1
|
Li T, Li R, Zhang T, Zhang H, Song X, Zhai X, Wang J, Xing B, Hou X, Wei L. Identification, cloning, and characterization of Cherry Valley duck CD4 and its antiviral immune responses. Poult Sci 2021; 100:101262. [PMID: 34273645 PMCID: PMC8287243 DOI: 10.1016/j.psj.2021.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 05/12/2021] [Indexed: 11/19/2022] Open
Abstract
CD4 protein is a single chain transmembrane glycoprotein and has a broad functionality beyond cell-mediated immunity. In this study, we cloned the full-length coding sequence (CDS) of duck CD4 (duCD4) and analyzed its sequence and structure, and expression levels in several tissues. It consists of 1,449 nucleotides and encodes a 482 amino acid protein. The putative protein of duCD4 consisted of an N-terminal signal peptide, three immunoglobulins and one immunoglobulins-like domain in its central, one terminal transmembrane region, and a C-terminal domain of the CD4 T cell receptor. The duCD4 also has the typical signature “CXC” of CD4s. The multiple sequence alignment suggests duCD4 has four potential N-glycosylation sites and the phylogenetic analysis suggests duCD4 shares greater similarity with avian than other vertebrates. Quantitative real-time PCR analysis showed that duCD4 mRNA transcripts are widely distributed in the healthy Cherry Valley duck, and the highest level in the thymus. During the virus infection, the obvious change of duCD4 expression was observed in the spleen, lung and brain, which suggesting that duCD4 could be involved in the host's immune response to multiple types of viruses. Our research studied the characterization, tissue distribution, and antiviral immune responses of duCD4.
Collapse
Affiliation(s)
- Tianxu Li
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Rong Li
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Tingting Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, College of Basic Medical Sciences, Shandong First Medical University, Tai'an City, Shandong Province 271000, China
| | - Huihui Zhang
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xingdong Song
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xinyu Zhai
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Jinchao Wang
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Bin Xing
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xiaolan Hou
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Liangmeng Wei
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, College of Basic Medical Sciences, Shandong First Medical University, Tai'an City, Shandong Province 271000, China.
| |
Collapse
|
2
|
Vaccination by Two DerG LEAPS Conjugates Incorporating Distinct Proteoglycan (PG, Aggrecan) Epitopes Provides Therapy by Different Immune Mechanisms in a Mouse Model of Rheumatoid Arthritis. Vaccines (Basel) 2021; 9:vaccines9050448. [PMID: 34063326 PMCID: PMC8147650 DOI: 10.3390/vaccines9050448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/14/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) can be initiated and driven by immune responses to multiple antigenic epitopes including those in cartilage proteoglycan (PG, aggrecan) and type II collagen. RA is driven by T helper 1 (Th1) or Th17 pro-inflammatory T cell responses. LEAPS (Ligand Epitope Antigen Presentation System) DerG peptide conjugate vaccines were prepared using epitopes from PG that elicit immune responses in RA patients: epitope PG70 (DerG-PG70, also designated CEL-4000) and the citrullinated form of another epitope (PG275Cit). The LEAPS peptides were administered alone or together in Seppic ISA51vg adjuvant to mice with PG G1 domain-induced arthritis (GIA), a mouse model of RA. Each of these LEAPS peptides and the combination modulated the inflammatory response and stopped the progression of arthritis in the GIA mouse model. Despite having a therapeutic effect, the DerG-PG275Cit vaccine did not elicit significant antibody responses, whereas DerG-PG70 (alone or with DerG-PG275Cit) induced both therapy and antibodies. Spleen T cells from GIA mice, vaccinated with the DerG LEAPS peptides, preferentially produced anti-inflammatory (IL-4 and IL-10) rather than pro-inflammatory (IFN-γ or IL-17) cytokines in culture. Similarly, cytokines secreted by CD4+ cells of unvaccinated GIA mice, differentiated in vitro to Th2 cells and treated with either or both DerG vaccine peptides, exhibited an anti-inflammatory (IL-4, IL-10) profile. These results suggest that the two peptides elicit different therapeutic immune responses by the immunomodulation of disease-promoting pro-inflammatory responses and that the combination of the two LEAPS conjugates may provide broader epitope coverage and, in some cases, greater efficacy than either conjugate alone.
Collapse
|
3
|
Nine residues in HLA-DQ molecules determine with susceptibility and resistance to type 1 diabetes among young children in Sweden. Sci Rep 2021; 11:8821. [PMID: 33893332 PMCID: PMC8065060 DOI: 10.1038/s41598-021-86229-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 03/04/2021] [Indexed: 11/09/2022] Open
Abstract
HLA-DQ molecules account over 50% genetic risk of type 1 diabetes (T1D), but little is known about associated residues. Through next generation targeted sequencing technology and deep learning of DQ residue sequences, the aim was to uncover critical residues and their motifs associated with T1D. Our analysis uncovered (αa1, α44, α157, α196) and (β9, β30, β57, β70, β135) on the HLA-DQ molecule. Their motifs captured all known susceptibility and resistant T1D associations. Three motifs, “DCAA-YSARD” (OR = 2.10, p = 1.96*10−20), “DQAA-YYARD” (OR = 3.34, 2.69*10−72) and “DQDA-YYARD” (OR = 3.71, 1.53*10−6) corresponding to DQ2.5 and DQ8.1 (the latter two motifs) associated with susceptibility. Ten motifs were significantly associated with resistance to T1D. Collectively, homozygous DQ risk motifs accounted for 43% of DQ-T1D risk, while homozygous DQ resistant motifs accounted for 25% protection to DQ-T1D risk. Of the identified nine residues five were within or near anchoring pockets of the antigenic peptide (α44, β9, β30, β57 and β70), one was the N-terminal of the alpha chain (αa1), one in the CD4-binding region (β135), one in the putative cognate TCR-induced αβ homodimerization process (α157), and one in the intra-membrane domain of the alpha chain (α196). Finding these critical residues should allow investigations of fundamental properties of host immunity that underlie tolerance to self and organ-specific autoimmunity.
Collapse
|
4
|
Zhao LP, Papadopoulos GK, Kwok WW, Moustakas AK, Bondinas GP, Larsson HE, Ludvigsson J, Marcus C, Samuelsson U, Wang R, Pyo CW, Nelson WC, Geraghty DE, Lernmark Å. Motifs of Three HLA-DQ Amino Acid Residues (α44, β57, β135) Capture Full Association With the Risk of Type 1 Diabetes in DQ2 and DQ8 Children. Diabetes 2020; 69:1573-1587. [PMID: 32245799 PMCID: PMC7306123 DOI: 10.2337/db20-0075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
Abstract
HLA-DQA1 and -DQB1 are strongly associated with type 1 diabetes (T1D), and DQ8.1 and DQ2.5 are major risk haplotypes. Next-generation targeted sequencing of HLA-DQA1 and -DQB1 in Swedish newly diagnosed 1- to 18 year-old patients (n = 962) and control subjects (n = 636) was used to construct abbreviated DQ haplotypes, converted into amino acid (AA) residues, and assessed for their associations with T1D. A hierarchically organized haplotype (HOH) association analysis allowed 45 unique DQ haplotypes to be categorized into seven clusters. The DQ8/9 cluster included two DQ8.1 risk and the DQ9 resistant haplotypes, and the DQ2 cluster included the DQ2.5 risk and DQ2.2 resistant haplotypes. Within each cluster, HOH found residues α44Q (odds ratio [OR] 3.29, P = 2.38 * 10-85) and β57A (OR 3.44, P = 3.80 * 10-84) to be associated with T1D in the DQ8/9 cluster representing all ten residues (α22, α23, α44, α49, α51, α53, α54, α73, α184, β57) due to complete linkage disequilibrium (LD) of α44 with eight such residues. Within the DQ2 cluster and due to LD, HOH analysis found α44C and β135D to share the risk for T1D (OR 2.10, P = 1.96 * 10-20). The motif "QAD" of α44, β57, and β135 captured the T1D risk association of DQ8.1 (OR 3.44, P = 3.80 * 10-84), and the corresponding motif "CAD" captured the risk association of DQ2.5 (OR 2.10, P = 1.96 * 10-20). Two risk associations were related to GAD65 autoantibody (GADA) and IA-2 autoantibody (IA-2A) but in opposite directions. CAD was positively associated with GADA (OR 1.56, P = 6.35 * 10-8) but negatively with IA-2A (OR 0.59, P = 6.55 * 10-11). QAD was negatively associated with GADA (OR 0.88; P = 3.70 * 10-3) but positively with IA-2A (OR 1.64; P = 2.40 * 10-14), despite a single difference at α44. The residues are found in and around anchor pockets 1 and 9, as potential T-cell receptor contacts, in the areas for CD4 binding and putative homodimer formation. The identification of three HLA-DQ AAs (α44, β57, β135) conferring T1D risk should sharpen functional and translational studies.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - George K Papadopoulos
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Antonis K Moustakas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, Argostoli, Cephalonia, Greece
| | - George P Bondinas
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| | - Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Claude Marcus
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Samuelsson
- Crown Princess Victoria Children's Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ruihan Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Wyatt C Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University CRC, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
5
|
Zhao LP, Papadopoulos GK, Kwok WW, Xu B, Kong M, Moustakas AK, Bondinas GP, Carlsson A, Elding-Larsson H, Ludvigsson J, Marcus C, Persson M, Samuelsson U, Wang R, Pyo CW, Nelson WC, Geraghty DE, Lernmark Å. Eleven Amino Acids of HLA-DRB1 and Fifteen Amino Acids of HLA-DRB3, 4, and 5 Include Potentially Causal Residues Responsible for the Risk of Childhood Type 1 Diabetes. Diabetes 2019; 68:1692-1704. [PMID: 31127057 PMCID: PMC6692811 DOI: 10.2337/db19-0273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/21/2019] [Indexed: 12/25/2022]
Abstract
Next-generation targeted sequencing of HLA-DRB1 and HLA-DRB3, -DRB4, and -DRB5 (abbreviated as DRB345) provides high resolution of functional variant positions to investigate their associations with type 1 diabetes risk and with autoantibodies against insulin (IAA), GAD65 (GADA), IA-2 (IA-2A), and ZnT8 (ZnT8A). To overcome exceptional DR sequence complexity as a result of high polymorphisms and extended linkage disequilibrium among the DR loci, we applied a novel recursive organizer (ROR) to discover disease-associated amino acid residues. ROR distills disease-associated DR sequences and identifies 11 residues of DRB1, sequences of which retain all significant associations observed by DR genes. Furthermore, all 11 residues locate under/adjoining the peptide-binding groove of DRB1, suggesting a plausible functional mechanism through peptide binding. The 15 residues of DRB345, located respectively in the β49-55 homodimerization patch and on the face of the molecule shown to interact with and bind to the accessory molecule CD4, retain their significant disease associations. Further ROR analysis of DR associations with autoantibodies finds that DRB1 residues significantly associated with ZnT8A and DRB345 residues with GADA. The strongest association is between four residues (β14, β25, β71, and β73) and IA-2A, in which the sequence ERKA confers a risk association (odds ratio 2.15, P = 10-18), and another sequence, ERKG, confers a protective association (odds ratio 0.59, P = 10-11), despite a difference of only one amino acid. Because motifs of identified residues capture potentially causal DR associations with type 1 diabetes, this list of residuals is expected to include corresponding causal residues in this study population.
