qassim University, Saudi Arabia
| |
Collapse
|
31
|
Development of a quantitative methylation-specific polymerase chain reaction method for monitoring beta cell death in type 1 diabetes. PLoS One 2012; 7:e47942. [PMID: 23144715 PMCID: PMC3483298 DOI: 10.1371/journal.pone.0047942] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 09/25/2012] [Indexed: 01/05/2023] Open
Abstract
DNA methylation is a mechanism by which cells control gene expression, and cell-specific genes often exhibit unique patterns of DNA methylation. We previously reported that the mouse insulin-2 gene (Ins2) promoter has three potential methylation (CpG) sites, all of which are unmethylated in insulin-producing cells but methylated in other tissues. In this study we examined Ins2 exon 2 and found a similar tissue-specific methylation pattern. These methylation patterns can differentiate between DNA from insulin-producing beta cells and other tissues. We hypothesized that damaged beta cells release their DNA into circulation at the onset of type 1 diabetes mellitus (T1DM) and sought to develop a quantitative methylation-specific polymerase chain reaction (qMSP) assay for circulating beta cell DNA to monitor the loss of beta cells. Methylation-specific primers were designed to interrogate two or more CpG in the same assay. The cloned mouse Ins2 gene was methylated in vitro and used for development of the qMSP assay. We found the qMSP method to be sensitive and specific to differentiate between insulin-producing cells and other tissues with a detection limit of 10 copies in the presence of non-specific genomic DNA background. We also compared different methods for data analysis and found that the Relative Expression Ratio method is the most robust method since it incorporates both a reference value to normalize day-to-day variability as well as PCR reaction efficiencies to normalize between the methylation-specific and bisulfite-specific components of the calculations. The assay was applied in the streptozotocin-treated diabetic mouse model and detected a significant increase in circulating beta cell DNA before the rise in blood glucose level. These results demonstrate that this qMSP assay can be used for monitoring circulating DNA from insulin-producing cells, which will provide the basis for development of assays to detect beta cell destruction in early T1DM.
Collapse
|
32
|
Kverka M, Tlaskalova-Hogenova H. Two faces of microbiota in inflammatory and autoimmune diseases: triggers and drugs. APMIS 2012; 121:403-21. [DOI: 10.1111/apm.12007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 09/13/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Miloslav Kverka
- Department of Immunology and Gnotobiology, Institute of Microbiology; Academy of Sciences of the Czech Republic; Prague; Czech Republic
| | - Helena Tlaskalova-Hogenova
- Department of Immunology and Gnotobiology, Institute of Microbiology; Academy of Sciences of the Czech Republic; Prague; Czech Republic
| |
Collapse
|
33
|
M. paratuberculosis Heat Shock Protein 65 and Human Diseases: Bridging Infection and Autoimmunity. Autoimmune Dis 2012; 2012:150824. [PMID: 23056923 PMCID: PMC3465878 DOI: 10.1155/2012/150824] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/27/2012] [Accepted: 08/29/2012] [Indexed: 01/19/2023] Open
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) is the known infectious cause of Johne's disease, an enteric inflammatory disease mostly studied in ruminant animals. MAP has also been implicated in the very similar Crohn's disease of humans as well as sarcoidosis. Recently, MAP has been associated with juvenile sarcoidosis (Blau syndrome), autoimmune diabetes, autoimmune thyroiditis, and multiple sclerosis. While it is intuitive to implicate MAP in granulomatous diseases where the microbe participates in the granuloma, it is more difficult to assign a role for MAP in diseases where autoantibodies are a primary feature. MAP may trigger autoimmune antibodies via its heat shock proteins. Mycobacterial heat shock protein 65 (HSP65) is an immunodominant protein that shares sequential and conformational elements with several human host proteins. This molecular mimicry is the proposed etiopathology by which MAP stimulates autoantibodies associated with autoimmune (type 1) diabetes, autoimmune (Hashimoto's) thyroiditis, and multiple sclerosis. This paper proposes that MAP is a source of mycobacterial HSP65 and acts as a trigger of autoimmune disease.