Collapse
Affiliation(s)
- Lue Ping Zhao
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Corresponding authors: Lue Ping Zhao, ; George K. Papadopoulos, ; and Åke Lernmark,
| | - George K. Papadopoulos
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece, presently known as Department of Agriculture, University of Ioannina, Ioannina, Greece
| | | | - Bryan Xu
- College of Letters and Sciences, University of California, Berkeley, CA
| | - Matthew Kong
- Department of Computer Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - Antonis K. Moustakas
- Department of Food Science and Technology, Ionian University, Argostoli, Cephalonia, Greece
| | - George P. Bondinas
- Laboratory of Biophysics, Biochemistry, Biomaterials and Bioprocessing, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, Arta, Greece, presently known as Department of Agriculture, University of Ioannina, Ioannina, Greece
| | - Annelie Carlsson
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | | | - Johnny Ludvigsson
- Crown Princess Victoria Children’s Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Claude Marcus
- Department of Clinical Science and Education and Institution of Medicine, Clinical Epidemiology, Karolinska Institutet, Solna, Sweden
| | - Martina Persson
- Department of Clinical Science and Education and Institution of Medicine, Clinical Epidemiology, Karolinska Institutet, Solna, Sweden
| | - Ulf Samuelsson
- Crown Princess Victoria Children’s Hospital, Region Östergötland, and Division of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ruihan Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Wyatt C. Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Daniel E. Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
6
|
Wang HY, Cui Z, Pei ZY, Fang SB, Chen SF, Zhu L, Chen M, Chen N, Zhao MH. Risk HLA class II alleles and amino acid residues in myeloperoxidase-ANCA-associated vasculitis. Kidney Int 2019; 96:1010-1019. [PMID: 31471160 DOI: 10.1016/j.kint.2019.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 06/05/2019] [Accepted: 06/19/2019] [Indexed: 01/09/2023]
Abstract
A genome-wide association study (GWAS) indicated that myeloperoxidase-ANCA associated vasculitis (AAV) is associated with HLA-DQ. However, susceptibility alleles in these loci have been under-investigated. Here we genotyped 258 Chinese patients with myeloperoxidase-AAV and 597 healthy control individuals at HLA DRB1, DQA1, DQB1 and DPB1, and extracted the encoded amino acid sequences from the IMGT/HLA database. The replication cohort included 97 cases and 107 controls. T cell epitopes of myeloperoxidase were predicted and docked to the HLA molecules. We found DQA1∗0302 (odds ratio 2.34 (95% confidence interval 1.75-3.14)) and DQB1∗0303 (odds ratio 1.89 (1.45-2.48)) were risk alleles for myeloperoxidase-AAV. They are in overt linkage disequilibrium (r2 0.69) and the haplotype DQA1∗0302-DQB1∗0303 presents a significant risk (haplotype score 6.39) as well. Aspartate160 on the DQ α chain (odds ratio 2.06 (1.60-2.67)), encoded by DQA1∗0302, and isoleucine185 on the DQ β chain (odds ratio 1.73 (1.38-2.18)), encoded by DQB1∗0303, both located in the α2β2 domains, conferred significant risk for myeloperoxidase-AAV. Homologous modeling showed that DQα∗160D may confer susceptibility to myeloperoxidase-AAV by altering dimerization of the HLA molecules. Thus, more attention should be paid to the roles of amino acids in the α2β2 domains in addition to the α1β1 binding groove of HLA class II molecules.
Collapse
Affiliation(s)
- Huai-Yu Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Zhao Cui
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.
| | | | | | - Su-Fang Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
| | - Nan Chen
- Department of Nephrology, Institute of Nephrology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China; Institute of Nephrology, Peking University, Beijing, China; Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW This review will cover what is known regarding exosomes and allergy, and furthermore discuss novel mechanism of exosome-mediated immune modulation and metabolic regulation via the transfer of mitochondria. RECENT FINDINGS Exosomes are nano-sized extracellular vesicles (EVs) derived from the endosome that play a direct role in governing physiological and pathological conditions by transferring bioactive cargo such as proteins, enzymes, nucleic acids (miRNA, mRNA, DNA), and metabolites. Recent evidence suggest that exosomes may signal in autocrine but, most importantly, in paracrine and endocrine manner, being taken up by neighboring cells or carried to distant sites. Exosomes also mediate immunogenic responses, such as antigen presentation and inflammation. In asthma and allergy, exosomes facilitate cross-talk between immune and epithelial cells, and drive site-specific inflammation through the generation of pro-inflammatory mediators like leukotrienes. Recent studies suggest that myeloid cell-generated exosomes transfer mitochondria to lymphocytes. Exosomes are nano-sized mediators of the immune system which can modulate responses through antigen presentation, and the transfer of pro- and anti-inflammatory mediators. In addition to conventional mechanisms of immune modulation, exosomes may act as a novel courier of functional mitochondria that is capable of modulating the recipient cells bioenergetics, resulting in altered cellular responses. The transfer of mitochondria and modulation of bioenergetics may result in immune activation or dampening depending on the context.
Collapse
Affiliation(s)
- K P Hough
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, 1900 University Boulevard, THT-433, Birmingham, AL, 35294, USA
| | - J S Deshane
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, 1900 University Boulevard, THT-433, Birmingham, AL, 35294, USA.
| |
Collapse
|
8
|
Mikecz K, Glant TT, Markovics A, Rosenthal KS, Kurko J, Carambula RE, Cress S, Steiner HL, Zimmerman DH. An epitope-specific DerG-PG70 LEAPS vaccine modulates T cell responses and suppresses arthritis progression in two related murine models of rheumatoid arthritis. Vaccine 2017; 35:4048-4056. [PMID: 28583308 DOI: 10.1016/j.vaccine.2017.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/13/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune joint disease maintained by aberrant immune responses involving CD4+ T helper (Th)1 and Th17 cells. In this study, we tested the therapeutic efficacy of Ligand Epitope Antigen Presentation System (LEAPS™) vaccines in two Th1 cell-driven mouse models of RA, cartilage proteoglycan (PG)-induced arthritis (PGIA) and PG G1-domain-induced arthritis (GIA). The immunodominant PG peptide PG70 was attached to a DerG or J immune cell binding peptide, and the DerG-PG70 and J-PG70 LEAPS vaccines were administered to the mice after the onset of PGIA or GIA symptoms. As indicated by significant decreases in visual and histopathological scores of arthritis, the DerG-PG70 vaccine inhibited disease progression in both PGIA and GIA, while the J-PG70 vaccine was ineffective. Splenic CD4+ cells from DerG-PG70-treated mice were diminished in Th1 and Th17 populations but enriched in Th2 and regulatory T (Treg) cells. In vitro spleen cell-secreted and serum cytokines from DerG-PG70-treated mice demonstrated a shift from a pro-inflammatory to an anti-inflammatory/regulatory profile. DerG-PG70 peptide tetramers preferentially bound to CD4+ T-cells of GIA spleen cells. We conclude that the DerG-PG70 vaccine (now designated CEL-4000) exerts its therapeutic effect by interacting with CD4+ cells, which results in an antigen-specific down-modulation of pathogenic T-cell responses in both the PGIA and GIA models of RA. Future studies will need to determine the potential of LEAPS vaccination to provide disease suppression in patients with RA.
Collapse
Affiliation(s)
- Katalin Mikecz
- Rush University Medical Center, Department of Orthopedic Surgery, 1735 W. Harrison St., Cohn Research Building, Chicago, IL 60612, United States.
| | - Tibor T Glant
- Rush University Medical Center, Department of Orthopedic Surgery, 1735 W. Harrison St., Cohn Research Building, Chicago, IL 60612, United States.
| | - Adrienn Markovics
- Rush University Medical Center, Department of Orthopedic Surgery, 1735 W. Harrison St., Cohn Research Building, Chicago, IL 60612, United States.
| | - Kenneth S Rosenthal
- Roseman University of Health Sciences College of Medicine, 10530 Discovery Dr., Las Vegas, NV 89135, United States.
| | - Julia Kurko
- Rush University Medical Center, Department of Orthopedic Surgery, 1735 W. Harrison St., Cohn Research Building, Chicago, IL 60612, United States.
| | - Roy E Carambula
- CEL-SCI Corporation, 8229 Boone Blvd., Suite 802, Vienna, VA 22182, United States.
| | - Steve Cress
- CEL-SCI Corporation, 8229 Boone Blvd., Suite 802, Vienna, VA 22182, United States.
| | - Harold L Steiner
- CEL-SCI Corporation, 8229 Boone Blvd., Suite 802, Vienna, VA 22182, United States.
| | - Daniel H Zimmerman
- CEL-SCI Corporation, 8229 Boone Blvd., Suite 802, Vienna, VA 22182, United States.
| |
Collapse
|
9
|
Glassman CR, Parrish HL, Deshpande NR, Kuhns MS. The CD4 and CD3δε Cytosolic Juxtamembrane Regions Are Proximal within a Compact TCR-CD3-pMHC-CD4 Macrocomplex. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4713-22. [PMID: 27183595 PMCID: PMC4875830 DOI: 10.4049/jimmunol.1502110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/29/2016] [Indexed: 12/15/2022]
Abstract
TCRs relay information about peptides embedded within MHC molecules (pMHC) to the ITAMs of the associated CD3γε, CD3δε, and CD3ζζ signaling modules. CD4 then recruits the Src kinase p56(Lck) (Lck) to the TCR-CD3 complex to phosphorylate the ITAMs, initiate intracellular signaling, and drive CD4(+) T cell fate decisions. Whereas the six ITAMs of CD3ζζ are key determinants of T cell development, activation, and the execution of effector functions, multiple models predict that CD4 recruits Lck proximal to the four ITAMs of the CD3 heterodimers. We tested these models by placing FRET probes at the cytosolic juxtamembrane regions of CD4 and the CD3 subunits to evaluate their relationship upon pMHC engagement in mouse cell lines. The data are consistent with a compact assembly in which CD4 is proximal to CD3δε, CD3ζζ resides behind the TCR, and CD3γε is offset from CD3δε. These results advance our understanding of the architecture of the TCR-CD3-pMHC-CD4 macrocomplex and point to regions of high CD4-Lck + ITAM concentrations therein. The findings thus have implications for TCR signaling, as phosphorylation of the CD3 ITAMs by CD4-associated Lck is important for CD4(+) T cell fate decisions.