Collapse
|
34
|
Immune recognition of the 60kD heat shock protein: implications for subsequent fertility. Infect Dis Obstet Gynecol 2012; 4:152-8. [PMID: 18476087 PMCID: PMC2364488 DOI: 10.1155/s1064744996000336] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/1996] [Accepted: 10/01/1996] [Indexed: 11/18/2022] Open
Abstract
The 60kD heat shock protein (hsp60) is a highly conserved protein and a dominant antigen of
most pathogenic bacteria. In some women, chronic or repeated upper genital tract infections with
Chlamydia trachomatis, and possibly with other microorganisms, induces immune sensitization to
epitopes of hsp60 that are present in both the microbial and human hsp60. Once a woman becomes
sensitized to these conserved epitpes, any subsequent induction of human or bacterial hsp60
expression will reactivate hsp60-sensitized lymphocytes and initiate a pro-inflammatory immune
response. Hsp60 is expressed during the early stages of pregnancy, by both the embryo and the
maternal decidua. We examined, therefore, whether women who were sensitized to hsp60 experienced
less successful pregnancy outcomes compared to women who were not sensitized to this
antigen. In women undergoing in vitro fertilization (IVF), the presence of cervical IgA antibodies
reactive with the C. trachomatis hsp60 correlated with implantation failure after embryo transfer.
Further analysis revealed that an immunodominant epitope for these IgA antibodies was an hsp60
epitope shared between C. trachomatis and man. In subsequent studies of women not undergoing
IVF, cervical IgA antibodies to the human hsp60 were identified in 13 of 91 reproductive age
women. This antibody was most prevalent in those women with a history of primary infertility
(p = 0.003). In addition, cervical anti-hsp60 IgA correlated with the detection of the pro-inflammatory
cytokines interferon-γ (p = 0.001) and tumor necrosis factor-α (p = 0.02) in the cervix.
Conversely, women with proven fertility had the highest prevalence of the anti-inflammatory cytokine,
interleukin 10, in their cervices (p = 0.001). In an analysis of serum samples in a third study,
women with a history of two or more consecutive first trimester spontaneous abortions had a higher
prevalence (p = 0.01) of IgG antibodies to the human hsp60 (36.8%) than did age matched fertile
women (11.1%) or women with primary infertility (11.8%). Immune sensitization to epitopes expressed
by the human hsp60 may reduce the probability of a successful pregnancy outcome due to
reactivation of hsp60-reactive lymphocytes, induction of a pro-inflammatory cytokine response and
interference with early embryo development and/or implantation.
Collapse
|
35
|
Singh I, Yadav AR, Mohanty KK, Katoch K, Bisht D, Sharma P, Sharma B, Gupta UD, Sengupta U. Molecular mimicry between HSP 65 of Mycobacterium leprae and cytokeratin 10 of the host keratin; role in pathogenesis of leprosy. Cell Immunol 2012; 278:63-75. [PMID: 23121977 DOI: 10.1016/j.cellimm.2012.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 06/09/2012] [Accepted: 06/26/2012] [Indexed: 12/12/2022]
Abstract
Mycobacteria are known to induce autoimmune response in the host. Anti-host keratrin antibodies (AkAbs) might be responsible for the autoimmune phenomena in leprosy patients as majority of leprosy lesions are manifested in the skin and occurrence of keratosis is not an uncommon feature. The aim of this study was to find out the level of AkAbs in leprosy patients across the spectrum and to explore its correlation with the clinical manifestation of the disease. Further, mimicking epitopes of keratin and Mycobacterium leprae components were characterized. We screened 140 leprosy patients (27 BT, 28 BL, 41 LL, 25 T1R, 19 ENL), 74 healthy controls (HC) and 3 psoriasis patients as positive control. Highest AkAbs level was observed in the psoriasis patients followed by T1R, LL, BL, ENL, TT/BT. AkAbs level was significantly (p<0.05) higher in all the groups of leprosy patients except TT/BT in comparison to HC. Significant positive correlation was found between number of lesions and level of AkAbs in leprosy patients. Highest lympho-proliferation for keratin protein was observed in T1R, followed by BL/LL, TT/BT, ENL. Lympho-proliferation was significantly (p<0.05) higher in all groups of leprosy patients except ENL in comparison to HC. Interestingly, it was noted that hyperimmunization of inbred strains of female BALB/c mice and rabbit with M. leprae soluble antigen (MLSA) induce higher level of AkAbs. The percentage of FoxP3(+) expressing Treg cells (total CD4(+)CD25(+)FoxP3(+) andCD4(+)CD25(+hi)FoxP3(+)) in splenocytes and lymph nodes of hyperimmunized mice were declined in comparison to control mice. Further, it was found that this autoimmune response can be adoptively transferred in naïve mice by splenocytes and lymph node cells as well as T cells. Comparative molecular characterization between keratin and MLSA noted a cross-reactivity/similarity between these two antigens. The cross-reactive protein of keratin was found to be in molecular weight range ≈74-51kDa and at pI 4.5 while the cross-reactive protein of MLSA was found to be in molecular weight ≈65kDa and at pI 4-4.5. Cross-reactive protein of keratin and MLSA was identified and characterized by MALDI-TOF/TOF analysis and Mascot software. It was found that the keratin (host protein) which reacted with anti-M. leprae sera is cytokeratin-10 and MLSA which reacted with anti-keratin sera is heat shock protein 65 (HSP 65). Seven B-cell epitopes of cytokeratin-10 and HSP 65 was found to be similar by multiple sequence alignment using ClustalW server and out of which 6 B-cell epitopes were found to be on the surface of HSP 65. In conclusion, our study provides evidence for the existence of molecular mimicry between cytokeratin-10 of keratin (host protein) and 65kDa HSP (groEL2) of M. leprae. Presence of heightened CMI response of leprosy patients to keratin and positive correlation of AkAbs level with number of lesions of leprosy patients showed the clinical evidence for its role in the pathogenesis in leprosy.
Collapse
Affiliation(s)
- Itu Singh
- Department of Immunology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, (ICMR) Tajganj, Agra, India
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hinke SA. Inverse vaccination with islet autoantigens to halt progression of autoimmune diabetes. Drug Dev Res 2011. [DOI: 10.1002/ddr.20488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
37
|
The complex exocrine-endocrine relationship and secondary diabetes in exocrine pancreatic disorders. J Clin Gastroenterol 2011; 45:850-61. [PMID: 21897283 DOI: 10.1097/mcg.0b013e31822a2ae5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pancreas is a dual organ with exocrine and endocrine functions. The interrelationship of the endocrine-exocrine parts of the pancreas is a complex one, but recent clinical and experimental studies have expanded our knowledge. Many disorders primarily of the exocrine pancreas, often solely in the clinical realm of gastroenterologists are associated with diabetes mellitus (DM). Although, the DM becoming disorders are often grouped with type 2 diabetes, the pathogenesis, clinical manifestations and management differ. We review here data on the association of exocrine-endocrine pancreas, the many hormones of the pancreas and their possible effects on the exocrine functions followed by data on the epidemiology, pathogenesis, and management of DM in chronic pancreatitis, cystic fibrosis, pancreatic cancer, and clinical states after pancreatic surgery.
Collapse
|
38
|
Parada CA, Portaro F, Marengo EB, Klitzke CF, Vicente EJ, Faria M, Sant’Anna OA, Fernandes BL. Autolytic Mycobacterium leprae Hsp65 fragments may act as biological markers for autoimmune diseases. Microb Pathog 2011; 51:268-76. [DOI: 10.1016/j.micpath.2011.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 05/31/2011] [Accepted: 06/02/2011] [Indexed: 10/18/2022]
|
39
|
Sané F, Moumna I, Hober D. Group B coxsackieviruses and autoimmunity: focus on Type 1 diabetes. Expert Rev Clin Immunol 2011; 7:357-66. [PMID: 21595602 DOI: 10.1586/eci.11.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Group B coxsackieviruses (CVB) and/or their components have been found in the blood and pancreas of patients with Type 1 diabetes (T1D). CVB infections lead to the activation of the innate and adaptive immune systems, which can result in the induction or aggravation of autoimmune processes. Persistent and/or repeated infections of pancreas islet β cells with CVB and the resulting production of IFN-α and inflammatory mediators, combined with a predisposed genetic background, may induce bystander activation of autoimmune effector T cells and an autoreactive response to islet self-antigens through molecular mimicry. Moreover, the antibody-dependent enhancement of CVB infection of monocytes, as well as infection of the thymus can intervene in the pathogenesis of T1D. In contrast with the deleterious effect of CVB, it has been shown that these viruses can protect against the development of T1D under certain experimental conditions. The role of CVB in autoimmunity is complex, and therefore a better understanding of the inducer versus protective effects of these viruses in T1D will help to design new strategies to treat and prevent the disease.