Collapse
Affiliation(s)
- Caleb R Glassman
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724
| | - Heather L Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724
| | - Neha R Deshpande
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724; The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ 85724; and
| | - Michael S Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ 85724; The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, AZ 85724; and The BIO5 Institute, The University of Arizona College of Medicine, Tucson, AZ 85724
| |
Collapse
|
10
|
Jönsson P, Southcombe JH, Santos AM, Huo J, Fernandes RA, McColl J, Lever M, Evans EJ, Hudson A, Chang VT, Hanke T, Godkin A, Dunne PD, Horrocks MH, Palayret M, Screaton GR, Petersen J, Rossjohn J, Fugger L, Dushek O, Xu XN, Davis SJ, Klenerman D. Remarkably low affinity of CD4/peptide-major histocompatibility complex class II protein interactions. Proc Natl Acad Sci U S A 2016; 113:5682-7. [PMID: 27114505 PMCID: PMC4878507 DOI: 10.1073/pnas.1513918113] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The αβ T-cell coreceptor CD4 enhances immune responses more than 1 million-fold in some assays, and yet the affinity of CD4 for its ligand, peptide-major histocompatibility class II (pMHC II) on antigen-presenting cells, is so weak that it was previously unquantifiable. Here, we report that a soluble form of CD4 failed to bind detectably to pMHC II in surface plasmon resonance-based assays, establishing a new upper limit for the solution affinity at 2.5 mM. However, when presented multivalently on magnetic beads, soluble CD4 bound pMHC II-expressing B cells, confirming that it is active and allowing mapping of the native coreceptor binding site on pMHC II. Whereas binding was undetectable in solution, the affinity of the CD4/pMHC II interaction could be measured in 2D using CD4- and adhesion molecule-functionalized, supported lipid bilayers, yielding a 2D Kd of ∼5,000 molecules/μm(2) This value is two to three orders of magnitude higher than previously measured 2D Kd values for interacting leukocyte surface proteins. Calculations indicated, however, that CD4/pMHC II binding would increase rates of T-cell receptor (TCR) complex phosphorylation by threefold via the recruitment of Lck, with only a small, 2-20% increase in the effective affinity of the TCR for pMHC II. The affinity of CD4/pMHC II therefore seems to be set at a value that increases T-cell sensitivity by enhancing phosphorylation, without compromising ligand discrimination.
Collapse
Affiliation(s)
- Peter Jönsson
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom; Department of Chemistry, Lund University, SE-22100 Lund, Sweden
| | - Jennifer H Southcombe
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom; Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Ana Mafalda Santos
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Jiandong Huo
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Ricardo A Fernandes
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - James McColl
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Melissa Lever
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Edward J Evans
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Alexander Hudson
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Veronica T Chang
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Tomáš Hanke
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Andrew Godkin
- Department of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom
| | - Paul D Dunne
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Mathew H Horrocks
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Matthieu Palayret
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Gavin R Screaton
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Jamie Rossjohn
- Department of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XN, United Kingdom; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Lars Fugger
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom; Nuffield Department of Clinical Neurosciences, Division of Clinical Neurology, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Xiao-Ning Xu
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom;
| | - Simon J Davis
- MRC Human Immunology Unit, University of Oxford, Oxford OX3 9DS, United Kingdom; Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom;
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom;
| |
Collapse
|
11
|
Owen GR, Channell JA, Forsyth VT, Haertlein M, Mitchell EP, Capovilla A, Papathanasopoulos M, Cerutti NM. Human CD4 Metastability Is a Function of the Allosteric Disulfide Bond in Domain 2. Biochemistry 2016; 55:2227-37. [DOI: 10.1021/acs.biochem.6b00154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Gavin R. Owen
- HIV
Pathogenesis Research Unit, Department of Molecular Medicine and Haematology,
Faculty of Health Sciences, University of the Witwatersrand, 7
York Road, Parktown, 2193, Johannesburg, South Africa
| | - Jennifer A. Channell
- Faculty
of Natural Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
- Life
Sciences Group, Institut Laue-Langevin, 71 Avenue des Martyrs, 38042, Grenoble, France
- European Synchrotron Radiation Facility, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - V. Trevor Forsyth
- Faculty
of Natural Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
- Life
Sciences Group, Institut Laue-Langevin, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Michael Haertlein
- Life
Sciences Group, Institut Laue-Langevin, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Edward P. Mitchell
- Faculty
of Natural Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
- European Synchrotron Radiation Facility, 71 Avenue des Martyrs, 38042, Grenoble, France
| | - Alexio Capovilla
- HIV
Pathogenesis Research Unit, Department of Molecular Medicine and Haematology,
Faculty of Health Sciences, University of the Witwatersrand, 7
York Road, Parktown, 2193, Johannesburg, South Africa
| | - Maria Papathanasopoulos
- HIV
Pathogenesis Research Unit, Department of Molecular Medicine and Haematology,
Faculty of Health Sciences, University of the Witwatersrand, 7
York Road, Parktown, 2193, Johannesburg, South Africa
| | - Nichole M. Cerutti
- HIV
Pathogenesis Research Unit, Department of Molecular Medicine and Haematology,
Faculty of Health Sciences, University of the Witwatersrand, 7
York Road, Parktown, 2193, Johannesburg, South Africa
| |
Collapse
|
12
|
Parrish HL, Glassman CR, Keenen MM, Deshpande NR, Bronnimann MP, Kuhns MS. A Transmembrane Domain GGxxG Motif in CD4 Contributes to Its Lck-Independent Function but Does Not Mediate CD4 Dimerization. PLoS One 2015; 10:e0132333. [PMID: 26147390 PMCID: PMC4493003 DOI: 10.1371/journal.pone.0132333] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/14/2015] [Indexed: 12/24/2022] Open
Abstract
CD4 interactions with class II major histocompatibility complex (MHC) molecules are essential for CD4+ T cell development, activation, and effector functions. While its association with p56lck (Lck), a Src kinase, is important for these functions CD4 also has an Lck-independent role in TCR signaling that is incompletely understood. Here, we identify a conserved GGxxG motif in the CD4 transmembrane domain that is related to the previously described GxxxG motifs of other proteins and predicted to form a flat glycine patch in a transmembrane helix. In other proteins, these patches have been reported to mediate dimerization of transmembrane domains. Here we show that introducing bulky side-chains into this patch (GGxxG to GVxxL) impairs the Lck-independent role of CD4 in T cell activation upon TCR engagement of agonist and weak agonist stimulation. However, using Forster’s Resonance Energy Transfer (FRET), we saw no evidence that these mutations decreased CD4 dimerization either in the unliganded state or upon engagement of pMHC concomitantly with the TCR. This suggests that the CD4 transmembrane domain is either mediating interactions with an unidentified partner, or mediating some other function such as membrane domain localization that is important for its role in T cell activation.
Collapse
Affiliation(s)
- Heather L. Parrish
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Caleb R. Glassman
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Madeline M. Keenen
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Neha R. Deshpande
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
- The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Matthew P. Bronnimann
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Michael S. Kuhns
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
- The Arizona Center on Aging, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
- The BIO-5 Institute, The University of Arizona College of Medicine, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
13
|
Rosenthal KS, Mikecz K, Steiner HL, Glant TT, Finnegan A, Carambula RE, Zimmerman DH. Rheumatoid arthritis vaccine therapies: perspectives and lessons from therapeutic ligand epitope antigen presentation system vaccines for models of rheumatoid arthritis. Expert Rev Vaccines 2015; 14:891-908. [PMID: 25787143 DOI: 10.1586/14760584.2015.1026330] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The current status of therapeutic vaccines for autoimmune diseases is reviewed with rheumatoid arthritis as the focus. Therapeutic vaccines for autoimmune diseases must regulate or subdue responses to common self-antigens. Ideally, such a vaccine would initiate an antigen-specific modulation of the T-cell immune response that drives the inflammatory disease. Appropriate animal models and types of T helper cells and signature cytokine responses that drive autoimmune disease are also discussed. Interpretation of these animal models must be done cautiously because the means of initiation, autoantigens, and even the signature cytokine and T helper cell (Th1 or Th17) responses that are involved in the disease may differ significantly from those in humans. We describe ligand epitope antigen presentation system vaccine modulation of T-cell autoimmune responses as a strategy for the design of therapeutic vaccines for rheumatoid arthritis, which may also be effective in other autoimmune conditions.
Collapse
|
14
|
Allorecognition of HLA-DP by CD4+ T cells is affected by polymorphism in its alpha chain. Mol Immunol 2014; 59:19-29. [DOI: 10.1016/j.molimm.2013.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 11/18/2013] [Accepted: 12/27/2013] [Indexed: 11/21/2022]
|
15
|
Cerutti N, Killick M, Jugnarain V, Papathanasopoulos M, Capovilla A. Disulfide reduction in CD4 domain 1 or 2 is essential for interaction with HIV glycoprotein 120 (gp120), which impairs thioredoxin-driven CD4 dimerization. J Biol Chem 2014; 289:10455-10465. [PMID: 24550395 PMCID: PMC4036167 DOI: 10.1074/jbc.m113.539353] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/12/2014] [Indexed: 11/06/2022] Open
Abstract
Human CD4 is a membrane-bound glycoprotein expressed on the surface of certain leukocytes, where it plays a key role in the activation of immunostimulatory T cells and acts as the primary receptor for human immunodeficiency virus (HIV) glycoprotein (gp120). Although growing evidence suggests that redox exchange reactions involving CD4 disulfides, potentially catalyzed by cell surface-secreted oxidoreductases such as thioredoxin (Trx) and protein disulfide isomerase, play an essential role in regulating the activity of CD4, their mechanism(s) and biological utility remain incompletely understood. To gain more insights in this regard, we generated a panel of recombinant 2-domain CD4 proteins (2dCD4), including wild-type and Cys/Ala variants, and used these to show that while protein disulfide isomerase has little capacity for 2dCD4 reduction, Trx reduces 2dCD4 highly efficiently, catalyzing the formation of conformationally distinct monomeric 2dCD4 isomers, and a stable, disulfide-linked 2dCD4 dimer. Moreover, we show that HIV gp120 is incapable of binding a fully oxidized, monomeric 2dCD4 in which both domain 1 and 2 disulfides are intact, but binds robustly to reduced counterparts that are the ostensible products of Trx-mediated isomerization. Finally, we demonstrate that Trx-driven dimerization of CD4, a process believed to be critical for the establishment of functional MHCII-TCR-CD4 antigen presentation complexes, is impaired when CD4 is bound to gp120. These observations reinforce the importance of cell surface redox activity for HIV entry and posit the intriguing possibility that one of the many pathogenic effects of HIV may be related to gp120-mediated inhibition of oxidoreductive CD4 isomerization.