Collapse
Affiliation(s)
- Famara Sané
- Laboratory of Virology EA3610, University Lille 2, Faculty of Medecine, CHRU Lille, 59037 Lille, France
| | | | | |
Collapse
|
40
|
Nicholas D, Odumosu O, Langridge WHR. Autoantigen based vaccines for type 1 diabetes. DISCOVERY MEDICINE 2011; 11:293-301. [PMID: 21524383 PMCID: PMC6474774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Type 1 diabetes is an organ-specific autoimmune disease caused by chronic inflammation (insulitis), which damages the insulin producing β-cells of the pancreatic Islets of Langerhans. Dendritic cells (DCs) are generally the first cells of the immune system to process β-cell autoantigens and, by promoting autoreactivity, play a major role in the onset of insulitis. Although no cure for diabetes presently exists, the onset of insulitis can be diminished in the non-obese diabetic (NOD) mouse type 1 diabetes model by inoculation with endogenous β-cell autoantigens. These include the single peptide vaccines insulin, GAD(65) (glutamic acid decarboxylase), and DiaPep277 (an immunogenic peptide from the 60-kDa heat shock protein). DiaPep277 is the only autoantigen so far to demonstrate positive results in human clinical trials. Diamyd (an alum adjuvant + recombinant GAD(65) protein formulation) has shown great promise for suppressing β-cell autoreactivity in phase I and II clinical trials. While Diamyd preserved residual insulin secretion in early-onset type 1 diabetes patients, it did not reduce the amounts of insulin required to maintain euglycemia. Recently, multi-component vaccines composed of the anti-inflammatory cytokine (IL-10) and insulin or GAD(55) linked to an immunostimulatory molecule, the cholera toxin B subunit, were shown to safely and completely inhibit diabetes onset in NOD mice. This result suggests that multi-component vaccine strategies are promising for prevention and reversal of diabetes autoimmunity in humans. Here we focus on the development of autoantigen vaccines for type 1 diabetes and demonstrate that multi-component vaccines are promising candidates for type 1 diabetes clinical studies.
Collapse
Affiliation(s)
- Dequina Nicholas
- Center for Health Disparities and Molecular Medicine, Department of Biochemistry, Loma Linda University School of Medicine, California 92354, USA
| | | | | |
Collapse
|
41
|
Rodríguez-Narciso C, Pérez-Tapia M, Rangel-Cano RM, Silva CL, Meckes-Fisher M, Salgado-Garciglia R, Estrada-Parra S, López-Gómez R, Estrada-García I. Expression of Mycobacterium leprae HSP65 in tobacco and its effectiveness as an oral treatment in adjuvant-induced arthritis. Transgenic Res 2011; 20:221-9. [PMID: 20526808 DOI: 10.1007/s11248-010-9404-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 05/13/2010] [Indexed: 12/22/2022]
Abstract
Transgenic plants are able to express molecules with antigenic properties. In recent years, this has led the pharmaceutical industry to use plants as alternative systems for the production of recombinant proteins. Plant-produced recombinant proteins can have important applications in therapeutics, such as in the treatment of rheumatoid arthritis (RA). In this study, the mycobacterial HSP65 protein expressed in tobacco plants was found to be effective as a treatment for adjuvant-induced arthritis (AIA). We cloned the hsp65 gene from Mycobacterium leprae into plasmid pCAMBIA 2301 under the control of the double 35S promoter from cauliflower mosaic virus. Agrobacterium tumefaciens bearing the pChsp65 plasmid was used to transform tobacco plants. Incorporation of the hsp65 gene was confirmed by PCR, reverse transcription-PCR, histochemistry, and western blot analyses in several transgenic lines of tobacco plants. Oral treatment of AIA rats with the HSP65 protein allowed them to recover body weight and joint inflammation was reduced. Our results suggest a synergistic effect between the HSP65 expressed protein and metabolites presents in tobacco plants.
Collapse
Affiliation(s)
- César Rodríguez-Narciso
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Edificio B1, Francisco J Mújica S/N Col. Felicitas del Rio, CP 58060, Morelia, Michoacán, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
King C, Sarvetnick N. The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions. PLoS One 2011; 6:e17049. [PMID: 21364875 PMCID: PMC3045412 DOI: 10.1371/journal.pone.0017049] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 01/14/2011] [Indexed: 01/23/2023] Open
Abstract
In the NOD mouse, the incidence of type-1 diabetes is thought to be influenced by the degree of cleanliness of the mouse colony. Studies collectively demonstrate that exposure to bacterial antigen or infection in the neonatal period prevents diabetes [1], [2], [3], [4], [5], [6], [7], [8], [9], [10], supporting the notion that immunostimulation can benefit the maturation of the postnatal immune system [11]. A widely accepted extrapolation from this data has been the notion that NOD mice maintained under germ-free conditions have an increased incidence of diabetes. However, evidence supporting this influential concept is surprisingly limited [12]. In this study, we demonstrate that the incidence of diabetes in female NOD mice remained unchanged under germ-free conditions. By contrast, a spontaneous monoculture with a gram-positive aerobic spore-forming rod delayed the onset and reduced the incidence of diabetes. These findings challenge the view that germ-free NOD mice have increased diabetes incidence and demonstrate that modulation of intestinal microbiota can prevent the development of type-1 diabetes.