Collapse
Affiliation(s)
- Nichole Cerutti
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of Witwatersrand Medical School, 7 York Road Parktown, 2193 Johannesburg, South Africa
| | - Mark Killick
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of Witwatersrand Medical School, 7 York Road Parktown, 2193 Johannesburg, South Africa
| | - Vinesh Jugnarain
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of Witwatersrand Medical School, 7 York Road Parktown, 2193 Johannesburg, South Africa
| | - Maria Papathanasopoulos
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of Witwatersrand Medical School, 7 York Road Parktown, 2193 Johannesburg, South Africa
| | - Alexio Capovilla
- HIV Pathogenesis Research Laboratory, Department of Molecular Medicine and Haematology, University of Witwatersrand Medical School, 7 York Road Parktown, 2193 Johannesburg, South Africa.
| |
Collapse
|
16
|
Kim JW, Bilusic M, Heery CJ, Madan RA. Therapeutic cancer vaccines in prostate cancer: the quest for intermediate markers of response. Cancers (Basel) 2012; 4:1229-46. [PMID: 24213505 PMCID: PMC3712729 DOI: 10.3390/cancers4041229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/09/2012] [Accepted: 11/14/2012] [Indexed: 11/17/2022] Open
Abstract
Despite recent advances in cancer immunotherapy, no prospectively validated intermediate biomarkers exist to predict response. These biomarkers are highly desirable given modern immunotherapy's paradoxical pattern of clinical benefit; that is, improvement in overall survival without short-term change in progression. Immunotherapy clinical trials have evaluated biomarkers that may correlate with clinical outcomes. Many of them are performed on peripheral blood to evaluate the systemic response, such as tumor-targeted humoral and cellular immunity, and cytokine responses. Accumulating evidence suggests that immune infiltrates in tumors may suggest evidence for the therapy's mechanism of action, and have greater potential for providing prognostic and predictive information. In addition, a non-immunologic biomarker, such as tumor growth kinetics, may explain this paradoxical pattern of clinical benefit, and predict survival in patients treated with an immunotherapy. Prospective assessment and validation of these and other intermediate markers would be required to better understand their potential clinical role.
Collapse
Affiliation(s)
- Joseph W Kim
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
17
|
Becker C, Bopp T, Jonuleit H. Boosting regulatory T cell function by CD4 stimulation enters the clinic. Front Immunol 2012; 3:164. [PMID: 22719741 PMCID: PMC3376463 DOI: 10.3389/fimmu.2012.00164] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/31/2012] [Indexed: 01/14/2023] Open
Abstract
Understanding tolerance mechanisms at the cellular and molecular level holds the promise to establish novel immune intervention therapies in patients with allergy or autoimmunity and to prevent transplant rejection. Administration of mAb against the CD4 molecule has been found to be exceptionally well suited for intentional tolerance induction in rodent and non-human primate models as well as in humanized mouse models. Recent evidence demonstrated that regulatory T cells (Treg) are directly activated by non-depleting CD4 ligands and suggests Treg activation as a central mechanism in anti-CD4-mediated tolerance induction. This review summarizes the current knowledge on the role of Treg in peripheral tolerance, addresses the putative mechanisms of Treg-mediated suppression and discusses the clinical potential of harnessing Treg suppressive activity through CD4 stimulation.
Collapse
Affiliation(s)
- Christian Becker
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | | | |
Collapse
|
18
|
Wan QH, Zhang P, Ni XW, Wu HL, Chen YY, Kuang YY, Ge YF, Fang SG. A novel HURRAH protocol reveals high numbers of monomorphic MHC class II loci and two asymmetric multi-locus haplotypes in the Père David's deer. PLoS One 2011; 6:e14518. [PMID: 21267075 PMCID: PMC3022581 DOI: 10.1371/journal.pone.0014518] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 12/17/2010] [Indexed: 11/22/2022] Open
Abstract
The Père David's deer is a highly inbred, but recovered, species, making it interesting to consider their adaptive molecular evolution from an immunological perspective. Prior to this study, genomic sequencing was the only method for isolating all functional MHC genes within a certain species. Here, we report a novel protocol for isolating MHC class II loci from a species, and its use to investigate the adaptive evolution of this endangered deer at the level of multi-locus haplotypes. This protocol was designated “HURRAH” based on its various steps and used to estimate the total number of MHC class II loci. We confirmed the validity of this novel protocol in the giant panda and then used it to examine the Père David's deer. Our results revealed that the Père David's deer possesses nine MHC class II loci and therefore has more functional MHC class II loci than the eight genome-sequenced mammals for which full MHC data are currently available. This could potentially account at least in part for the strong survival ability of this species in the face of severe bottlenecking. The results from the HURRAH protocol also revealed that: (1) All of the identified MHC class II loci were monomorphic at their antigen-binding regions, although DRA was dimorphic at its cytoplasmic tail; and (2) these genes constituted two asymmetric functional MHC class II multi-locus haplotypes: DRA1*01 ∼ DRB1 ∼ DRB3 ∼ DQA1 ∼ DQB2 (H1) and DRA1*02 ∼ DRB2 ∼ DRB4 ∼ DQA2 ∼ DQB1 (H2). The latter finding indicates that the current members of the deer species have lost the powerful ancestral MHC class II haplotypes of nine or more loci, and have instead fixed two relatively weak haplotypes containing five genes. As a result, the Père David's deer are currently at risk for increased susceptibility to infectious pathogens.
Collapse
Affiliation(s)
- Qiu-Hong Wan
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Pei Zhang
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao-Wei Ni
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Hai-Long Wu
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
- College of Life Science, Anhui Normal University, Wuhu, People's Republic of China
| | - Yi-Yan Chen
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Ye-Ye Kuang
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Yun-Fa Ge
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
| | - Sheng-Guo Fang
- The Key Laboratory of Conservation Biology for Endangered Wildlife of the Ministry of Education, College of Life Sciences, State Conservation Center for Gene Resources of Endangered Wildlife, Zhejiang University, Hangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
19
|
Fournier M, Peyrou M, Bourgoin L, Maeder C, Tchou I, Foti M. CD4 dimerization requires two cysteines in the cytoplasmic domain of the molecule and occurs in microdomains distinct from lipid rafts. Mol Immunol 2010; 47:2594-603. [DOI: 10.1016/j.molimm.2010.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 05/06/2010] [Accepted: 06/21/2010] [Indexed: 01/27/2023]
|
20
|
Yasukochi Y, Kurosaki T, Yoneda M, Koike H. Identification of the expressed MHC class II DQB gene of the Asiatic black bear, Ursus thibetanus, in Japan. Genes Genet Syst 2010; 85:147-55. [PMID: 20558901 DOI: 10.1266/ggs.85.147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Genetic diversity estimation of the major histocompatibility complex (MHC) gene may be an important tool in the assessment of immune response ability against infectious disease. We were able to identify a near full-length expressed DQB sequence by RACE-PCR method from the Asiatic black bear, Ursus thibetanus in Japan. This is the first such full length expression in the Ursidae. The bear had at least one functional DQB locus. In phylogenetic tree analysis its DQB amino acid sequence formed a monophyletic group with DQB sequences from members of the order Carnivora and had a 90% nucleotide sequence similarity with the DQB allele of the California sea lion, Zalophus californianus. We compared the DQB amino acid composition of U. thibetanus with those of several other mammalian species including Homo sapiens. Amino acid residues known to be functionally important for human MHC genes, tended to be also conserved among other mammalian species while PBRs in the beta1 domain were heterogeneous among mammalian species. The DQB sequence obtained from the bear had not only no putative frameshifts or deletions but also no abnormal amino acid mutations such as had been observed in human DQB molecules. This suggests that the bear DQB sequence was an apparently functional DQB allele. As a preliminary study, we sequenced the exon 2 region of DQB alleles from genomic DNA, and succeeded to amplify the exon 2 of DQB loci. Our study will provide useful information for conservation genetics of the U. thibetanus as well as more generally regarding the mammalian MHC region.
Collapse
Affiliation(s)
- Yoshiki Yasukochi
- Department of Biosystems Science, the Graduate University for Advanced Studies, Hayama.
| | | | | | | |
Collapse
|
21
|
Vignali DAA. CD4 on the road to coreceptor status. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:5933-4. [PMID: 20483776 PMCID: PMC3142941 DOI: 10.4049/jimmunol.1090037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA.
| |
Collapse
|
22
|
MHC class II exacerbates demyelination in vivo independently of T cells. J Neuroimmunol 2009; 203:23-32. [PMID: 18805594 DOI: 10.1016/j.jneuroim.2008.06.034] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 06/14/2008] [Accepted: 06/16/2008] [Indexed: 11/17/2022]
Abstract
We have shown previously the importance of MHC class II for central nervous system remyelination; however, the function of MHC class II during cuprizone-induced demyelination has not been examined. Here, we show that I-A(beta)-/- mice exhibit significantly reduced inflammation and demyelination. RAG-1(1/1) mice are indistinguishable from controls, indicating T cells may not play a role. The role of MHC class II depends on an intact cytoplasmic tail that leads to the production of IL-1beta, TNF-alpha, and nitric oxide, and oligodendrocyte apoptosis. Thus, the function of MHC class II cytoplasmic tail appears to increase microglial proliferation and activation that exacerbates demyelination.
Collapse
|
23
|
J2 prolongs the corneal allograft survival through inhibition of the CD4+ T cell-mediated response in vivo. Transpl Immunol 2007; 18:130-7. [PMID: 18005857 DOI: 10.1016/j.trim.2007.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Accepted: 05/22/2007] [Indexed: 11/22/2022]
Abstract
In our previous report, we described a novel non-peptidic organic ligand of CD4 D1, designated J2, as a potential inhibitor of CD4 D1 and thus CD4-dependent T cell responses in vitro. In this work, we further used a murine model of corneal allograft rejection to determine its in vivo immunosuppressive activities. To mimic the situation in high-risk human eyes, the recipient mice corneas were all induced by intrastromal sutures to serve as neovascularized graft beds. J2 was administrated by mouth 3 h before transplantation and thereafter on consecutive 12 days. The results showed that J2 could significantly prolong the median survival time of the corneal allografts, compared to the untreated control group. And the subsequent functional assays, including T cell phenotype analysis, delayed-type hypersensitivity (DTH) and enzyme-linked immunospot (ELISPOT) assays revealed that the immunosuppressive activity of J2 was associated with its inhibitory effects on the CD4(+) T cells and these cells-mediated responses. All these results suggest that J2 is a potential lead for the development of new immunosuppressive agents to prevent the corneal allograft rejection.
Collapse
|
24
|
Bondinas GP, Moustakas AK, Papadopoulos GK. The spectrum of HLA-DQ and HLA-DR alleles, 2006: a listing correlating sequence and structure with function. Immunogenetics 2007; 59:539-53. [PMID: 17497145 DOI: 10.1007/s00251-007-0224-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 04/03/2007] [Indexed: 10/23/2022]
Abstract
The list of alleles in the HLA-DRB, HLA-DQA, and HLA-DQB gene loci has grown enormously since the last listing in this journal 8 years ago. Crystal structure determination of several human and mouse HLA class II alleles, representative of two gene loci in each species, enables a direct comparison of ortholog and paralog loci. A new numbering system is suggested, extending earlier suggestions by [Fremont et al. in Immunity 8:305-317, (1998)], which will bring in line all the structural features of various gene loci, regardless of animal species. This system allows for structural equivalence of residues from different gene loci. The listing also highlights all amino acid residues participating in the various functions of these molecules, from antigenic peptide binding to homodimer formation, CD4 binding, membrane anchoring, and cytoplasmic signal transduction, indicative of the variety of functions of these molecules. It is remarkable that despite the enormous number of unique alleles listed thus far (DQA = 22, DQB = 54, DRA = 2, and DRB = 409), there is invariance at many specific positions in man, but slightly less so in mouse or rat, despite their much lower number of alleles at each gene locus in the latter two species. Certain key polymorphisms (from substitutions to an eight-residue insertion in the cytoplasmic tail of certain DQB alleles) that have thus far gone unnoticed are highly suggestive of differences or diversities in function and thus call for further investigation into the properties of these specific alleles. This listing is amenable to supplementation by future additions of new alleles and the highlighting of new functions to be discovered, providing thus a unifying platform of reference in all animal species for the MHC class II allelic counterparts, aiding research in the field and furthering our understanding of the functions of these molecules.
Collapse
Affiliation(s)
- George P Bondinas
- Laboratory of Biochemistry and Biophysics, Faculty of Agricultural Technology, Epirus Institute of Technology, GR47100 Arta, Greece
| | | | | |
Collapse
|
25
|
Thedrez A, de Lalla C, Allain S, Zaccagnino L, Sidobre S, Garavaglia C, Borsellino G, Dellabona P, Bonneville M, Scotet E, Casorati G. CD4 engagement by CD1d potentiates activation of CD4+ invariant NKT cells. Blood 2007; 110:251-8. [PMID: 17363727 DOI: 10.1182/blood-2007-01-066217] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The CD4 coreceptor is crucial in the activation of major histocompatibility complex (MHC) class II restricted CD4 (+) T lymphocytes by binding the same MHC class as the T-cell receptor (TCR) and by potentiating TCR-dependent signaling. CD4 is also expressed by invariant natural killer T cells (iNKT), which recognize natural and synthetic lipid antigens, such as alpha-galactosyl ceramide (alpha-GalCer), in association with the MHC class I-like CD1d molecule. Human iNKT cells can be divided into 2 major subsets depending on CD4 expression: CD4 (+) iNKT preferentially produce T-helper (Th)0/Th2 cytokines, whereas CD4(-) iNKT cells produce Th1 cytokines after antigenic activation. Cytokines produced by iNKT may have immunomodulatory roles in various physiopathologic contexts, but their mode of regulation by iNKT cells remains ill-defined. Using blocking reagents neutralizing CD4 binding, experimental systems where MHC class II molecules are absent and recombinant alpha-GalCer/CD1d complexes, we show that CD4 potentiates human iNKT cell activation by engaging CD1d molecules. These results indicate that the CD4 coreceptors may contribute to the fine tuning of iNKT cells reactivity.
Collapse
Affiliation(s)
- Aurelie Thedrez
- Institute Nationale Scientifique et Recherche Medicale, Nantes, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Moldovan MC, Sabbagh L, Breton G, Sékaly RP, Krummel MF. Triggering of T cell activation via CD4 dimers. THE JOURNAL OF IMMUNOLOGY 2006; 176:5438-45. [PMID: 16622011 DOI: 10.4049/jimmunol.176.9.5438] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The onset of activation in Th cells is triggered by localized co-engagement of TCRs and the coreceptor CD4. A CD4 crystal suggested that CD4 may form dimers in some circumstances. In this study, we use live-cell fluorescence resonance energy transfer imaging to demonstrate that CD4 dimers are present at a basal level on the cell surface and accumulate at the synapse. Mechanistically, we reveal two conditions under which dimers are highly relevant. First, CD4 dimers are more proficient in mediating prolonged cell contacts with APCs in the presence or absence of Ag. This is consistent with a model whereby the dimer functions to increase T-APC avidity. Second, we show that dimer mutations result in an increased level of an inactive lckTyr(505) bound to the CD4 molecule relative to dimer-competent CD4. We also find a consistent defect in signaling onset in these cells. This supports a role for CD4 dimerization in maintaining active signaling machinery. We suggest that modulation of the dimer/monomer ratio may permit tuning of activation thresholds during initial engagement.
Collapse
|
27
|
Maekawa A, Schmidt B, Fazekas de St Groth B, Sanejouand YH, Hogg PJ. Evidence for a Domain-Swapped CD4 Dimer as the Coreceptor for Binding to Class II MHC. THE JOURNAL OF IMMUNOLOGY 2006; 176:6873-8. [PMID: 16709847 DOI: 10.4049/jimmunol.176.11.6873] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CD4 is a coreceptor for binding of T cells to APC and the primary receptor for HIV. The disulfide bond in the second extracellular domain (D2) of CD4 is reduced on the cell surface, which leads to formation of disulfide-linked homodimers. A large conformational change must take place in D2 to allow for formation of the disulfide-linked dimer. Domain swapping of D2 is the most likely candidate for the conformational change leading to formation of two disulfide-bonds between Cys130 in one monomer and Cys159 in the other one. Mild reduction of the extracellular part of CD4 resulted in formation of disulfide-linked dimers, which supports the domain-swapped model. The functional significance of dimer formation for coreceptor function was tested using cells expressing wild-type or disulfide-bond mutant CD4. Eliminating the D2 disulfide bond markedly impaired CD4's coreceptor function. Modeling of the complex of the TCR and domain-swapped CD4 dimer bound to class II MHC and Ag supports the domain-swapped dimer as the immune coreceptor. The known involvement of D4 residues Lys318 and Gln344 in dimer formation is also accommodated by this model. These findings imply that disulfide-linked dimeric CD4 is the preferred coreceptor for binding to APC.
Collapse
Affiliation(s)
- Akiko Maekawa
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | | | | | | | | |
Collapse
|
28
|
van der Veken LT, Hagedoorn RS, van Loenen MM, Willemze R, Falkenburg JHF, Heemskerk MHM. Alphabeta T-cell receptor engineered gammadelta T cells mediate effective antileukemic reactivity. Cancer Res 2006; 66:3331-7. [PMID: 16540688 DOI: 10.1158/0008-5472.can-05-4190] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Retroviral transfer of T-cell receptors (TCR) to peripheral blood-derived T cells generates large numbers of T cells with the same antigen specificity, potentially useful for adoptive immunotherapy. One drawback of this procedure is the formation of mixed TCR dimers with unknown specificities due to pairing of endogenous and introduced TCR chains. We investigated whether gammadelta T cells can be an alternative effector population for TCR gene transfer because the gammadeltaTCR is not able to form dimers with the alphabetaTCR. Peripheral blood-derived gammadelta T cells were transduced with human leukocyte antigen (HLA) class I- or HLA class II-restricted minor histocompatibility antigen (mHag) or virus-specific TCRs. Because most gammadelta T cells do not express CD4 and CD8, we subsequently transferred these coreceptors. The TCR-transduced gammadelta T cells exerted high levels of antigen-specific cytotoxicity and produced IFN-gamma and IL-4, particularly in the presence of the relevant coreceptor. gammadelta T cells transferred with a TCR specific for the hematopoiesis-specific mHag HA-2 in combination with CD8 displayed high antileukemic reactivity against HA-2-expressing leukemic cells. These data show that transfer of alphabetaTCRs to gammadelta T cells generated potent effector cells for immunotherapy of leukemia, without the expression of potentially hazardous mixed TCR dimers.
Collapse
MESH Headings
- CD3 Complex/biosynthesis
- CD3 Complex/genetics
- CD3 Complex/immunology
- Gene Transfer Techniques
- Genetic Engineering
- HLA-A2 Antigen/immunology
- HLA-B7 Antigen/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia/immunology
- Leukemia/therapy
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Retroviridae/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Lars T van der Veken
- Laboratory of Experimental Hematology, Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | |
Collapse
|
29
|
Marracci GH, Marquardt WE, Strehlow A, McKeon GP, Gross J, Buck DC, Kozell LB, Bourdette DN. Lipoic acid downmodulates CD4 from human T lymphocytes by dissociation of p56(Lck). Biochem Biophys Res Commun 2006; 344:963-71. [PMID: 16631599 DOI: 10.1016/j.bbrc.2006.03.172] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 03/22/2006] [Indexed: 01/03/2023]
Abstract
Lipoic acid is an antioxidant that suppresses and treats a model of multiple sclerosis, experimental autoimmune encephalomyelitis. We now demonstrate that treatment of human PBMC and T cell lines with LA downmodulated CD4 expression in a concentration-dependent manner. LA treatment of Con A stimulated PBMC specifically removed CD4 from the T-cell surface, but not CD3. Epitope masking by LA was excluded by using monoclonal antibodies targeting different domains of CD4. Incubation on ice inhibited CD4 removal following LA treatment, suggesting that endocytosis was involved in its downmodulation. LA is in a unique category of compounds that induce CD4 downmodulation by various mechanisms (e.g., gangliosides). We hypothesized that LA might induce dissociation of p56(Lck) from CD4, thus leading to its downmodulation. Immunoblot analyses demonstrated reduced co-precipitation of p56(Lck) from Jurkat T-cells following LA treatment and precipitation of CD4. This unique immunomodulatory effect of LA warrants further investigation.
Collapse
Affiliation(s)
- Gail H Marracci
- Portland Veterans Affairs Medical Center, R&D-65, 3710 SW US Veterans Hospital Road, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Hayball JD, Lake RA. The immune function of MHC class II molecules mutated in the putative superdimer interface. Mol Cell Biochem 2005; 273:1-9. [PMID: 16013435 DOI: 10.1007/s11010-005-5281-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Analysis of the crystal structure of human class II (HLA-DR1) molecules suggests that the alphabeta heterodimer may be further ordered as a dimer of heterodimers (superdimer), leading to the hypothesis that T cell receptor dimerisation is a mechanism for initiating signaling events preceding T cell activation. The interface between pairs of molecules is stabilised by both salt bridges, polar and hydrophobic interactions. The residues that form the superdimer interface occur in three areas distinct from the antigen-binding groove. They can be defined as follows: region 1, beta-beta contacts in the helix of the beta1 domain; region 2, alpha-alpha contacts near the alpha 1/alpha2 domain junction and region 3; alpha-beta contacts in the alpha2/beta2 domains adjacent to the plasma membrane. To determine whether salt bridges and polar interactions formed within these regions are involved in the immune function of the murine MHC class II molecule, I-A(b), appropriate residues in both the alpha and beta chain were identified and mutated to uncharged alanine. Cell lines transfected with different combinations of mutated alpha and beta chains were generated and tested for MHC class II expression, peptide binding capabilities, and ability to present antigenic peptide to an OVA-specific T cell hybridoma. With the exception of two residues in region 2, the substitutions tested did not modulate MHC class II expression, or peptide binding function. When tested for ability to present peptide to an antigen-specific T cell hybridoma, with the exception of mutations in region 2, the substitutions did not appear to abrogate the ability of I-A(b) to stimulate the T cells. These results suggest that mutation of residues in region 2 of the putative superdimer interface have a gross effect on the ability of I-A(b) to be expressed on the cell surface. However, abrogation of salt bridges in region 1 and 3 do not influence I-A(b) cell surface expression, peptide binding or ability to stimulate antigen-specific T cells.