Collapse
Affiliation(s)
- Cecile King
- Department of Immunology, The Scripps Research Institute, La Jolla, California, United States of America.
| | | |
Collapse
|
43
|
Quintana FJ, Cohen IR. The HSP60 immune system network. Trends Immunol 2010; 32:89-95. [PMID: 21145789 DOI: 10.1016/j.it.2010.11.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 11/05/2010] [Accepted: 11/05/2010] [Indexed: 02/06/2023]
Abstract
Heat shock proteins (HSPs) were initially discovered as participants in the cellular response to stress. It is now clear, however, that self and microbial HSPs also play an important role in the control of the immune response. Here, we focus on HSP60 and its interactions with both the innate and adaptive immune system in mammals. We also consider that circulating HSP60 and the quantities and specificities of serum antibodies to HSP60 provide a biomarker to monitor the immune status of the individual. Thus, the dual role of HSP60 as an immune modulator and a biomarker, provides an opportunity to modulate immunity for therapeutic purposes, and to monitor the immune response in health and disease.
Collapse
Affiliation(s)
- Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston MA, USA.
| | | |
Collapse
|
44
|
Li Q, Ge C, Liu R, Zhang K, Wu G, Huo W. Administration of dendritic cells dual expressing DcR3 and GAD65 mediates the suppression of T cells and induces long-term acceptance of pancreatic-islet transplantation. Vaccine 2010; 28:8300-5. [DOI: 10.1016/j.vaccine.2010.09.094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 09/17/2010] [Accepted: 09/26/2010] [Indexed: 12/22/2022]
|
45
|
Abstract
The widely used nonobese diabetic (NOD) mouse model of autoimmune (Type 1) diabetes mellitus shares multiple characteristics with the human disease, and studies employing this model continue to yield clinically relevant and important information. Here, we review some of the recent key findings obtained from NOD mouse investigations that have both advanced our understanding of disease pathogenesis and suggested new therapeutic targets and approaches. Areas discussed include antigen discovery, identification of genes and pathways contributing to disease susceptibility, development of strategies to image islet inflammation and the testing of therapeutics. We also review recent technical advances that, combined with an improved understanding of the NOD mouse model's limitations, should work to ensure its popularity, utility and relevance in the years ahead.
Collapse
Affiliation(s)
- Rodolfo José Chaparro
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Teresa P DiLorenzo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
46
|
Babad J, Geliebter A, DiLorenzo TP. T-cell autoantigens in the non-obese diabetic mouse model of autoimmune diabetes. Immunology 2010; 131:459-65. [PMID: 21039471 DOI: 10.1111/j.1365-2567.2010.03362.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The non-obese diabetic (NOD) mouse model of autoimmune (type 1) diabetes has contributed greatly to our understanding of disease pathogenesis and has facilitated the development and testing of therapeutic strategies to combat the disease. Although the model is a valuable immunological tool in its own right, it reaches its fullest potential in areas where its findings translate to the human disease. Perhaps the foremost example of this is the field of T-cell antigen discovery, from which diverse benefits can be derived, including the development of antigen-specific disease interventions. The majority of NOD T-cell antigens are also targets of T-cell autoimmunity in patients with type 1 diabetes, and several of these are currently being evaluated in clinical trials. Here we review the journeys of these antigens from bench to bedside. We also discuss several recently identified NOD T-cell autoantigens whose translational potential warrants further investigation.