Collapse
Affiliation(s)
- John D Hayball
- School of Pharmacy and Medical Sciences, University of South Australia, North Tce, Adelaide, South Australia, Australia.
| | | |
Collapse
|
31
|
Huan JY, Meza-Romero R, Mooney JL, Chou YK, Edwards DM, Rich C, Link JM, Vandenbark AA, Bourdette DN, Bächinger HP, Burrows GG. Rationally designed mutations convert complexes of human recombinant T cell receptor ligands into monomers that retain biological activity. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2005; 80:2-12. [PMID: 22973070 PMCID: PMC3438139 DOI: 10.1002/jctb.1086] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Single-chain human recombinant T cell receptor ligands derived from the peptide binding/TCR recognition domain of human HLA-DR2b (DRA*0101/DRB1*1501) produced in Escherichia coli with and without amino-terminal extensions containing antigenic peptides have been described previously. While molecules with the native sequence retained biological activity, they formed higher order aggregates in solution. In this study, we used site-directed mutagenesis to modify the β-sheet platform of the DR2-derived RTLs, obtaining two variants that were monomeric in solution by replacing hydrophobic residues with polar (serine) or charged (aspartic acid) residues. Size exclusion chromatography and dynamic light scattering demonstrated that the modified RTLs were monomeric in solution, and structural characterization using circular dichroism demonstrated the highly ordered secondary structure of the RTLs. Peptide binding to the `empty' RTLs was quantified using biotinylated peptides, and functional studies showed that the modified RTLs containing covalently tethered peptides were able to inhibit antigen-specific T cell proliferation in vitro, as well as suppress experimental autoimmune encephalomyelitis in vivo. These studies demonstrated that RTLs encoding the Ag-binding/TCR recognition domain of MHC class II molecules are innately very robust structures, capable of retaining potent biological activity separate from the Ig-fold domains of the progenitor class II structure, with prevention of aggregation accomplished by modification of an exposed surface that was buried in the progenitor structure.
Collapse
Affiliation(s)
- Jianya Y Huan
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Roberto Meza-Romero
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeffery L Mooney
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Yuan K Chou
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - David M Edwards
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Cathleen Rich
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, OR 97207, USA
| | - Jason M Link
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, OR 97207, USA
| | - Arthur A Vandenbark
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, OR 97207, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Dennis N Bourdette
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | | | - Gregory G Burrows
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
32
|
Ettinger RA, Moustakas AK, Lobaton SD. Open reading frame sequencing and structure-based alignment of polypeptides encoded by RT1-Bb, RT1-Ba, RT1-Db, and RT1-Da alleles. Immunogenetics 2004; 56:585-96. [PMID: 15517241 DOI: 10.1007/s00251-004-0725-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Revised: 09/13/2004] [Indexed: 10/26/2022]
Abstract
MHC class II genes are major genetic components in rats developing autoimmunity. The majority of rat MHC class II sequencing has focused on exon 2, which forms the first external domain. Sequence of the complete open reading frame for rat MHC class II haplotypes and structure-based alignment is lacking. Herein, the complete open reading frame for RT1-Bbeta, RT1-Balpha, RT1-Dbeta, and RT1-Dalpha was sequenced from ten different rat strains, covering eight serological haplotypes, namely a, b, c, d, k, l, n, and u. Each serological haplotype was unique at the nucleotide level of the sequenced RT1-B/D region. Within individual genes, the number of alleles identified was seven, seven, six, and three and the degree of amino-acid polymorphism between allotypes for each gene was 22%, 16%, 19%, and 0.4% for RT1-Bbeta, RT1-Balpha, RT1-Dbeta, and RT1-Dalpha, respectively. The extent and distribution of amino-acid polymorphism was comparable with mouse and human MHC class II. Structure-based alignment identified the beta65-66 deletion, the beta84a insertion, the alpha9a insertion, and the alpha1a-1c insertion in RT1-B previously described for H2-A. Rat allele-specific deletions were found at RT1-Balpha76 and RT1-Dbeta90-92. The mature RT1-Dbeta polypeptide was one amino acid longer than HLA-DRB1 due to the position of the predicted signal peptide cleavage site. These data are important to a comprehensive understanding of MHC class II structure-function and for mechanistic studies of rat models of autoimmunity.
Collapse
Affiliation(s)
- Ruth A Ettinger
- Robert H. Williams Lab, Department of Immunology, University of Washington, Box 357710, 1959 NE Pacific St, HSB K-165, Seattle, WA 98195-7710, USA.
| | | | | |
Collapse
|
33
|
Li QJ, Dinner AR, Qi S, Irvine DJ, Huppa JB, Davis MM, Chakraborty AK. CD4 enhances T cell sensitivity to antigen by coordinating Lck accumulation at the immunological synapse. Nat Immunol 2004; 5:791-9. [PMID: 15247914 DOI: 10.1038/ni1095] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Accepted: 06/18/2004] [Indexed: 11/09/2022]
Abstract
How T cells respond with extraordinary sensitivity to minute amounts of agonist peptide and major histocompatibility complex (pMHC) molecules on the surface of antigen-presenting cells bearing large numbers of endogenous pMHC molecules is not understood. Here we present evidence that CD4 affects the responsiveness of T helper cells by controlling spatial localization of the tyrosine kinase Lck in the synapse. This finding, as well as further in silico and in vitro experiments, led us to develop a molecular model in which endogenous and agonist pMHC molecules act cooperatively to amplify T cell receptor signaling. At the same time, activation due to endogenous pMHC molecules alone is inhibited. A key feature is that the binding of agonist pMHC molecules to the T cell receptor results in CD4-mediated spatial localization of Lck, which in turn enables endogenous pMHC molecules to trigger many T cell receptors. We also discuss broader implications for T cell biology, including thymic selection, diversity of the repertoire of self pMHC molecules and serial triggering.
Collapse
Affiliation(s)
- Qi-Jing Li
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Grandjean I, Duban L, Bonney EA, Corcuff E, Di Santo JP, Matzinger P, Lantz O. Are major histocompatibility complex molecules involved in the survival of naive CD4+ T cells? ACTA ACUST UNITED AC 2003; 198:1089-102. [PMID: 14517277 PMCID: PMC2194222 DOI: 10.1084/jem.20030963] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The exact role of major histocompatibility complex (MHC) molecules in the peripheral survival of naive T cells is controversial, as some studies have suggested that they are critically required whereas others have suggested that they are not. Here we controlled for some of the features that differed among the earlier studies, and analyzed both the survival and expansion of naive CD4+ T cells transferred into MHC syngeneic, allogeneic, or MHC negative environments. We found that naive T cells transferred into MHC negative or allogeneic environments often fail to survive because of rejection and/or competition by natural killer (NK) cells, rather than failure to recognize a particular MHC allele. In the absence of NK cells, naive CD4+ T cells survived equally well regardless of the MHC type of the host. There was, however, an MHC requirement for extensive space-induced “homeostatic” expansion. Although the first few divisions occurred in the absence of MHC molecules, the cells did not continue to divide or transit to a CD44hi phenotype. Surprisingly, this MHC requirement could be satisfied by alleles other than the restricting haplotype. Therefore, space-induced expansion and survival are two different phenomena displaying different MHC requirements. Memory CD4+ T cells, whose survival and expansion showed no requirements for MHC molecules at all, dampened the space-induced expansion of naive cells, showing that the two populations are not independent in their requirements for peripheral niches.
Collapse
Affiliation(s)
- Isabelle Grandjean
- Laboratoire d'Immunologie, Institut Curie, 26 rue d'Ulm, 75005 Paris, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Denning TL, Qi H, König R, Scott KG, Naganuma M, Ernst PB. CD4+ Th cells resembling regulatory T cells that inhibit chronic colitis differentiate in the absence of interactions between CD4 and class II MHC. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2279-86. [PMID: 12928372 DOI: 10.4049/jimmunol.171.5.2279] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Regulatory CD4+ Th cells can prevent many autoimmune diseases; however, the factors selecting for these cells remain poorly defined. In transgenic mice with a mutation in the CD4 binding region on class II MHC, the disruption of CD4-class II interactions selected for CD4+ Th cells that expressed surface markers and cytokines associated with regulatory Th cells. Th cells from these mice were enriched for CD45RB(low) as well as CD25+, while they expressed high levels of the transcription factor associated with regulatory T cells, Foxp3, and cytokines, including IL-4, IL-10, and IFN-gamma mRNA and protein. These regulatory Th cells inhibited the function of APCs via IL-10 production, and adoptive transfer of these cells prevented weight loss and inflammation in a model of colitis. CD4+ regulatory Th cells emerged only when interactions between CD4 and class II MHC were deficient on cells of nonhemopoietic origin. These data support a novel model controlling the differentiation of regulatory Th cells and suggest that interactions between CD4 and class II MHC may a useful target for re-educating T cells as a treatment for inflammatory diseases.
Collapse
Affiliation(s)
- Timothy L Denning
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | | | |
Collapse
|
36
|
Zhou W, König R. T cell receptor-independent CD4 signalling: CD4-MHC class II interactions regulate intracellular calcium and cyclic AMP. Cell Signal 2003; 15:751-62. [PMID: 12781868 DOI: 10.1016/s0898-6568(03)00037-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
CD4 is a coreceptor on T helper (Th) cells that interacts with MHC class II molecules (MHCII). The mechanisms mediating the effects of CD4 on responses by T helper cells to stimulation of the antigen-specific T cell receptor (TCR) are still poorly understood. Here, we demonstrate T cell costimulation via CD4 signalling independent of T cell receptor-mediated signals. Incubation of T helper cells with peptide mimetics of the CD4-binding region on the MHC class II beta2 domain caused intracellular calcium mobilization in the absence of antigen or other T cell receptor stimuli. Engagement of CD4 by peptide mimetics or wild-type MHC class II, but not by mutant MHC class II molecules incapable of engaging CD4, inhibited the T cell receptor-mediated increase in cyclic AMP (cAMP) concentrations in T helper cells. CD4-mediated signals activated cyclic AMP phosphodiesterases (PDEs) and inhibited adenylyl cyclase. Full activation and clonal expansion of antigen-stimulated T helper cells required the CD4-mediated regulation of cyclic AMP. Our results suggest a costimulatory mechanism of CD4 function that acts on the second messengers, calcium and cyclic AMP.