Collapse
Affiliation(s)
- Jeffrey Babad
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
47
|
Abstract
Not only T cells but also B cells play a role in the autoimmune process. Both monoclonal antiCD3 and antiCD20 antibodies seem efficacious. However, such treatments need to be refined to minimize adverse events. Use of autoantigens to create tolerance is a concept with great potential. GAD65 treatment has shown efficacy without adverse events thus far, and administration of the insulin B chain shows interesting immunologic effects. Other more or less speculative approaches to modulate the immune process need further studies with good design. Risks that are too serious cannot be motivated. In addition, as the beta cells may die even though the autoimmune process is stopped, protective measures may be valuable (eg, active insulin treatment, and perhaps interleukin-1 receptor antagonists to reduce the nonautoimmune inflammation). Combination of immune intervention, protection of the beta cells, and stimulation of regeneration may lead to a milder disease or even a cure in the future, and prevention is no longer unrealistic.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Department of Clinical and Experimental Medicine, Division of Pediatrics, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
48
|
Wood KL, Nunley DR, Moffatt-Bruce S, Pope-Harman A, Huang Q, Shamo EN, Phillips GS, Baran C, Batra S, Marsh CB, Doseff AI. The role of heat shock protein 27 in bronchiolitis obliterans syndrome after lung transplantation. J Heart Lung Transplant 2010; 29:786-91. [PMID: 20456980 PMCID: PMC2902709 DOI: 10.1016/j.healun.2010.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 01/25/2010] [Accepted: 03/02/2010] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Heat shock proteins (Hsps) are a family of evolutionary conserved proteins classified according to their size as small and large Hsps. They have a cytoprotective role and have been shown to be immunogenic molecules. In addition, self-reactivity to Hsps has been implicated in various autoimmune diseases and in the development of alloimmunity. This study examined the relationship of large and small Hsps and anti-Hsp antibodies in lung transplant (LTx) recipients who have bronchiolitis obliterans syndrome (BOS). METHODS Anti-Hsp27 and Hsp70 antibodies, and Hsp27, Hsp60, and Hsp70 protein expression levels were evaluated in serum and bronchoalveolar lavage samples collected from 8 LTx recipients with established BOS and 8 recipients without BOS (controls). Serum from 8 healthy individuals was examined for Hsp levels as a comparison. RESULTS Elevated serum Hsp27 levels were observed in recipients with BOS compared with controls or healthy individuals, whereas Hsp70 and Hsp60 expression showed no difference. Anti-Hsp27 antibody levels were significantly higher in the BAL of recipients with BOS than in those without BOS. In contrast, anti-Hsp70 antibodies levels in serum or BAL showed no difference between groups. CONCLUSIONS These results support the novel concept that Hsp27, but not the classic Hsp60 and Hsp70, may be associated with the development BOS. The expression of anti-Hsp27 antibodies found only in the BAL fluid suggests a local response occurring at the level of the alveoli and terminal airways.
Collapse
Affiliation(s)
- Karen L Wood
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
HIV-1 protease has a genetic T-cell adjuvant effect which is negatively regulated by proteolytic activity. J Virol 2010; 84:7743-9. [PMID: 20484507 DOI: 10.1128/jvi.00747-10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV protease (PR) mediates the processing of human immunodeficiency virus (HIV) polyproteins and is necessary for the viral production. Recently, HIV PR was shown to possess both cytotoxic and chaperone like activity. We demonstrate here that HIV PR can serve as a genetic adjuvant that enhances the HIV Env and human papillomavirus (HPV) DNA vaccine-induced T-cell response in a dose-dependent manner, only when codelivered with DNA vaccine. Interestingly, the T-cell adjuvant effects of HIV PR were increased by introducing several mutations that inhibited its proteolytic activity, indicating that the adjuvant properties were inversely correlated with its proteolytic activity. Conversely, the introduction of a mutation in the flap region of HIV PR limiting the access to the core domain of HIV PR inhibited the T-cell adjuvant effect, suggesting that the HIV PR chaperone like activity may play a role in mediating T-cell adjuvant properties. A similar adjuvant effect was also observed in adenovirus vaccine, indicating vaccine type independency. These findings suggest that HIV PR can modulate T-cell responses elicited by a gene-based vaccine positively by inherent chaperone like activity and negatively by its proteolytic activity.
Collapse
|
50
|
Liang J, Aihua Z, Yu W, Yong L, Jingjing L. HSP65 serves as an immunogenic carrier for a diabetogenic peptide P277 inducing anti-inflammatory immune response in NOD mice by nasal administration. Vaccine 2010; 28:3312-7. [DOI: 10.1016/j.vaccine.2010.02.100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 02/22/2010] [Accepted: 02/23/2010] [Indexed: 11/30/2022]
|