Collapse
Affiliation(s)
- Wenhong Zhou
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | |
Collapse
|
37
|
Moldovan MC, Yachou A, Lévesque K, Wu H, Hendrickson WA, Cohen EA, Sékaly RP. CD4 dimers constitute the functional component required for T cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6261-8. [PMID: 12444132 DOI: 10.4049/jimmunol.169.11.6261] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CD4 molecule plays a key role in the development and activation of helper T cells. Dimerization and oligomerization is often a necessary step in the function of several cell surface receptors. Herein, we provide direct biochemical evidence confirming the presence of CD4 as dimers in transfected cells from hemopoetic and fibroblastic origin as well as in primary T cells. Such dimers are also observed with murine CD4 confirming selective pressure during evolution to maintain such a structure. Using a series of point mutations, we have precisely mapped the dimerization site at residues K318 and Q344 within the fourth extracellular domain of CD4. These residues are highly conserved and their mutation results in interference with dimer formation. More importantly, we demonstrate that dimer formation is essential for the coligand and coreceptor functions of CD4 in T cell activation. These data strongly suggest that CD4 dimerization is necessary for helper T cell function.
Collapse
|
38
|
König R, Shen X, Maroto R, Denning TL. The role of CD4 in regulating homeostasis of T helper cells. Immunol Res 2002; 25:115-30. [PMID: 11999166 DOI: 10.1385/ir:25:2:115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Intrathymic T cell selection and peripheral activation of mature T cells are crucial for self-recognition and the general immune response to viral, bacterial, and tumor antigens. The T cell coreceptors, CD4 and CD8, contribute to the regulation of these processes. The importance of interactions between CD4 and molecules encoded by the class II major histocompatibility complex (MHC) for thymic T cell selection has been clearly established, however, the role of CD4-MHC class II interactions in T helper (TH) cell differentiation, in the maintenance of homeostasis in the peripheral immune system, and in the generation of memory TH cells is largely unclear. Here, we present evidence for a role of CD4 in controlling homeostasis in the peripheral immune system. We also demonstrate the importance of CD4-MHC class II interactions in inducing these previously not recognized functions of CD4.
Collapse
Affiliation(s)
- Rolf König
- Department of Microbiology and Immunology and the Sealy Center for Molecular Science, The University of Texas Medical Branch, Galveston 77555-1070, USA.
| | | | | | | |
Collapse
|
39
|
Horie T, Shen Y, Kajino K, Gaubin M, Bonomi G, Mani JC, Berezov A, Piatier-Tonneau D, Guardiola J, Hillard B, Rostami A, Greene M, Murali R. Study of disabling T-cell activation and inhibiting T-cell-mediated immunopathology reveals a possible inverse agonist activity of CD4 peptidomimetics. Exp Mol Pathol 2002; 73:93-103. [PMID: 12231211 DOI: 10.1006/exmp.2002.2444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We designed a new class of aromatically modified exocyclic peptides based on the structure of CD4 by engineering one of the cysteine residues in a peptidomimetic derived from the CDR3 region of the CD4 molecule. All three species mediate inhibition of T-cell proliferation at concentrations ranging from 10 to 100 microM. The mimetics CD4-Cys and CD4-Met bind to sCD4 with affinities ranging from 1 to 2 microM, while CD4-Ser shows poor binding in radioisotope assay. Though these mimetics have similar structures, they exhibit different biochemical and biological functions. Activation of T-cells as measured by thymidine incorporation or IL-2 production revealed that CD4-Cys and CD4-Ser mimetics behave as classical antagonists. On the other hand, the CD4-Met species inhibited T-cell proliferation with an IC(50) of 30 microM but unexpectedly increased IL-2 secretion modestly at a less than 3 microM concentration. In experimental autoimmune encephalitis (EAE), CD4-Ser and CD4-Cys mimetics reduced the severity of EAE symptoms while the CD4-Met mimetic exacerbated the conditions. We propose that CD4-Cys and CD4-Ser are classical antagonists, but CD4-Met may possess properties of an inverse agonist. The structure-activity relationship of mimetics reveals that a minor change in the net hydropathic value is enough to alter the dynamic nature of the receptor-ligand complex.
Collapse
MESH Headings
- Animals
- Biotechnology
- CD4 Antigens/chemistry
- CD4 Antigens/metabolism
- CD4 Antigens/pharmacology
- Complementarity Determining Regions/chemistry
- Complementarity Determining Regions/metabolism
- Computer Simulation
- Dimerization
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Guinea Pigs
- Humans
- In Vitro Techniques
- Interleukin-2/biosynthesis
- Interleukin-2/immunology
- Lymphocyte Activation/drug effects
- Major Histocompatibility Complex
- Mice
- Mice, Inbred C3H
- Mice, Inbred Strains
- Models, Biological
- Models, Molecular
- Peptide Fragments/chemistry
- Peptide Fragments/metabolism
- Peptide Fragments/pharmacology
- Protein Conformation
- Receptors, Antigen, T-Cell/metabolism
- Structure-Activity Relationship
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Takeo Horie
- Department of Pathology and School of Medicine, University of Pensylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rudolph MG, Luz JG, Wilson IA. Structural and thermodynamic correlates of T cell signaling. ANNUAL REVIEW OF BIOPHYSICS AND BIOMOLECULAR STRUCTURE 2002; 31:121-49. [PMID: 11988465 DOI: 10.1146/annurev.biophys.31.082901.134423] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The first crystal structures of intact T cell receptors (TCRs) bound to class I peptide-MHC (pMHCs) antigens were determined in 1996. Since then, further structures of class I TCR/pMHC complexes have explored the degree of structural variability in the TCR-pMHC system and the structural basis for positive and negative selection. The recent determination of class II and allogeneic class I TCR/pMHC structures, as well as those of accessory molecules (e.g., CD3), has pushed our knowledge of TCR/pMHC interactions into new realms, shedding light on clinical pathologies, such as graft rejection and graft-versus-host disease. Furthermore, the determination of coreceptor structures lays the foundation for a more comprehensive structural description of the supramolecular TCR signaling events and those assemblies that arise in the immunological synapse. While these telling photodocumentaries of the TCR/pMHC interaction are composed mainly from static crystal structures, a full description of the biological snapshots in T cell signaling requires additional analytical methods that record the dynamics of the process. To this end, surface plasmon resonance (SPR), isothermal titration calorimetry (ITC), and ultracentrifugation (UC) have furnished both affinities and kinetics of the TCR/pMHC association. In the past year, structural, biochemical, and molecular biological data describing TCR/pMHC interactions have sublimely coalesced into a burgeoning well of understanding that promises to deliver further insights into T cell recognition. The coming years will, through a more intimate union of structural and kinetic data, allow many pressing questions to be addressed, such as how TCR/pMHC ligation is affected by coreceptor binding and what is the mechanism of TCR signaling in both early and late stages of T cell engagement with antigen-presenting cells.
Collapse
Affiliation(s)
- Markus G Rudolph
- Department of Molecular Biology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
41
|
Marschner S, Hünig T, Cambier JC, Finkel TH. Ligation of human CD4 interferes with antigen-induced activation of primary T cells. Immunol Lett 2002; 82:131-9. [PMID: 12008044 DOI: 10.1016/s0165-2478(02)00028-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The CD4 molecule functions to enhance T cell activation when it is co-aggregated with the T cell receptor for antigen (TCR) by MHC class II antigenic peptide complexes. However, independent ligation of CD4 has been shown to negatively effect signaling through the TCR in vitro. The interaction between the HIV-1 envelope glycoprotein gp120 and CD4 is a central event in the pathogenesis of AIDS and may contribute to immune deficiency via both direct and indirect mechanisms, including lytic infection of T cells and induction of CD4 signaling events resulting in apoptosis and anergy. Analysis of the consequences of interactions between CD4 and gp120 have yielded contradictory results presumably because most of these studies have focused on T cell clones of questionable relevance to the in vivo target of the virus. Here, we analyzed the effects of CD4 ligation on freshly isolated cells of human CD4 transgenic mice, and show that huCD4 preligation, in the absence of human CXCR4, has an inhibitory effect on both early and late T cell activation events. CD4 signaling negatively regulates the response to antigen, as well as to anti-TCR mAb. In addition, we show here that this negative signal requires the cytoplasmic tail of CD4. These results suggest that in HIV infected patients the interaction of gp120 with CD4 induces unresponsiveness of CD4+ T cells to subsequent activation by antigen.
Collapse
Affiliation(s)
- Susanne Marschner
- Integrated Department of Immunology, National Jewish Medical and Research Center, University of Colorado Health Sciences Center, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
42
|
Moustakas AK, Papadopoulos GK. Molecular properties of HLA-DQ alleles conferring susceptibility to or protection from insulin-dependent diabetes mellitus: keys to the fate of islet beta-cells. AMERICAN JOURNAL OF MEDICAL GENETICS 2002; 115:37-47. [PMID: 12116175 DOI: 10.1002/ajmg.10342] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The major histocompatibility complex Class II alleles, HLA-DQ, and the related HLA-DR, are the chief genetic elements of human type 1 diabetes. These genes code for polymorphic heterodimeric proteins, whose chief function is to trap peptide antigens in the endosome and present them on the surface of antigen-presenting cells (dendritic cells, B lymphocytes, monocytes/macrophages) to CD4(+) T helper cells. A systematic investigation of the molecular properties of HLA-DQ alleles linked to susceptibility or resistance to type 1 diabetes has shown that these properties segregate along lines of susceptibility or resistance. A correlation of these features with the function of each particular segment of the HLA-DQ molecule yields interesting insights into the possible pathways leading to type 1 diabetes. There remain, however, areas to be clarified, including mechanisms by which dominant protection is conferred by certain alleles, the interplay between HLA-DQ and the related locus HLA-DR, that also shows autoantigen-specific reactivity, and the cross-Class help delivered to CD8(+) T cells, the final effectors in pancreatic beta-cell destruction. Clarification of these issues may lead to ways to prevent diabetes in predisposed individuals already exhibiting the genetic and immunological characteristics, and perhaps a cure in those with the disease, by means of transplantation, and measures for prevention of disease recurrence.
Collapse
|
43
|
Edling AE, Choksi S, Huang Z, Korngold R. An organic CD4 inhibitor reduces the clinical and pathological symptoms of acute experimental allergic encephalomyelitis. J Autoimmun 2002; 18:169-79. [PMID: 11908949 DOI: 10.1006/jaut.2001.0576] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CD4(+) T cells have an important role in mediating the pathogenesis of many human and experimental autoimmune diseases including experimental allergic encephalomyelitis (EAE), a demyelinating animal model for multiple sclerosis (MS). We applied a computer screening approach to select a small organic molecule, TJU103, that would specifically inhibit autoreactive CD4(+) T cells by disrupting the function of the CD4 molecule during activation. Upon studying the therapeutic effect of TJU103 in acute EAE, it was found that administration shortly before or after the onset of clinical symptoms reduced the severity of disease in both SJL and SWXJ-14 mouse models. In addition, TJU103 treatment could affect both in vivo responses to EAE rechallenge and secondary in vitro proliferation and cytokine production of T cells responding to proteolipid protein epitope 139-151 (PLPe). These results demonstrate the potential of the TJU103 organic inhibitor for future clinical application in CD4(+) T cell-mediated diseases.
Collapse
Affiliation(s)
- Andrea E Edling
- Kimmel Cancer Institute, Jefferson Medical College, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Genetic experiments indicate similarity between binding sites on MHC class I (MHCI) for CD8 and on MHCII for CD4, but the crystal structures of CD8/MHCI and CD4/MHCII complexes suggest critical differences between the interfaces in the two complexes. Biophysical analyses using ectodomains of co-receptors and MHC molecules demonstrate extremely fast kinetics and low-affinity interactions. Experiments with soluble multimeric MHC ligands suggest that CD4 and CD8 may differ in the mechanisms by which they promote the formation of ternary TCR/MHC/co-receptor complexes. Co-receptor-influenced duration of TCR signaling controls thymocyte selection. In naïve T cells, CD4/MHCII interactions may promote T-cell survival. Temporal and spatial analysis of TCR and CD4 co-clustering in the immunological synapse suggests that CD4 recruitment is regulated by the half-life of the initial TCR/MHCII complex. Diverse experimental systems have yielded conflicting data that have helped to formulate revised mechanistic models of co-receptor function.
Collapse
Affiliation(s)
- Rolf König
- Department of Microbiology and Immunology and the Sealy Center for Molecular Science, The University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| |
Collapse
|
45
|
Wang Q, Malherbe L, Zhang D, Zingler K, Glaichenhaus N, Killeen N. CD4 promotes breadth in the TCR repertoire. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4311-20. [PMID: 11591754 DOI: 10.4049/jimmunol.167.8.4311] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A diverse population of MHC class II-restricted CD4 lineage T cells develops in mice that lack expression of the CD4 molecule. In this study, we show that the TCR repertoire selected in the absence of CD4 is distinct, but still overlapping in its properties with that selected in the presence of CD4. Immunization of mice lacking CD4 caused the clonal expansion of T cells that showed less breadth in the range of Ag-binding properties exhibited by their TCRs. Specifically, the CD4-deficient Ag-specific TCR repertoire was depleted of TCRs that demonstrated low-affinity binding to their ligands. The data thus suggest a key role for CD4 in broadening the TCR repertoire by potentiating productive TCR signaling and clonal expansion in response to the engagement of low-affinity antigenic ligands.
Collapse
Affiliation(s)
- Q Wang
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
46
|
Wang JH, Meijers R, Xiong Y, Liu JH, Sakihama T, Zhang R, Joachimiak A, Reinherz EL. Crystal structure of the human CD4 N-terminal two-domain fragment complexed to a class II MHC molecule. Proc Natl Acad Sci U S A 2001; 98:10799-804. [PMID: 11535811 PMCID: PMC59561 DOI: 10.1073/pnas.191124098] [Citation(s) in RCA: 186] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The structural basis of the interaction between the CD4 coreceptor and a class II major histocompatibility complex (MHC) is described. The crystal structure of a complex containing the human CD4 N-terminal two-domain fragment and the murine I-A(k) class II MHC molecule with associated peptide (pMHCII) shows that only the "top corner" of the CD4 molecule directly contacts pMHCII. The CD4 Phe-43 side chain extends into a hydrophobic concavity formed by MHC residues from both alpha 2 and beta 2 domains. A ternary model of the CD4-pMHCII-T-cell receptor (TCR) reveals that the complex appears V-shaped with the membrane-proximal pMHCII at the apex. This configuration excludes a direct TCR-CD4 interaction and suggests how TCR and CD4 signaling is coordinated around the antigenic pMHCII complex. Human CD4 binds to HIV gp120 in a manner strikingly similar to the way in which CD4 interacts with pMHCII. Additional contacts between gp120 and CD4 give the CD4-gp120 complex a greater affinity. Thus, ligation of the viral envelope glycoprotein to CD4 occludes the pMHCII-binding site on CD4, contributing to immunodeficiency.
Collapse
Affiliation(s)
- J H Wang
- Laboratory of Immunobiology and Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gáspár R, Bagossi P, Bene L, Matkó J, Szöllosi J, Tozsér J, Fésüs L, Waldmann TA, Damjanovich S. Clustering of class I HLA oligomers with CD8 and TCR: three-dimensional models based on fluorescence resonance energy transfer and crystallographic data. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5078-86. [PMID: 11290789 DOI: 10.4049/jimmunol.166.8.5078] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fluorescence resonance energy transfer (FRET) data, in accordance with lateral mobility measurements, suggested the existence of class I HLA dimers and oligomers at the surface of live human cells, including the B lymphoblast cell line (JY) used in the present study. Intra- and intermolecular class I HLA epitope distances were measured on JY B cells by FRET using fluorophore-conjugated Ag-binding fragments of mAbs W6/32 and L368 directed against structurally well-characterized heavy and light chain epitopes, respectively. Out-of-plane location of these epitopes relative to the membrane-bound BODIPY-PC (2-(4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza-s-indacene-3-pentanoyl)-1-hexadecanoyl-sn-glycero-3-phosphocholine) was also determined by FRET. Computer-simulated docking of crystallographic structures of class I HLA and epitope-specific Ag-binding fragments, with experimentally determined interepitope and epitope to cell surface distances as constraints, revealed several sterically allowed and FRET-compatible class I HLA dimeric and tetrameric arrangements. Extension of the tetrameric class I HLA model with interacting TCR and CD8 resulted in a model of a supramolecular cluster that may exist physiologically and serve as a functionally significant unit for a network of CD8-HLA-I complexes providing enhanced signaling efficiency even at low MHC-peptide concentrations at the interface of effector and APCs.
Collapse
Affiliation(s)
- R Gáspár
- Department of Biophysics, Biophysics Research Group of the Hungarian Academy of Sciences, University of Debrecen, Medical and Health Science Center, Debrecen, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lindstedt R, Monk N, Lombardi G, Lechler R. Amino acid substitutions in the putative MHC class II "dimer of dimers" interface inhibit CD4+ T cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:800-8. [PMID: 11145653 DOI: 10.4049/jimmunol.166.2.800] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of T lymphocytes is dependent on multiple ligand-receptor interactions. The possibility that TCR dimerization contributes to T cell triggering was raised by the crystallographic analysis of MHC class II molecules. The MHC class II molecules associated as double dimers, and in such a way that two TCR (and two CD4 molecules) could bind simultaneously. Several subsequent studies have lent support to this concept, although the role of TCR cross-linking in T cell activation remains unclear. Using DRA cDNAs modified to encode two different C-terminal tags, no evidence of constitutive double dimer formation was obtained following immunoprecipitation and Western blotting from cells transiently transfected with wild-type DRB and tagged DRA constructs, together with invariant chain and HLA-DM. To determine whether MHC class II molecules contribute actively to TCR-dependent dimerization and consequent T cell activation, panels of HLA-DR1beta and H2-E(k) cDNAs were generated with mutations in the sequences encoding the interface regions of the MHC class II double dimer. Stable DAP.3 transfectants expressing these cDNAs were generated and characterized biochemically and functionally. Substitutions in either interface region I or III did not affect T cell activation, whereas combinations of amino acid substitutions in both regions led to substantial inhibition of proliferation or IL-2 secretion by human and murine T cells. Because the amino acid-substituted molecules were serologically indistinguishable from wild type, bound antigenic peptide with equal efficiency, and induced Ag-dependent CD25 expression indicating TCR recognition, the reduced ability of the mutants to induce full T cell activation is most likely the result of impaired double dimer formation. These data suggest that MHC class II molecules, due to their structural properties, actively contribute to TCR cross-linking.
Collapse
Affiliation(s)
- R Lindstedt
- Department of Immunology, Division of Medicine, Imperial College of Science, Technology, and Medicine, Hammersmith Hospital, Du Cane Road, London, United Kingdom
| | | | | | | |
Collapse
|
49
|
Edling AE, Choksi S, Huang Z, Korngold R. Effect of a cyclic heptapeptide based on the human CD4 domain 1 CC' loop region on murine experimental allergic encephalomyelitis: inhibition of both primary and secondary responses. J Neuroimmunol 2001; 112:115-28. [PMID: 11108940 DOI: 10.1016/s0165-5728(00)00393-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The 802-2 peptide, designed from the conserved D1-CC' loop region of human CD4, can disrupt CD4(+) T cell activation in both human and murine systems. Here, 802-2 was investigated for efficacy in acute murine experimental allergic encephalomyelitis (EAE) models, and was found to significantly reduce the severity of disease when administered either before or after the onset of symptoms. 802-2 treatment during PLP139-151 induction of EAE rendered the mice more resistant to subsequent rechallenge with antigen, and was also efficacious when initially administered during a secondary EAE response. T cells from 802-2-treated mice proliferated poorly to in vitro restimulation with PLP139-151 and exhibited decreased frequencies of IL-2, IL-4, and IFN-gamma producing cells, but were still able to respond to third-party antigens. These combined results suggest the potential therapeutic value of 802-2 for inhibition of CD4(+) T cell neuroimmunological responses.
Collapse
Affiliation(s)
- A E Edling
- Department of Microbiology and Immunology, Jefferson Medical College, 233 S. 10th Street, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
50
|
Poussin MA, Goluszko E, Hughes TK, Duchicella SI, Christadoss P. Suppression of experimental autoimmune myasthenia gravis in IL-10 gene-disrupted mice is associated with reduced B cells and serum cytotoxicity on mouse cell line expressing AChR. J Neuroimmunol 2000; 111:152-60. [PMID: 11063833 DOI: 10.1016/s0165-5728(00)00385-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To analyze the role of interleukin-10 (IL-10) in experimental autoimmune myasthenia gravis (EAMG) pathogenesis, we induced clinical EAMG in C57BL/6 and IL-10 gene-knockout (KO) mice. IL-10 KO mice had a lower incidence and severity of EAMG, with less muscle acetylcholine receptor (AChR) loss. AChR-immunized IL-10 KO mice showed a significantly higher AChR-specific proliferative response, altered cytokine response, lower number of class II-positive cells and B-cells, but a greater CD5(+)CD19(+) population than C57BL/6 mice. The lower clinical incidence in IL-10 KO could be explained not by a reduction of the quantity, but by a possible difference in the pathogenicity of anti-AChR antibodies.
Collapse
MESH Headings
- Adjuvants, Immunologic/genetics
- Animals
- Antigens, CD19/analysis
- Autoantibodies/blood
- B-Lymphocytes/chemistry
- B-Lymphocytes/cytology
- B-Lymphocytes/immunology
- Blood Proteins/immunology
- CD5 Antigens/analysis
- Cell Division/immunology
- Cell Line
- Cytotoxins/immunology
- Epitopes/immunology
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/immunology
- Immunization
- Immunodominant Epitopes/immunology
- In Vitro Techniques
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-6/biosynthesis
- Interleukin-6/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/chemistry
- Muscle, Skeletal/immunology
- Myasthenia Gravis, Autoimmune, Experimental/genetics
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Receptors, Cholinergic/genetics
- Receptors, Cholinergic/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- M A Poussin
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555-1070, Galveston, TX, USA
| | | | | | | | | |
Collapse
|